)..n . : .: ‘1 vulva“ I!!! m rvg-cnn In! ‘ "away-I wmw 1 ; urr-v -n V 0:“ :flug-gm;'~pn= u «a... "an .1 qua-I kync¢y I 39! .l‘? ulnu {go-z.- "4.‘(3Ql"""' a . 1. 1.93 .n :1 tin... .5? \F .1 . ‘ . This is to certify that the dissertation entitled THE PRESENCE OF A LOCAL SEROTONERGIC SYSTEM IN PERIPHERAL ARTERIES presented by Wei Ni has been accepted towards fulfillment of the requirements for the Ph.D. degree in Pharmacology and Toxicology w Waté; Major Professor’s Signature' IZi/Ia/AQOOE Date MSU is an Affirmative Action/Equal Opportunity Institution “LIBRARY Michigan State University PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 p:/ClRC/DateDue.indd-p.1 THE PRESENCE OF A LOCAL SEROTONERGIC SYSTEM IN PERIPHERAL ARTERIES Wei Ni A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pharmacology and Toxicology 2006 ABSTRACT THE PRESENCE OF A LOCAL SEROTONERGIC SYSTEM IN PERIPHERAL ARTERIES By Wei Ni Serotonin (5-hydroxytryptamine, 5—HT), originally discovered in the intestinal tract and blood, has multiple functions in physiological and pathological conditions. Increases in reactivity to 5-HT have been observed in a number of different forms of hypertension models including DOCA-salt hypertensive rats, spontaneously hypertensive rats and in human patients. However, whether there is a local serotonergic system in peripheral arteries has not been studied. We hypothesized that the existence of a local serotonergic system (including 5-HT synthesis or metabolism, reuptake or release) in peripheral arteries and that this local regulation of 5-HT is altered in hypertension. We discovered: (1) the presence of essential enzymes for 5-HT synthesis and metabolism in peripheral arteries; (2) peripheral arteries are capable of taking up and releasing 5-HT; (3) the presence of a functional serotonin transporter (SERT), which is the major protein responsible for 5-HT uptake; (4) SERT expression is increased but arterial 5-HT uptake function is decreased in mineralocorticoid hypertension (DOCA—salt hypertensive rats) and nitric oxide synthase inhibited hypertension (LNNA hypertensive rats), but not in spontaneously hypertensive rats compared to their non'notensive control. Our studies showed that a serotonergic system exists in peripheral arteries and may play a role in local control of total peripheral resistance. DEDICATION To my parents, Hengjin Ni and Suli Cai, who gave me life and encouraged me to go after my dream. ACKNOWLEDGEMENT First of all, | wish to acknowledge my mentor Dr. Stephanie W. Watts, who taught me to think, to speak, to write and to do research as a scientist. Thank you. I would like to thank my committee members: Dr. Greg Fink, Dr. JR Haywood, Dr. Marc Bailie and Dr. Keith Lookingland for their guidance, their challenges, and their suggestions. I am appreciated to be in Dr. Watts’s lab. I had great time to be with people worked there. To my friend and classmate Dr. Keshari Thakali, thanks her for being my good friend and the person I can always count on. To Janice Thompson, Theo Irina Szasz, Jessica Diaz and Robert Burnett for their friendship and support. I have learned so many things, science and life, from all of you. At last, I would also like to thank my husband, Yue Huang, for his love and unconditional supports! TABLE OF CONTENT List of Tables .............................................................................. x List of Figures ............................................................................. xi List of Abbreviations ................................................................... xv Introduction ................................................................................... 1 l. Biosynthesis, storage, and metabolism of 5-HT .......................... 2 1. Biosynthesis ....................................................................... 2 A. Biochemistry .............................................................. 2 B. Location .................................................................... 2 C. Regulation ................................................................. 4 D. Pharmacology ............................................................ 6 2. Storage .............................................................................. 6 A. Location .................................................................... 6 B. Mechanisms ............................................................... 7 C. Regulation and Pharmacology ...................................... 8 3. Metabolism ......................................................................... 9 A. Biochemistry ............................................................. 9 B. Location ................................................................... 10 C. Regulation and Pharmacology ...................................... 10 II. Uptake and release of 5- HT ................................................... 11 1. The major component responsible for 5- HT uptake and release--- SERT ..................................................................................... 1 1 A. Molecular Biochemistry ................................................ 11 8. Location .................................................................... 13 C. Function and Physiology .............................................. 13 D. Pharmacology ............................................................ 15 E. Regulation ................................................................. 17 2. Other components responsible for 5-HT uptake and release--- Non-SERT dependent ............................................................... 20 Ill. 5-HT Function Mechanisms---- in General ................................. 21 1. Extracellular 5-HT functions (5-HT receptors) ............................ 21 2. Intracellular 5-HT functions .................................................... 21 IV. 5-HT and SERT in Physiological Cardiovascular System .............. 22 1. 5-HT in Physiological Cardiovascular System ............................ 22 A. Blood ........................................................................ 22 B Brain / Sympathetic Nervous System ............................... 23 C Heart ........................................................................ 23 D. Kidney ...................................................................... 24 E. Blood Vessels ............................................................ 24 F. Whole Body ............................................................... 26 2. SERT in the Physiological Cardiovascular System ...................... 26 A Pulmonary Vasculature ................................................ 26 B Platelet Function ......................................................... 27 vi C. Cardiac Function ........................................................ 28 D. Other Cardiovascular Organs ........................................ 28 V. Serotonergic System in Cardiovascular Diseases ....................... 29 1. Serotonergic System and MI ................................................... 3O 2. Serotonergic System and PPH ............................................... 30 3. Serotonergic System and Hypertension .................................... 31 A. Free Circulating 5-HT Concentrations and 5-HT Receptors Activation ................................................................................. 31 8. Uses of 5-HT Receptor Antagonists in the Treatment of Hypertension ........................................................................... 33 C. High L-tryptophan Diet and Blood Pressure ....................... 35 D. TPH Function Inhibition and Blood Pressure ..................... 36 E. Other Evidence Supporting the Involvement of Serotonergic System in Hypertension ............................................................ 37 F. An Unanswered Question --- The Existence of Local Regulation of 5-HT in Peripheral Arteries ...................................... 38 Hypotheses ................................................................................ 39 Methods ..................................................................................... 42 l. Animal Uses ........................................................................ 42 II. Euthanasia .......................................................................... 42 III. Animal Models ..................................................................... 42 IV. BP Measurements ................................................................ 44 V. 5-HT Basal Level Measurement .............................................. 44 VI. TPH Activity Assay ............................................................... 45 VII. 5-HT Storage Assay .............................................................. 46 VIII. 5-HT Uptake Assay ............................................................... 46 IX. 5-HT Release Assay ............................................................. 46 X. 5-HIAA and 5-HT Measurement .............................................. 47 XI. Real Time RT-PCR ............................................................... 48 XII. lmmunohistochemistry ........................................................... 49 XIII. lmmunocytochemistry ........................................................... 50 XIV. Protein Isolation ................................................................... 51 XV. BCA Protein Assay ............................................................... 51 XVI. Western Blotting ................................................................... 52 XVII. Isolated Smooth Muscle Contractility Measurement ..................... 52 XVIII. Data Analysis and Statistics ................................................... 53 Results ....................................................................................... 55 Hypothesis #1 ............................................................................... 55 I.The Presence of 5-HT in Peripheral Arteries .................................... 55 II. The Presence of 5-HT Synthesis and Metabolism in Peripheral Arteries ........................................................................................ 64 vii 1. The Existence of TPH1 mRNA and Protein in Peripheral Arteries... 64 2. The Activity of TPH in Peripheral Arteries .................................. 70 3. The Presence of AADC in Rat Aorta and Superior Mesenteric Artery ...................................................................................... 73 4. The Presence of MAO A in Rat Aorta and Superior Mesenteric Artery ...................................................................................... 73 5. The Activity of MAO A in Rat and Superior Mesenteric Artery ........ 81 III. The Lack of 5-HT Storage in Peripheral Artery ........................... 84 1. Investigation of Whether Rat Aorta Has Ability to Store 5-HT ........ 84 2. Investigation of “Serotonylated” 5-HT in Rat Aorta ....................... 89 IV. The Presence of Uptake and Release of 5-HT in Peripheral Arteries ......................................................................................... 95 1. The Presence of SERT in Peripheral Arteries ............................. 95 2. The Presence of Active 5-HT Uptake in Peripheral Arteries ........... 100 A. 5-HT Uptake Time Course Study ..................................... 100 B. Investigation of SERT-dependent 5-HT Uptake .................. 106 C. Investigation of SERT-independent 5-HT Uptake ............... 110 3. The Presence of 5-HT Release in Peripheral Arteries ................... 125 Hypothesis #2 .................................................................................. 132 I. Measurement of SERT Expression in Aorta from Normotensive and Hypertensive rats ........................................................................ 132 II. Comparison of the Basal Level of 5-HT in DOCA-salt Rats, LNNA Rats and SHR with Their Normotensive Control Rats ............................ 135 III. 5-HT Uptake Comparison in DOCA—salt Rats, LNNA Rats and SHR with Their Normotensive Control Rats ........................................... 138 IV. SERT Function in Contractility- Comparison of DOCA-salt Rats 145 with Normal Rats ............................................................................. Discussion ..................................................................................... 150 I.Biochemical Proof of the Presence of 5-HT and its Metabolite in Peripheral Normal Arteries ................................................................. 150 II. Investigation of 5-HT Synthesis in Peripheral Normal Arteries ....... 151 III. Investigation of Metabolism of 5-HT in Peripheral Arteries ............ 153 IV. Investigation of Storage of 5-HT in Peripheral Arteries ................. 154 V. Investigation of Functional SERT in Peripheral Normal Arteries ..... 155 VI. Investigation of SERT-independent 5-HT Uptake Mechanisms in Peripheral Arteries ............................................................................ 156 VII. Investigation of 5-HT Release Mechanisms in Peripheral Arteries. 158 VIII. Physiological Relevance of 5-HT Uptake in Peripheral Arteries and Speculation ..................................................................................... 159 IX. Pathological Relevance of 5-HT Uptake in Peripheral Arteries and Speculation ...................................................................................... 160 X. Basal 5-HT Concentrations in Aorta of DOCA-salt, LNNA Hypertensive Rats and SHR .............................................................. 162 viii XI. 5-HT Uptake Ability is Reduced in Aorta of DOCA—salt and LNNA Hypertensive Rats, but not in SHR ..................................................... 164 XII. Other Arterial 5-HT-uptake Mechanisms in Hypertensive Animals. 167 XIII. The Effect of SERT Function on Arterial Contractility in Hypertension .................................................................................... 166 XIV. Future Research ..................................................................... 169 XV. Limitations ............................................................................. 171 Conclusion ..................................................................................... 172 Reference ....................................................................................... 1 74 Bibliography .................................................................................. 193 Table 1. Table 2. Table 3. Table 4. Table 5. Table 6. LIST OF TABLES The CT values in real time RT-PCR experiments detecting the expression of tph1 and B-Z-microglobulin mRNA in rat aorta and superior mesenteric artery ................................ 67 Quantification of 5—HIAA and 5-HT in A 5-HT storage study in aorta from normal rats ............................................... 88 Quantification of 5-HIAA and 5-HT in A 5-HT storage study in aorta from pargyline-treated rats .................................... 91 Quantification of 5-HIAA and 5-HT in A 5-HT storage study in 5-HT—preloaded aorta from pargyline—treated rats ........... 93 Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in aorta from wild type and tph1-/- mice ................................ 109 Comparison of the effect of fluoxetine and fluvoxamine on EC50 and threshold of 5—HT-induced contraction in aorta from SHAM and DOCA rats ............................................. 149 Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. Figure 12. Figure 13. Figure 14. LIST OF FIGURES 5-HT Biosynthesis and Metabolism Pathway ...................... 3 Top: Proposed Topology of Plasma-membrane SERT Bottom: Mechanism of 5-HT Transporter .......................... 12 Diagram of Working Hypotheses ...................................... 41 Top: Chromatogram showing separation of 1 ng standards using HPLC. Bottom: Detection of basal levels of 5-HIAA and 5-HT in thoracic aorta ................................................................ 56 Basal levels of 5-HIAA and 5-HT in peripheral arteries from normal animals .............................................................. 58 Top: Representative of IHC using guinea pig ileum myenteric plexus as positive control for 5-HT staining Bottom: 5-HT IHC staining in rat aorta ............................... 61 5-HT staining in freshly dissociated rat aortic smooth muscle cells ............................................................................. 63 A. Real time RT-PCR amplification curves of tph1 mRNA expression in normal rat peripheral arteries ....................... 66 B. Real time RT-PCR dissociation curves of tph1 mRNA expression in normal rat peripheral arteries ....................... 67 A. TPH staining in freshly dissociated rat aortic and superior mesenteric artery smooth muscle cells B. TPH IHC staining in rat aorta ...................................... 69 Quantification of 5-HIAA, 5-HT and 5-HTP in an in vivo TPH activity study ................................................................ 72 Quantification of 5-HIAA, 5-HT and 5-HTP in A 5-HTP uptake study ................................................................. 75 Chromatogram showing the measurement of 5-HTP in tissues in An in vitro TPH activity study ............................. 77 Western analysis of AADC in rat peripheral arteries ............ 78 A. Western analysis examining MAO A in rat peripheral xi Figure 15. Figure 16. Figure 17. Figure 18. Figure 19. Figure 20. Figure 21. Figure 22. Figure 23. Figure 24. Figure 25. Figure 26 Figure 27. Figure 28. Figure 29. anefies B. MAO A IHC staining in rat peripheral arteries ................. Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in arteries from normal rats ................................................. Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in arteries from pargyline-treated rats .................................. Quantification of 5-HIAA and 5-HT in A 5-HT storage study in arteries from normal rats ............................................ Quantification of 5-HIAA and 5-HT in A 5-HT storage study in arteries from pargyline-treated rats ................................ Quantification of 5-HIAA and 5-HT in A 5-HT storage study in 5-HT-preloaded aorta from pargyline-treated rats ........... Western analysis examining 5-HT covalently bound protein in rat aorta .................................................................... Top: RT-PCR of SERT in rat aorta Bottom: Western analysis of SERT in rat aorta, superior mesenteric artery and mouse aorta ................................ SERT IHC staining in aorta from normal rat and mouse ...... SERT staining in freshly dissociated rat aortic smooth muscle cells .................................................................. Time course study of 5-HT uptake in normal rat superior mesenteric artery ........................................................... Time course study of 5-HT uptake in aorta from pargyline- treated rats at 37°C (top) and room temperature (bottom) ..... Time course study of 5-HT uptake in superior mesenteric artery from pargyline-treated rats at room temperature ........ Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in aorta from wild type and tph1-/- mice ................................. Effect of fluvoxamine and fluoxetine on 5-HT uptake in peripheral arteries from pargyline-treated rats ..................... Effect of fluvoxamine on 5-HT uptake in aorta from xii 80 83 86 88 91 93 94 97 99 102 103 105 107 109 112 Figure 30. Figure 31. Figure 32. Figure 33. Figure 34. Figure 35. Figure 36. Figure 37. Figure 38. Figure 39. Figure 40. pargyline-treated wild type, SERT HET and SERT KO mice.. Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in peripheral arteries from normal rats in Na*-free PSS ............ A. Glyoxylic acid fluorescence in small mesenteric resistance arteries from vehicle or 6-OHDA-treated rats B. Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in peripheral arteries from 6-OHDA-treated rats ........ Effect of nisoxetine on 5-HT uptake in peripheral arteries from normal rats ............................................................ Effect of corticosterone on 5-HT uptake in peripheral arteries from normal rats ............................................................ Quantification of 5-HIAA and 5-HT in A 5-HT uptake study in endothelium intact and denuded aorta normal rats .............. Quantification of 5-HIAA and 5-HT in fluramines-induced 5- HT release in aorta from pargyline-treated rats ................... Quantification of 5-HIAA and 5-HT in (+)-fenfluramine- induced 5-HT release in superior mesenteric artery from pargyline-treated rats ...................................................... Effect of fluoxetine on (+)-fenfluramine-induced 5-HT release in aorta from pargyline-treated rats ....................... Western analysis examining SERT expression in aorta from hypertensive and normotensive rats ................................. Quantification of basal levels of 5-HIAA and 5-HT in aorta from hypertensive and normotensive rats .......................... Effect of fluvoxamine and fluoxetine on 5-HT uptake in aorta from pargyline-treated SHAM(D) (Top) and DOCA rats (Middle) Comparison of total 5-HT uptake and SERT dependent 5- HT uptake in aorta from SHAM(D) and DOCA rats (Bottom)... xiii 114 116 119 120 122 124 127 129 131 134 137 140 Figure 41. Figure 42. Figure 43. Effect of fluvoxamine and fluoxetine on 5-HT uptake in aorta from pargyline-treated SHAMM (Top) and LNNA rats (Middle) Comparison of total 5-HT uptake and SERT dependent 5- HT uptake in aorta from SHAMM and LNNA rats (Bottom)... 142 Effect of fluvoxamine and fluoxetine on 5-HT uptake in aorta from pargyline-treated WKY (Top) and SHR rats (Middle) Comparison of total 5-HT uptake and SERT dependent 5- HT uptake in aorta from WKY and SHR rats (Bottom) .......... 144 Top: Effect of fluoxetine on a 5-HT concentration response curve in aorta from SHAM and DOCA rats Bottom: Effect of fluvoxamine on a 5-HT concentration response curve in aorta from SHAM and DOCA rats ............ 148 xiv LIST OF ABBREVIATIONS 5-HIAA 5-Hyd roxyindoacetic acid 5-HT 5-Hydroxytryptamine, serotonin 5-HTP 5—Hydroxytryptophan AADC Amino acid decarboxylase ACh Acetylcholine Ang II Angiotensin II AT 1 Angiotensin II receptor 1 BOA Bicinchoninic Acid BH4 Tetrahydrobiopterin BP Blood pressure BSA Bovine serum albumin CNS Central nervous system DOCA Deoxycorticosterone acetate 08 Dissociation solution DTI' Dithiothreitol ET—1 Endothelin- 1 L-NNA Nm-nitro-L-arginine MAPK Mitogen-activated protein kinase MAO Monoamine oxidase mCPP m-Chlorophenylpiperazine (:I:)-MDMA 3,4-methylene-dioxymethamphetamine MI Myocardial Infarction XV NE Norepinephrine NET Norepinephrine transporter NOS Nitric Oxide Synthase NSD 1015 3-Hydroxybenzylhydrazine OCT Organic cation transporters PBS Phosphate-buffered saline PCPA Para-chlorophenylalanine PE Phenylephrine PKA Protein kinase A PKC Protein kinase C PP2A Protein phosphatase 2A PPH Primary pulmonary hypertension PSS Physiological salt solution SBP Serotonin-binding protein SERT Serotonin transporter SHR Spontaneously hypertensive rats SNPs Single Nucleotide Polymorphisms SNS Sympathetic Nervous System SSRIs Selective serotonin reuptake inhibitors TBS Tris Buffered Saline TBS-T Tris Buffered Saline + Tween TPH Tryptophan hydoxylase VMAT Vesicular monoamine transporter TGs Transglutaminases TM Transmembrane domains TPR Total peripheral resistance WKY Wistar-Kyoto rats xvii Introduction: Serotonin (5-hydroxytryptamine, 5-HT), as a neurotransmitter in the brain and gastrointestinal tract, is involved in a wide variety of physiological functions such as mood control, urine storage and voiding, the regulation of sleep and body temperature, food intake and intestinal motility. 5-HT was first found as a powerful vasoconstrictor over a century ago and has been recognized as an arterial smooth muscle cell mitogen in the last 20 years (Nemecek et al., 1986). The vascular effects of 5-HT are associated with many vascular diseases including migraine (Buzzi et al., 2005), Raynaud’s phenomenon (Cooke JP and Marshall JM, 2005), atherosclerosis (Hara et al., 2004) and were suggested to contribute to liver tissue hypoperfusion following hepatic ischemia-reperfusion (Murata et al., 2003). In the past decade, a strong argument for the role of 5-HT and the serotonin transporter (SERT) in pulmonary hypertension has been built (Marcos et al., 2003; Eddahibi et al., 1999; Eddahibi et al., 2002; Eddahibi et al., 2000; MacLean et al., 2004; Morecroft et al., 1999). However, whether there is a functional serotonergic system present in peripheral arteries and the roles and mechanisms of circulating 5-HT in regulation of non-pulmonary, peripheral arterial function and its contributions to diseases are not clear. This section begins with an introduction of general background of 5-HT and SERT, followed by a summary of the roles of 5-HT and SERT played in physiological cardiovascular system and cardiovascular diseases, especially in hypertension. Biosynthesis, storage, and metabolism of 5-HT 1. Biosynthesis A. Biochemistry: The essential amino acid tryptophan is the precursor for 5-HT. Serotonin synthesis depends on the specific action and rate-limiting step of the enzyme tryptophan hydroxylase (TPH; EC 1.14.164), which transfers a hydroxyl group to the benzyl ring of tryptophan. Subsequent decarboxylation by amino acid decarboxylase (AADC) results in formation of 5-HT (Figure 1). The structure of 5-HT indicates it as a hydrophilic substance, which is preferably protonated at physiological pH. Thus, 5-HT does not pass the lipophilic blood—brain barrier and cell membrane readily. B. Location: Over 95% of 5-HT in the body is synthesized in the enterochromaffin cells of the intestine. 5-HT must be synthesized in the brain because 5-HT cannot cross the blood—brain barrier. Those areas responsible for synthesizing 5-HT are the clusters of cells in the midlinelraphe regions of the pons and upper brainstem. Other sources of 5-HT include neuroendothelial cells that line the lung, and a few other discrete sites (Cooper et al., 2003), some of which are in the cardiovascular system. TPH mRNA and protein have been detected in hamster heart (Slominski et al., 2002), and 5-HT synthesis has been measured both in an HL-1 cardiomyocyte cell line, which represents adult cardiomyocytes, and in neonatal rat ventricular cardiac myocytes (lkeda et al., 2005; Cote et al., 2004). Thus, the cardiomyocyte provides a local source of 5- HT in the heart. Both in vivo and in vitro experiments suggested a renal formation of serotonin by decarboxylation of its amino acid precursor L-5- 5-HT Biosynthesis and Metabolism Pathway /COOH CH2CHNH2 ©:§ Tryptophan NH Tryptophan Hydroxylase Rate Limiting /cooI-I HchNHz HO \ 5-Hydroxytryptophan (5-HTP) NH Amino Acid Decarboxvlase CH2CH2NH2 HO \ 5-Hydroxytryptamine (5-HT, Serotonin) 8 NH MAO Aldeh de Dehy rogenase <—— CH2COOH HO \ 5-Hydroxyindole Acetic Acid NH (5-HIAA) Figure 1. hydroxytryptophan (L-5-HTP) in rat (Stier et al., 1985; Stier et al., 1984). Serotonin synthesis has also been reported in adrenochromaffin cells of frogs (Delarue et al., 1992). Recent studies have revealed TPH in new and unexpected places such as the skin (Slominski et al., 2003). Sites of 5-HT synthesis in the non-pulmonary peripheral vasculature have not yet been identified, so our current understanding is that the systemic vasculature is exposed to 5-HT through release of 5-HT by the platelet or to freely circulating 5- HT. C. Regulation: TPH is an extremely labile enzyme, which can be affected by multiple factors. TPH uses Fe” as co-factor and Oz and tetrahydrobiopterin (BH4) as co-substrates to hydroxylate tryptophan. Additionally, an in vitro study suggested that TPH is an oxygen-inhibitable enzyme. The degree of TPH inactivation is dependent on the partial pressure of oxygen to which the enzyme is exposed and the temperature at which TPH is preincubated. TPH activity declined in a linear fashion as the temperature increased from 0 to 45 °C (Kuhn et al., 1979). The inactivation of TPH by oxygen could be recovered by anaerobic incubation in the presence of dithiothreitol (DTT) and Fe2+ (Kuhn et al., 1979). TPH activity can be irreversibly inactivated by nitric oxide (Kuhn and Arthur, 1997). The phosphorylation of TPH by protein kinase A (PKA) and CaZVcaImodulin-dependent protein kinase II results in increased TPH activity (Kowlessur and Kaufman 1999). The phosphorylation site for PKA has been reported at serine 58 (Kuhn et al., 1997) and at serine 58 and 260 for Ca2*/calmodulin-dependent protein kinase ll-induced phosphorylation (Jiang et al., 2000). The 14-3-3 proteins are a group of ubiquitous proteins that were first discovered in the brain (Aitken et al., 1995, review). It has been reported that the 14-3-3 proteins form a complex with phosphorylated rat brain TPH, thereby increasing TPH enzymatic activity and inhibiting the protein phosphatase-1 induced dephosphorylation (Banik et al., 1997). It is interesting that, unlike catecholamines, 5-HT synthesis by TPH is not inhibited by the end-product (5- HT) nor by the major metabolite (5-hydoxyindoleacetic acid, 5-HIAA) (Cooper JR et al., 2002). Thus, one way to study 5-HT synthesis is to inhibit AADC to examine the accumulation of 5-HTP. Recently, Bader and colleagues discovered that tph1 knock-out mice exhibit only a minor reduction of steady state 5-HT levels in brain regions (hippocampus and frontal cortex) with a lack of 5-HT in peripheral organs (gut, blood and pineal gland), suggesting the existence of a second tph gene, which they named tph2. The expression of tph1 is predominant in peripheral organs such as the intestine, spleen and pineal gland and tph2 is present in brain regions (Walther and Bader, 2003; Walther et al., 2003 a), which allows for distinct sources of peripheral and central 5-HT. TPH1 and TPH2 are highly homologous proteins exhibiting 71% of amino acid identity in human (Walther and Bader, 2003). All the residues important for tryptophan, iron, BH4 and 14-3- 3 proteins binding are identical in TPH1 and TPH2 (Walther and Bader, 2003). Consistently, both isoforms of TPH can be phosphorylated by Ca2*/calmodulin- dependent protein kinase II and PKA. However, the N-terminus of the two isoforms of TPH, which contain the regulatory domains, are quite different. The KM values of the purified and recombinant TPH from carcinoid tumors and pineal gland (TPH1) have been reported as 13 and 23 nM, respectively, while the TPH isolated from brain stem (TPH2) exhibits a value of 142 uM (Kowlessur and Kaufman, 1999), which suggests that the substrate of TPH, tryptophan, has higher affinity for TPH1. Pharmacology: Para-chlorophenylalanine (PCPA) is a potent, specific and irreversible inhibitor of TPH activity both in vivo and in vitro, which drastically reduces the 5-HT concentration in 5-HT neurons and terminals (Pandey et al., 1983). Amphetamine analogs inhibit TPH activity potentially via production of nitric oxide, superoxide and peroxynitrite (Kuhn and Geddes, 2000). The interaction of 5-HT and these reactive oxygen species produces tryptamine-4,5- dione, which rapidly and irreversibly inactivates TPH (Wrona and Dryhurst., 2001). 3-Hydroxybenzylhydrazine (NSD 1015) is an AADC inhibitor, which has been used in experiments to block the conversion of 5-HTP to 5-HT (Pandey et aL,1983) 2. Storage A. Location: In the periphery, the major 5-HT storage site is platelets. Excluding platelets, the free circulating levels of 5-HT in plasma were reported as 15-120 nM, much lower than the levels of 5-HT in whole blood (uM) (Martin 1994). Other than playing a crucial role in 5-HT synthesis, enterochromaffin cells are also a storage place of 5-HT (Nilsson et al., 1985). In the neuronal system, 5-HT is synthesized in the axon and stored in nerve terminals. Other 5-HT storage sites including mast cells (Pihel et al, 1998), adrenal medullary cells (Brownfield et al., 1985) and pinealocytes (Hayashi et al., 1999). B. Mechanisms: PM Platelets themselves do not synthesize 5-HT. However, they possess a high efficacy SERT, enabling them to take up 5-HT from the gut and lung. As the carrier and storage site of 5-HT, the platelets store 5-HT in dense, electron- opaque granules and release it in a thrombotic event (Vanhoutte, 1991). Storage of 5—HT in platelet granules requires active uptake of 5-HT from the cytoplasm by vesicular monoamine transporter 2 (VMAT 2). Transglutaminases (TGs) catalyze the calcium-dependent acyl transfer between the y-carboxamide of a bound glutamine residue and the s-amine group of a bound lysine, the primary amine group of a polyamine or possibly monoamine, such as 5-HT (Zhang et al., 1998). Recently, Walther and his colleagues reported that platelet intracellular 5-HT could be transamidated to small GTPases by TGs during activation and aggregation of platelets, rendering these GTPases constitutively active (Walther et al, 2003 b). This observation suggested that 5-HT could be covalently bound to proteins and thus to be stored in platelets. Enterochromaffin Cells: Study of the subcellular localization of serotonin immunoreactivity in rat enterochromaffin cells showed that 60% of 5-HT was located in the dense cores of the secretory granules following uptake via VMAT 1. However, a significant amount of 5-HT (40%) was located in the cytoplasm, outside of the secretory granules (Nilsson et al., 1985), which suggests two different forms of 5-HT storage exist, at least in enterochromaffin cells. New After being synthesized in neurons, 5-HT is taken up by VMAT 2 and stored in synaptic vesicles. Different from other non-neuronal 5-HT storing cells such as enterochromoffin cells, mast cells or platelets, soluble serotonin-binding protein (SBP) has been found in serotonergic synaptic vesicles of central and peripheral serotonergic neurons. This difference might come from different origins of the cells. SBP appears to be neuroectoderm-specific, while enterochromaffin cells are from endodermal origin, and mast cells and platelets are derived from mesoderm (Gershon et al., 1983). SBP binds with 5-HT with high affinity in the presence of Fe”. Two SBPs have been identified, with molecular weights of 45 Kd and 56 Kd. Thus, SBP binds 5-HT and functions as a storage protein for 5-HT in neurons (Gershon et al., 1983). Mast Cells: Mast cells store serotonin and histamine together within large secretory granules (Pihel et al, 1998). Moreover, two serotonin binding proteins were found in mast cells which are different from the SBP found in brain (Tamir etaL,1982) Thus, from what has been discovered, 5-HT could be stored in vesicles or bound to proteins in cells. C. Regulation and Pharmacology: VMAT functions as a proton-amine exchanger with a stoichiometry of two protons to one amine, which could be 5- HT, dopamine, histamine etc. The electrochemical gradient across secretory vesicle membranes (due to the activity of H+-ATPase) provides energy for the transport of monoamines. As indicated above, two types of VMAT have been discovered, VMAT 1 primarily in endocrine cells [enterochromaffin cells, pinealocytes (Hayashi et al., 1999)] and VMAT 2 in neuronal cells and platelets. The two VMATs differ in their ability to transport histamine, and in their sensitivity to certain inhibitors. For example, reserpine blocks both, but tetrabenzine is selective for VMAT2 (Peter et al. 1994). Recently, a study showed that the filling of 5-HT in vesicles initiates an activation of Gaq protein (probably on vesicle membrane), which by an unknown 5-HT receptor-independent mechanism downregulates VMAT 2 function in platelets (Holtje et al., 2003). Because VMAT function depends on the electrochemical gradient induced by H"-ATPase, any chemical (such as concanamycin and bafilomycin), which inhibits H+-ATPase will reduce VMAT function and therefore 5-HT storage in vesicles. TGs transamidate 5-HT to proteins. Cystamine inhibits TGs function and thus potentially reduces 5-HT storage in the form of 5-HT-transamidated proteins. 3. Metabolism A. Biochemistry: Metabolism of 5-HT primarily occurs through actions of deamination by monoamine oxidase (MAO) to form 5-hydroxyindole acetaldehyde, which in turn is oxidized by aldehyde dehydrogenase to produce 5-HIAA (Figure 1). Alternatively, 5-hydroxyindole acetaldehyde can be converted to 5- hydroxytryptophol, but this pathway is considered insignificant. Other metabolism pathways include transformation of 5-HT to melatonin. N—acetyl transferase induces acetylation of the amine group of 5-HT to N-acetyl-5-HT. Then, N-acetyl-5-HT is converted to melatonin by adding a methyl group (Cooper JR et al., 2003). B. Location: MAO is an intracellular enzyme, found primarily in mitochondria. 5-HT must be taken up inside a cell prior to being acted upon by MAO and both SERT and norepinephrine (NE) transporter (NET) facilitate this uptake (Kawasaki and Takasaki, 1987). Tissues or cells that contribute significantly to 5-HT metabolism include the lung, intestine and endothelial cells of the arterial system, but any cell that can take up 5-HT and possesses MAO has the potential to metabolize 5-HT. In pineal gland, melatonin synthesis uses 5-HT as a precursor. C. Regulation and Pharmacology: Two isoforms of MAO have been found, MAO A and MAO B. MAO A is more selective and has a much lower KM value (higher affinity with 5-HT) in metabolizing 5-HT compared to MAO B. MAO A activity increased in rats with low salt diet. This increased activity of MAO A was proved to be related to angiotensin II receptor 1 (AT 1) activation due to increased angiotensin II levels and an increased AT 1 receptor expression induced by low salt diet (De Luca Sarobe et al., 2005). Glucocorticoid increased both MAO A mRNA and protein expression via glucocorticoid receptor and Sp1 transcription factor function (Manoli et al., 2005). Pharmacologically, MAO could be inhibited by pargyline. 10 Il. Uptake and release of 5-HT 5-HT is a charged molecule (protonated) in physiological condition, which cannot cross lipid bilayer. SERT is the major protein responsible for uptake and release of 5-HT, which transports 5-HT in either direction, depending on the concentration gradient. SERT is important in that it plays a critical role in regulating the function of the 5-HT receptors and the serotonergic system via modulation of extracellular and intracellular cytoplasm 5-HT concentrations. 1. The major component responsible for 5-HT uptake and releaseu-SERT A. Molecular Biochemistry: SERT was cloned in 1991 by Hoffman and Blakely in the rat (Hoffman et al., 1991; Blakely et al., 1991). Human SERT was cloned in placental trophoblastic cells in 1993 (Ramamoorthy et al., 1993); the mouse SERT was cloned in 1996 (Chang et al., 1996). Mouse SERT protein has an 88% homology with the human SERT, compared to the rat SERT having 71% homology for the human SERT. SERT proteins are positioned in the plasma membrane. Hydropathy plots of the SERT protein, typically 630 amino acids long, have suggested that the protein spans the bilayer 12 times (12 transmembrane domains; TM) and that both amino and carboxy termini are intracellular (Figure 2 top). SERT possesses a large extracellular loop between TM3 and 4. This loop has sites of glycosylation, important for the trafficking and stability of SERT (Chen et al., 2002). In three sites of the protein, the amino terminus, the carboxy terminus and the intracellular loop between TM8 and TM9, consensus sites for 11 Proposed Topology of Plasma-membrane SERT Inhibitor Interaction, Cation Dependence ‘IL ,1 1 OUT . 112EI§5BILE§91> ho Ema—2n. Figure 3. 41 Methods: I. Animal Uses All animal procedures were followed in accordance with the institutional guidelines of Michigan State University. Normal male Sprague-Dawley rats (225- 250 g) were purchased from Charles River (Portage, MI) or Harlan Industries, Inc. (Indianapolis, IN). Male SHR and VWstar-Kyoto rats (WKY, 12 weeks) were purchased from Taconic Farms, Inc. (Gerrnantown, NY). Normal male CS7BLI6 mice (20-24 g) were purchased from Charles River (Portage, MI). The SERT targeted mutation mice and wild type male mice (C57BU6; 30-40 g) were received from Dr. Dennis L. Murphy, National Institute of Mental Health. The Tph1 -/- mice and wild type mice (C57BU6, 20-24 g) were received from Dr. Michael Bader, Max Delbriick Center for Molecular Medicine (Germany). Until surgery, the rats were kept in clear plastic boxes with free access to standard rat chow (Teklad ®) and tap water. II. Euthanasia: Rats were anesthetized with pentobarbital (60 mg/kg, i.p.). Mice were sacrificed by asphyxiation with carbon dioxide. III. Animal Models: Mineralocorticoig Hypertension: Male Sprague-Dawley rats (250-300 9; Charles River, Portage, MI) were anesthetized with isoflurane (IsoFlo®). Animals were uninephrectomized and a 42 Silastic® (Dow Corning, Midland, MI) implant impregnated with DOCA pellet (200 mg/kg) was placed subcutaneously on the back of the neck. Postoperatively, rats were given a solution of 1% NaCI and 0.2% KCI for drinking. Sham rats also received a uninephrectomy, but received no DOCA implant and drank normal tap water. Animals were fed standard rat chow and had ad Iibitum access to food and water. The animals remained on the regimen for four weeks prior to use. L-NNA Hypertension Male Sprague-Dawley rats (250-300 g; Harlan, Indianapolis, IN) were given tap water mixed with Nw—nitro-L-arginine (L-NNA, 0.5 g/L). The animals were remained on the regimen for two weeks prior to use. 6-OHDA Denervetion Sympathetic neuronal denervation was induced by 6-hydroxydopamine (6- OHDA) injections (McCafferty et al., 1997). Male Sprague-Dawley rats were treated with four doses of 6-OHDA over 7 days (50 mg/kg on days 1 and 2 and 100 mg/kg on days 6 and 7; 0.1% ascorbic acid in physiological saline as vehicle i.p.). Rats were euthanized (pentobarbital, 60 mg/kg i.p.), and arteries were removed on day 8. Denervation was validated by glyoxylic acid staining in mesenteric resistance arteries. Glyoxylic acid staining: Mesenteric resistance arteries were removed. After fat was trimmed off, arteries were immersed into glyoxylic acid (2%) for 5 min. Blood vessels were mounted on a microscope slide 43 and placed in an oven (100°C) for 5 min. The slides were removed and blood vessels mounted in mineral oil and cover-slipped. Vessels were viewed using a fluorescence microscope (Nikon LABOPHOT) and UV illumination (G-1A; excitation, 546/10 nm; barrier filter, BA580). The absence of a fluorescent network of staining of catecholamines in arteries confirms the effectiveness of 6- OHDA in causing sympathetic denervation. IV. BP Measurements: Systolic BP of conscious rats were determined by the tail cuff method using a pneumatic transducer. Briefly, the rat was placed in a plastic pail with wood shavings covering the bottom. This pail was then placed on a heating pad and the rat was contained in the bucket by a small metal cage. A warming light was placed over the bucket. The rat was warmed for approximately 6 min. This allowed vasodilatation of the tail artery, which facilitated the measurement of the BP. The rat was placed in a restraint and the blood pressure cuff and balloon transducer was placed on the tail and secured with tape. The BP was measured utilizing a sphygmomanometer in conjunction with the pulse transducer. Three blood pressure measurements were taken to obtain an average measurement. V. 5-HT Basal Level Measurement Aorta from normotensive and hypertensive animals were dissected, cleaned and placed in 75 IIL of 0.05 mM sodium phosphate & 0.03 mM citric acid 44 buffer (pH 2.5) containing 15% methanol. Samples were frozen at -80°C at least 4 hours. VI. TPH Activity Assay In vivo 5-HT §vnthefi Aorta, superior mesenteric artery and hypothalamus from NSD1015 (100 mg/kg, 30 min) or NSD1015 (100 mg/kg) + L-tryptophan methyl ester hydrochloride (300 mg/kg, 60 min)—treated rats were dissected, cleaned in physiological salt solution (PSS, 103 mM NaCl; 4.7 mM KCL; 1.18 mM KH2PO4; 1.17 mM MgSO4-7H20; 1.6 mM CaCl2-2H20; 14.9 mM NaHCO3; 5.5 mM Dextrose, and 0.03 mM CaNazEDTA) and placed in tissue buffer [0.05 mM sodium phosphate & 0.03 mM citric acid buffer (pH 2.5) containing 15% methanol]. Samples were frozen at —80 °C until assay. In vitro 5-HT synthesis Aorta, superior mesenteric artery and dorsal raphe nucleus from normal rats were dissected, cleaned in incubation solution [100 uM Fe(NH4)(SO4)2, 1 mM DTT, 50 mM Tris-HCI, pH 7.4]. Tissues were incubated in 1.5 ml plastic centrifuge tubes with incubation solution plus 3250 U/ml catalase and 10 mM NSD 1015 at 37 °C for 30 min. Then 200 uM BH4 and either H20 or 400 uM tryptophan were added. After 40 min incubation, tissues were rinsed in drug-free PSS, placed in 75 pL tissue buffer and saved at —80 °C until assay. 45 VII. 5-HT Storage Assay At room temperature, naive or 5-HT-Ioaded (5-HT 1 uM, 15 min) aortae from untreated or MAO A inhibitor pargyline (100 mg/Kg)-treated rats were incubated with PSS in 1.5 ml plastic centrifuge tubes for 4 or 8 hours. Tissues were then briefly dipped In drug-free PSS and placed in 75 uL tissue buffer. Samples were frozen at —80 °C until assay. VIII. 5-HT Uptake Assay: At room temperature (unless otherwise specified), dissected and cleaned arteries from normal or pargyline (100 mg/Kg)—treated animals were placed in PSS in 1.5 ml plastic centrifuge tubes containing either vehicle or inhibitor for 30 min. 5-HT (1 uM) or vehicle (water) was then added for a varying amount of time (time course studies) or for 15 min in experiments with inhibitor. Tissues were then briefly dipped in drug-free PSS and placed in tissue buffer. Samples were frozen at —80 °C until assay. In some experiments, the endothelium was removed by gently rubbing the luminal face of the artery with a moistened cotton swab. IX. 5-HT Release Assay: At room temperature, dissected and cleaned arteries from normal or pargyline (100 mg/Kg)-treated animals were placed in 100 uL PSS or SERT inhibitor (diluted in PSS) in 250 uL microcentrifuge tubes for 30 min. Water or (+)-norfenfluramine (1O uM), in some experiments (+)-fenfluramine (1, 10 uM), was then added for 20 min. Tissues were taken out of the tubes and the PSS 46 solution in tube was saved on ice (PSS samples) and used to test 5-HT and 5- HIAA release. Tissues were briefly dipped in drug-free PSS and placed in tissue buffer (tissue sample, used to test 5-HT and 5-HIAA left in tissues). PSS samples and tissue samples were frozen in -80 °C until assay. X. 5-HIAA and 5-HT Measurement: Samples were thawed, sonicated for 3 seconds and centrifuged for 30 seconds (10,000 g). Supernatant was collected and transferred to new tubes. Tissue pellets were dissolved in 1.0 M NaOH and assayed for protein. Concentrations of 5-HIAA and 5-HT in tissue supernatants and PSS samples from 5-HT release assay were determined by isocratic High Performance Liquid Chromatography (HPLC) coupled with electrochemical detection. Fifty microliters of tissue supernatant or PSS sample was injected onto a C18 reverse phase analytical column (Biosphere ODS, West Lafayett, IN) protected by a precolumn cartridge filter. This column was coupled to a single coulometric electrode conditional cell in series with dual electrode analytical cells (ESA, Bedfore, MA). The conditioning electrode potential was set at 0.4V, while the analytical electrodes were set at 0.12 and —0.31 V relative to the internal silver reference electrodes. Amounts of 5-HIAA and 5-HT were determined by comparing peaks areas in samples with those obtained from standards run the same day, and reported as a concentration relative to protein content. The lower limit of sensitivity for detection of 5-HIAA and 5-HT was 2-5 picogram/sample. 47 XI. Real Time RT-PCR: Two-step RT—PCR was performed using a GeneAMP 7500 Real Time PCR machine (Applied Biosystems, Foster City, CA). Total RNA from rat aorta and superior mesenteric artery were isolated using the MELT Total RNA isolation System Kit (Ambion, Austin, Taxes). Concentration of RNA was measured spectrophotometrically (A260/A280). One microgram of total RNA was reverse transcribed using a Taqman® reverse transcriptase kit (Applied Biosystems, Foster City, CA, USA; buffer, MgCIz 5.5 mM, dNTP 500 uM, of each random hexamer 2.5 uM, RNase inhibitor 0.4 pg/uL and MultiScribe Reverse Transcriptase 1.25 ug/uL; 10 min hold at 25°C, 30 min hold at 48°C, 5 min hold at 95°C). One microliter of this cDNA was taken through PCR using a SYBR® Green Master Mix (Applied Biosystems, Foster City, CA, USA). PCR conditions were: 95°C 10 min for AmpliTAQ® activation, 40 cycles of PCR (15 seconds 95°C, 60 seconds 60°C). Two sets of rat tph1 primer pairs were used. One of the rat tph1 primer pair, rat [it-actin primer pair and B-2-microglobulin (B2m) pair were purchased from Superarray (Frederick, MD). An alternative rat tph1 primer pair was designed based on RefSeq Accession number: XM-341862.1 and was synthesized by the Macromolecular Structures and Synthesis Facility at Michigan State University. The rat tph1 primer fon~ard= GCC TGC TTT CTT CCA TCA GT. The rat tph1 primer reverse= AGA CAT CCT GGA AGC 'ITG TGA. The SERT primer forward= GGC CAG TAC CAC CGA AAC, SERT primer reverse = CGG GGC AGA TCT TCC TCC ATA T. 48 CT values were derived as the threshold cycle at which product was first detected and are reported as cycle numbers. XII. lmmunohistochemistry: Prepagtion of frozen sections: Tissues were frozen in OCT compound and stored at -80°C until use. Sections were cut, cold acetone fixed, washed 3 times with phosphate-buffered saline (PBS). Prepaflttion of paraffin-embedded sections: Formalin-fixed, paraffin-embedded rat thoracic aorta and superior mesenteric artery were washed twice in xylenes and four times in 90% ethanol for 3 min each to dewax. Sections were unmasked by microwaving them twice for 3 min in Vector Antigen Unmasking Solution. Endogenous peroxidase in fixed frozen sections or dewaxed, unmasked paraffin-embedded section were blocked [0.3% H202 in phosphate-buffered saline, PBS, for 30 min]. Sections were blocked for non-specific binding in PBS containing 1.5% of competing serum. In a humidified chamber, samples were incubated overnight with antibody (10 pg/mL, anti-TPH antibody, Sigma; 5 ug/mL anti-SERT, C-20, Santa Cruz; 5 pg/mL, anti-5-HT YC5/45, Abcam, UK), antibody neutrialized with 5-fold excess of competing peptide or blocking serum with antibody. The remaining steps were carried out in accordance to the manufacturer’s instructions (Vector Laboratories, Burlingame, CA, USA). Sections were washed 3 times with PBS and incubated with a peroxidase- conjugated secondary antibody (30 min, room temp). Samples were washed and 49 incubated with Vectastain® ABC Elite reagent (30 min, room temp) followed by 3,3’-diaminobenzidine (DAB)/H2O2. The reaction was stopped with washing, sections were air dried, hematoxylin-stained, mounted and photographed using an inverted Nikon microscope with a Spot digital camera. XIII. lmmunocytochemistry: Rat thoracic aorta and superior mesenteric artery were dissected and cut into small rings in chilled dissociation solution [D8, containing (M): NaCI 0.137; KCI, 0.0056; MgCl2, 0.001; Na2HPO4, 0.00042; NaH2PO4, 0.00042; NaHCO3, 0.0042; Na nitroprusside 2.6 mg/L and HEPES, 2.383 mglL; pH=7.4]. Small vessel rings were incubated with enzymatic solution (containing papain, 26 U/ml and DTT, 1 mg/ml) for 35 min and other enzymatic solution (containing, type II collagenase 2.5 U/ml, elastase 0.15 mg/ml and soybean trypsin inhibitor, 1 mg/ml) for 45 min. We then draw off other enzymatic solution and gently added 3 ml of fresh DS and left on ice for 5 min, which was followed by draw off 08 without disturbance of the cells at the bottom. The cells were then triturated in D8 containing Na nitroprusside (0.01 mM). One hundred uL of titrated cells were put on polylysine coated coverslips and left in incubator (37°C, 5% CO2) for 30- 45 min. After the cells attached to coverslips, the DS was carefully removed and the cells were fixed with Zamboni’s fixative (20 min). The cells were rinsed with low-salt PBS twice (5 min each time) and cell membrane was perrneablized with 1% triton (5 min) followed by blocking serum incubation (5% serum, 20 min). Cells were then incubated with primary antibody for 2 hours (37°C, anti-5-HT 50 antibody, anti-TPH antibody, 1:200, Sigma, anti-SERT antibody 1:200). Cells were washed with low-salt PBS three times, followed by 1 hour incubation with goat-anti-mouse Cy3 antibody or donkey-anti-goat Cy3 antibody (37°C, 1:1000, Jackson ImmunoResearch). The coverslips were then mounted and photographed using an inverted Nikon microscope or a confocal microscope with a digital camera. Images in this dissertation are presented in color. XIV. Protein Isolation: Rat/mouse thoracic aorta and rat superior mesenteric artery were removed from the animal and placed in PSS and cleaned as described above. Arteries were quick frozen and pulverized in a liquid nitrogen-cooled mortar and pestle and solubilized in lysis buffer [0.5 M Tris HCI (pH 6.8), 10% SDS, 10% glycerol] with protease inhibitors [0.5 mM phenylmethylsulfonyl fluoride, 10 uglul aprotinin and 10 ug/ml Ieupeptin]. Homogenates were centrifuged (11,000 g for 10 min, 4 °C). We utilized the Bicinchoninic Acid (BCA) method for protein measurement (Sigma, St.Louis, MO). Samples were stored at —80 °C until use. XV. BCA Protein Assay: The bovine serum albumin (BSA) protein standard, consisting of a known concentration of BSA, was utilized to make the standard curve to which the protein samples were compared. The working reagent was made by mixing BCA with Copper (II) Sulfate (50:1). To determine the protein concentrations of 51 samples, 5 pL protein from each sample, 95 pl H20 and 2 mL working reagent were mixed and incubated for 30 min at 37 °C, no C02. The samples were analyzed on a spectrophotometer at an absorbance of 562 nm and the protein concentration determined by plotting these values on the standard curve. XVI. Western Blotting: Tissues homogenates (4:1 in denaturing sample buffer, boiled for 5 min) were separated on SDS-polyacrylamide gels and transferred to Immobilon-P membrane. Membranes were blocked for 3-4 hours (Tris Buffered Saline, TBS, 4% chick egg ovalbumin or 5% dry milk, 0.25% sodium azide). Blots were probed overnight with primary antibody (4 °C), rinsed in TBS-Tween (TBS-T, pH 7.6) (20 mM Tris, 137 mM sodium chloride and 0.1% Tween-20), with a final rinse in TBS (pH 7.6) (20 mM Tris and 137 mM sodium chloride) and incubated with secondary antibody for 1 hour at 4 °C following with TBS-T and TBS washes. Blots were incubated with ECL® reagents to visualize the bands. XVII. Isolated Smooth Muscle Contractility Measurement: The thoracic aorta was removed and placed in PSS. The aorta was cleaned of fat and connective tissue and cut into helical strips. Aortic strips were then mounted onto stainless steel rod holders and placed into 50 ml tissue baths for isometric tension recordings using Grass polygraphs and transducers. Strips were placed under optimum resting tension (1,500 mg for rat aorta, determined previously) and allowed to equilibrate for one hour before exposure to 52 compounds. In experiments testing tissues from hypertensive animals, one aortic strip isolated from a normotensive control and one aortic strip from a hypertensive rat were placed in the same bath, thereby controlling for potential experimental variations. Tissue baths contained warmed (37 °C), aerated (95% 02/002) PSS. Administration of an initial concentration of 10 pM PE was used to test arterial strip viability; the strips must contract to a minimum of 500 mg for rat aorta to be considered viable. To examine the status of the arterial endothelium, tissues were contracted with a half-maximal concentration of PE (10-100 nM) and once the contraction plateaued, the muscarinic agonist acetylcholine (ACh, 1 uM) was administered. The observation of a relaxation to ACh greater than 60% of the PE (10-100 nM) -induced contraction was considered as endothelium- intact. Cumulative concentration curves were performed to agonists. Antagonists, inhibitors or vehicle were incubated with the vessels for one hour prior to the experimentation. XVIII. Data Analysis and Statistics: Data are presented as means :I: standard error of the mean for the number of animals in parentheses. 5-HIAA and 5-HT concentrations were quantified using standards run the same day, and reported as a concentration relative to protein content. Contractions are reported as force (milligrams) or as a percentage of response to maximum contraction to PE. ECso values (agonist concentration necessary to produce a half-maximal response) were determined using non- 53 linear regression analysis in Prism version 4.0 (San Diego, CA) and were reported as the mean of the negative logarithm (-log) of the EC50 value (M). These values represent the concentration necessary to produce a half-maximal response in each tissue using the tissue’s own maximum response. Band density quantitation in Western analyses was performed using NIH Image (v.1.61). For each sample, the densities of the tested bands on Western blotting are normalized to the density of the corresponding actin band. When comparing two groups, the appropriate Student’s t-test was used. When comparing 3 or more groups, an ANOVA followed by Newman Keuls post hoc test was performed. In all cases, a p value less than or equal to 0.05 was considered statistically significant. 54 Results Hypothesis #1: A local serotonergic system, Including synthesis, metabolism, storage, uptake and release of 5-HT is present and functional In peripheral vasculature. We investigated the serotonergic system in peripheral arteries on the following aspects: (1) presence of 5-HT; (2) 5-HT synthesis and metabolism; (3) 5-HT storage; (4) 5-HT uptake and release. I. The Presence of 5-HT in Peripheral Arteries Whether peripheral arteries contain endogenous 5-HT has not been reported. We used three different experimental techniques, which provided the same results suggesting the presence of 5-HT in peripheral arteries. By using HPLC analyses, we detected the content of 5-HT and its MAO-A metabolite 5-HIAA in aorta, superior mesenteric artery and carotid artery. Figure 4 shows a standard chromatogram (Top, a representative of all standard tracings in data from HPLC) and a chromatogram of basal 5-HT and 5-HIAA in rat thoracic aorta (bottom). The quantification of basal 5-HIAA and 5-HT levels in rat aorta and superior mesenteric artery and mouse aorta are reported in Figure 5. Figure 6 depicts the results of IHC experiments using a 5-HT antibody, localizing 5-HT to rat aorta. The presence of 5-HT in peripheral arteries was 55 6 Mix Standard :25. 1:7" w 19.175 5-HIAA awry ” { 7 3m 19.319 {r 5-HT TII‘IET'FIBLE ST 0P Basal Level 5-HIAAI5-HT in Rat Aorta -I L———~— to .391 §' é 5-HIAA ID (.2 19.724 - rrrrerasu: srops'HT Figure 4. Top: Chromatogram showing separation of 1 ng standards using HPLC. Bottom: Detection of basal levels of 5-HIAA and 5-HT in thoracic aorta. 5-HIAA = 5-hydroxyindoleacetic acid. 5-HT = 5-hydroxytryptamine. 56 Figure 5. Basal levels of 5-HIAA and 5-HT in rat aorta (top), rat superior mesenteric artery (middle) and mouse aorta (bottom) from normal animals. 5-HlAA=5—hydroxyindoleacetic acid, 5-HT=5-hydroxytryptamine, SMA=superior mesenteric artery. 57 Rat Aorta (N=7I f o 2 5 1 c 1 a O 5-HT 5-HIAA 0 .5395 9:55 cozfiucoocoo Rat SMA (N=4) T 5-HT a 1 ..u. 1 g... 0 D 0 3355 9:35 :oSEEoocoo Mouse Aorta (N=4) Figure 5. 5-I'-IT o 2 5 1 o. 1 5 0 £ 0 .5995 9:55 cozfluceocoo 58 supported by staining of the aorta, throughout the smooth muscle layers and adventitia, with an antibody that recognizes 5-HT (Y05/45, Figure 6, bottom left). Staining was significantly diminished when the antibody was pre-incubated with a 5-fold excess of 5-HT itself (Figure 6, bottom right). The dark precipitate formed by DAB in serotonergic nerves of the guinea pig ileum myenteric plexus (Figure 6, top) was used as a positive control as this tissue contains a rich innervation of serotonergic nerves (Galligan et al., 1986). To eliminate the possibility that the 5- HT we observed in aorta section is actually in mast cells rather than in smooth muscle cells, we used Giemsa’s Stain to test the existence of mast cells in rat aorta. The aorta only rarely presented with recognizable mast cells while this staining detected abundant mast cells in a human lymph node (data not shown). Thus, it is unlikely that aortic 5-HT comes from mast cells. Intracellular staining of 5-HT using the 5-HT antibody (goat anti-rabbit 5- HT, Serotec, Raleigh, NC) and a fluorescent Cy3 secondary antibody in freshly isolated aortic smooth muscle cells further confirmed the existence of endogenous 5-HT in arterial smooth muscle (Figure 7, B). Using the same exposure time, no fluorescence or very low fluorescence was observed in cells with only secondary antibody incubation (Figure 7, C). The staining of a-actin verified that the cells were smooth muscle cells (Figure 7, A). 59 Figure 6. Top: Positive control for use of the 5-HT antibody (YC5/45, Abcam); guinea pig ileum myenteric plexus contains identifiable 5-HT containing neurons marked with the arrow. Bottom: Immunohistochemical staining for 5-HT in aorta from normal rats. Aortic section incubated with 5-HT primary antibody (left) compared with section incubated with primary antibody prequenched with 5-HT. Arrows indicate the placement of staining. Representative of 4 different experiments/rats. 5-HT = 5-hydroxytryptamine. 60 Presence of 5-HT in Rat Aorta Guinea Pig lleum Myenteric Plexus POSItIve Controlfor 5- HT Rat Aorta 5-HT Primary +, 5-HT‘ Prima fig; ' ' fl“ ’V’ I" ' f -’ 5}” ' Lumen Figure 6. 61 Figure 7. Freshly dissociated rat aortic smooth muscle cells stained with anti-a-actin antibody (A), with anti-5-HT antibody (B) or secondary antibody alone (C). Representatives of four separate experiments, each with a different rat. 5-HT = 5-hydroxytryptamine. 62 Presence of 5-HT in Freshly Isolated Rat Aortic Smooth Muscle Cells Rat Aorta No primary Figure 7. 63 II. The Presence of 5-HT Synthesis and Metabolism in Peripheral Arteries To investigate the synthesis and metabolism system in peripheral arteries, we tested the existence and activity of enzymes that are important for serotonin synthesis and metabolism: TPH, AADC and MAO A. 1. The Existence of TPH1 mRNA and Protein in Peripheral Arteries We first investigated the existence of mRNA in peripheral arteries. Two sets of rat tph1 primer pairs were used in our experiments. The first rat tph1 primer pair was purchased from Superarry (Frederick, MD). This primer was designed according to RefSeq Accession #: XM_341862.1. We also designed another rat tph1 primer. The sequences were rat tph1-L: GCCTGCTI'TCTTCCATCAGT and rat tph1-R: AGACATCCTGGAAGCTTGTGA. Figure 8 A. shows the amplification plots of tph1 and the housekeeping gene (3-2— microglobulin (B2m) in rat aorta and superior mesenteric artery. CT values were measured as the value at which measurable product was first observed in real time RT-PCR. The CT values for tph1 and B2m in rat aorta and superior mesenteric artery are reported in Table 1. Figure 8 B. shows the dissociation curve for tph1 (both primers) in aorta and superior mesenteric artery. Only one peak was observed in each of the dissociation curve, indicating the single product of tph1 primer. The translation of tph1 mRNA was confirmed by the immunocytochemistry experiments in which we observed an expression of TPH protein in freshly isolated aortic and superior mesenteric smooth muscle cells (Figure 9, top) using 64 Figure 8. A. Real time RT-PCR amplification curves of tph1 mRNA expression in normal rat aorta (top) and superior mesenteric artery (bottom) using tph1 primer purchased from SuperArray® (left, Frederick, MD) or using tph1 primer designed by us (right). B. Real time RT-PCR dissociation curves of tph1 mRNA expression in normal rat aorta and superior mesenteric artery using tph1 primer purchased from SuperArray® (top, Frederick, MD) or using tph1 primer designed by us (bottom). Table 1. The CT values in real time RT—PCR experiments detecting the expression of tph1 and B-2-microglobulin mRNA in rat aorta and superior mesenteric artery. B2m=8—2-microglobulin, tph=tryptophan hydroxylase, SMA=superior mesenteric artery. 65 A. tph1 SuperArray 1.0e+001 . -. 1.06.00, 1-°e*°°° W" 1.0e+000 ' .59 ' to E 1-09'003 251.012003 0 w ,. , 0 1.019004» " 1.06004 1.0e-oos~-~i——'— '1 .. « 1.064305 . , . Rat Aorta Rat Aorta 107006 5 15 25 35 1-°°‘°°° 5 15 25 35 Cycles Cycles tph1 S erArra tph1 1 08+001~~ up y 1,0e+001-- - .- 1 .Oe+000 1 .0e+000 1 .0e-001 1.0e-001 é 1.0e-002 0:: 1.09002 9 1.05003 9 105003 8 1.05004 8 ' 1.05005 1-06-004 mm“ 1 .0e-005 ,‘ "°°°°° Rat 8m Ralt SMA l - 1.0 005 - 1'06 007 5 15 25 35 e“ 5 15 25 35 Cycles Cycles Figure 8. 66 tph1 SuperArray Table 1. Derivative tph1 Derivative Dissociation Curve 0.40 0.35 I 0.30 0.25 0.20 0.15 0.10 0.05 -0.00 -o.05 ‘ so 65 7o 75 so 85 90 90 Temperature Dissociation Curve l 0.40 l . 0.35 ., j . 0.30 0.25 .. “We - _- 0.20 0.15-E . g...- 0.10 Em.--“- - .. -____,- 0.05 -._c. -0.00 -0.05 ‘ 60 65 70 75 80 85 90 90 Temperature Figure 8. tph1 (SuperArray) tph1 B2m Rat Aorta SMA 24.922057 27.02:1.06 28.77::033 16.811025 30.351021 177020.32 67 Figure 9. A. Freshly dissociated rat aortic smooth muscle cells (top) and superior mesenteric arterial smooth muscle cells (bottom) stained with anti-TPH antibody. Representatives of four separate experiments, each with a different rat. B. Immunohistochemical staining of the TPH (Sigma) in smooth muscle between cables of elastin/collagen in the rat aorta frozen section. Parallel sections were incubated with antibody (left) or no primary antibody (right). Arrows indicate the placement of staining. TPH= tryptophan hydroxylase, SMA=superior mesenteric artery. 68 Presence of TPH Protein in Freshly Isolated A. Arterial Smooth Muscle Cells Rat Aona Rat SMA B. Presence of TPH Protein in Rat Aorta Rat Aorta TPH Primary Primary Lumen t t 1 so» Figure 9. 69 an anti-TPH monoclonal antibody (Sigma, St Louis, MO, USA) and a fluorescent anti-mouse secondary antibody, which confirmed the existence of TPH protein in arterial smooth muscle. Again, smooth muscle cells were confirmed by a-actin staining (data not shown). No fluorescence or very low fluorescence was pictured in cells without primary antibody incubation (data not shown). We also did IHC experiments testing the expression of TPH in artery sections using the same anti-TPH antibody. However, we did not get very convincing results. Relatively darker DAB precipitates appeared in sections with primary antibody incubation (Figure 9, bottom left) compared to control sections (Figure 9, bottom right). 2. The Activity of TPH in Peripheral Arteries To test whether the arterial TPH is functional, we measured 5-HTP levels in peripheral arteries and in hypothalamus from NSD 1015 (100 mglKg, 30 min)- treated rats. NSD 1015 is an inhibitor of AADC and blocks the conversion of 5- HTP to 5-HT. Hypothalamic tissues, that contains terminals of central serotonergic neuron terminals, were used as positive control for this experiment. Using HPLC we found non-detectable 5-HTP in aorta, fairly low amount of 5-HTP in superior mesenteric artery (003510.021 ng/mg protein) and 1612026 ng/mg protein of 5-HTP in hypothalamus. In all three tissues from tryptophan (300 mglKg, 60 min) and NSD 1015-treated rats, we observed increases of 5-HTP concentrations (Figure 10). We also tested the possibility of peripheral arteries 70 Figure 10. Quantification of 5-HIAA, 5—HT and 5-HTP in aorta (top), superior mesenteric artery (middle) and hypothalamus (bottom) in rats treated (i.p.) with AADC inhibitor NSD 1015 (100 mglKg, 30 min) or NSD 1015 (100 mglKg, 30 min) + tryptophan (300 mg/Kg, 60 min). Bars represent means :I: SEM for the number of animals in parentheses. * p<0.05 compared to NSD1015 (alone)- treated animal. 5-HlAA=5-hydroxyindoleacetic acid, 5-HT=5-hydroxytryptamine, 5-HTP=5- hydroxytryptophan, AADC=amino acid decarboxylase, SMA=superior mesenteric artery. 71 A—‘l N» d g protein) 9 .° .-‘ I? 09 9 OD we Rat Aorta [:lNSD 1015 .NSD 1015+ tryptophan (N=4) Concentration(nglm 9 9,591!- Concentration(nglm protein) 9 .° .° .° '9 «P 9’ 9° ogtfli [Ii 5-HIAA 5-HTP Rat SMA DNSD 1015 .NSD 1015 +tryptophan (N=3-4) i? 0| .5 w l N Concentration(nglmg protein) 5i 5-HIAA 5-HTP 5-I-IT Rat Hypothalamus DNSD 1015 .NSD 1015+tryptophan (N=3) 5-I-IT 5-HIAA 5-I-ITP _ figure 10. 72 taking up 5-HTP. The concentration of 5-HTP increased in rat aorta and superior mesenteric artery after incubation with exogenous 5-HTP (1 (M) for 30 minutes (Figure 11). We also tested TPH activity in in vitro experiments. In the presence of AADC inhibitor, substrate and all the co-factors that TPH needs to synthesize 5- HT, no 5-HTP accumulation in arteries (with or without tryptophan Figure 12, A, B), a low level of 5-HTP (without tryptophan) and an increased 5-HTP peak (with tryptophan) in our positive control -- raphe nucleus (Figure 12, C, D) were observed. 3. The Presence of AADC in Rat Aorta and Superior Mesenteric Artery Figure 13 shows the Western analysis of the expression of AADC protein (Abcam, Cambridge, MA) in rat aorta and superior mesenteric artery. We used rat adrenal medulla as a positive control. The bands in rat aorta and superior mesenteric artery samples as well as adrenal medulla migrated at ~55kDa. 4. The Presence of MAO A in Rat Aorta and Superior Mesenteric Artery Western analysis using an antibody specifically recognizing MAO A protein (H-70, Santa Cruz, CA, USA) was performed in rat aorta and-superior mesenteric artery whole tissue homogenate supernatant. This MAO A antibody is a rabbit polyclonal antibody raised against amino acids 458-527 of MAO A of human origin. Figure 14, top shows the bands in both rat aorta and superior mesenteric artery samples migrated at ~ 70 kDa, consistent with our positive 73 Figure 11. Quantification of 5-HIAA, 5-HT and 5-HTP in aorta (top) and superior mesenteric artery (bottom) from MAO A inhibitor pargyline (100 mg/Kg)-treated rats after 20 minutes of vehicle or 5-HTP (1 uM) incubation. Bars represent means 1 SEM for the number of animals in parentheses. * p<0.05 compared to vehicle incubation. 5-HIAA=5-hydroxyindoleacetic acid, 5-HT=5-hydroxytryptamine, 5-HTP=5- hydroxytryptophan, MAO A=monoamine oxidase A, SMA=superior mesenteric artery. 74 A {‘3 SR Concentration(nglmg protein) .°.°.°.°.°."‘.'"‘ ‘P “1.67 In Concentration(nglmg prote O O O O A A A '9 i‘ “B °.° ‘-? '9 1‘ Rat Aorta DVehicle .5-HTP 1 ”M 20 min (N=4) l 5-HIAA 5-HT Rat SMA |:|Vehicle .5-HTP 1 11M 20 min (N=4) l 5-HIAA 5-i-IT 5-l-ITP Figure 11. 75 Figure 12. Chromatogram showing the measurement of 5-HTP in rat aorta (A: without tryptophan; B: with tryptophan) and raphe nucleus (C: without tryptophan; D: with tryptophan) samples incubated with NSD1015 and other cofactors for 5-HT synthesis. Solid line: sample tracing. Dot line: 3 mix standards including 20 pL, 75pg/uL 5-HT, 5-HTP and 5-HIAA. 5-HlAA=5-hydroxyindoleacetic acid, 5-HT=5-hydroxytryptamine, 5-HTP=5- hydroxytryptophan, SMA=superior mesenteric artery. 76 AEEV mEE. ad a.” a." QN w IHI a e... I t . in I . .2 'r** ’ n I i l . I : l . V v A q ' ‘ : ~.a '4 .LE'I 9 :1 -E . . . . .E . . . . ..‘.‘:I| . .. ‘ . 38 E XE. ms. w IJ 93mm . u n . .3... A . W .8... . -——'O‘- 1? 9 ......o/W . Tl «.... Acmcaofifuv .3... 3:5 05:. a.» QN ad 3.: lH-g'ilf. WIH-S dJ'H'g“"'-'-'-'-'-Li's-22212211....i .. .- Emfioaecv .- .1II mto< AEEV 9:: a.» ...... en 9% 6.” no“ a." «.5 WIH-g av .38 ms 5:. m2. . anew-B . 93mm .1 ~“-UIAU . “A'I.v Annnn . nIv .86 1... 86% . 2.9M; w ( w 3.6 AEEV mEE. m." 6.” ad QN m... ...... a 2.6 .0 1 9.9 s. I ....- i 7 ' ;.....;.......-,--.... “nu-u“.-. ‘ . ' cv’lj-‘l-J'I79".'-'—'-~.-.:222:21::‘.‘.'.'.‘.'.’.'.‘,.‘.‘ .. $.52me mto< - 8.0 .I? °°I° v . goo . 3.6 A . 0 8.0 9 . 8... 1A) v lllll‘ .l .. ~“-‘I.U 3.6 2.“: '9ure 12. F 77 AADC Protein Expression in Arteries Rat Adrenal Rat SMA Rat Aorta Medulla Figure 13. Western blot of AADC in homogenates from rat superior mesenteric artery and aorta, where each lane represents a different rat. Adrenal medulla was used as positive control. AADC=amino acid decarboxylase. SMA=superior mesenteric artery. 78 Figure 14. A: Western blot of MAO A in homogenates from rat superior mesenteric artery and aorta, where each lane represents a different rat. Rat gut mucosa was used as positive control. B: Immunohistochemical staining of the MAO A (Santa Cruz Biotechnology) in smooth muscle between cables of elastin/collagen in the rat aorta and superior mesenteric artery paraffin embeded section. Parallel sections were incubated with antibody (left) or no primary antibody (right). Arrows indicate the placement of staining. Representatives of four separate experiments, each with a different rat. MAO A = monoamine oxidase A. SMA=superior mesenteric artery. 79 MAO A Protein Expression in Arteries A. F...—-- ...- —----‘ Rat Gut Rat SMA Rat Aorta Mucosa B- MAO A Primary Rat Aona Rat SMA Figure 14. 80 control, rat gut mucosa. Immunohistochemical experiments using the same antibody localized the MAO A protein to smooth muscle and the endothelial layer of the rat aorta and superior mesenteric artery (Figure 14 B, bottom). The pictures on the right show rat aorta and superior mesenteric artery sections incubated with secondary antibody, but no primary antibody. 5. The Activity of MAO A in Rat and Superior Mesenteric Artery To investigate whether MAO A is functional in peripheral arteries, we compared 5-HT and 5-HIAA concentrations in peripheral arteries with in vitro incubation with either exogenous 5-HT (1 1M, 15 min) or vehicle. We measured a basal level of 5-HT and 5-HIAA in rat aorta (5-HT, 0.28:0.05 nglmg protein; 5- HIAA, 1.38:0.09 nglmg protein, Figure 15 top) and in superior mesenteric artery (5-HT, 0.32:0.16 nglmg protein; 5-HIAA, 1.210.08 ng/mg protein, Figure 15 bottom). After incubation with 5-HT (1 pM, 15 min), 5-HIAA concentrations were increased to 6782048 nglmg protein and 7921.2 nglmg protein in rat aorta and superior mesenteric artery respectively with minor change of 5-HT levels (rat aorta, 0.43:0.07 nglmg protein; superior mesenteric artery, 1.210.18 nglmg protein). Moreover, we confirmed that the metabolism of 5-HT in arteries was mediated via MAO A by doing the same experiment in arteries from pargyline- treated rats. We observed that 5-HIAA production was abolished and 5-HT levels were increased in arteries from pargyline-treated rats (rat aorta, 5-HT= 0.92:0.07 nglmg protein, 5-HIAA= 00025100025 nglmg protein; superior 81 Figure 15. Quantification of 5-HIAA and 5-HT in aorta (top) and superior mesenteric artery (bottom) incubated with vehicle or 1 uM exogenous 5-HT for 15 minutes. Arteries were from normal rats. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle-incubated tissues. 5-HIAA = 5-hydroxyindoleacetic acid. 5-HT = 5-hydroxytryptamine. SMA= superior mesenteric artery. 82 . ‘9 N“ 0" Concentration (nglmg protein) 1‘ 't’ ‘1’ ‘r ‘I' O Rat Aorta I:IVehicle .5-HT1 uM, 15min (N=5) r—ni Concentration (nglmg protein) <5 -.* N ‘1’ E 9' 1’ 1' 0." '9 5-I-IT Rat SMA * EJVehicle .5-HT 111M 15 min (N=4) III a: 5-H'IAA 5-HT Figure 15. 83 mesenteric artery, 5-HT= 22810.24 nglmg protein, 5-HIAA= 002510.008 nglmg protein, Figure 16). VVItI'I exogenous 5-HT incubation, the arterial 5-HT levels significantly increased with minor increase of 5-HIAA (rat aorta, 5-HT= 24910.37 nglmg protein, 5-HIAA= 001310.013 ng/mg protein; superior mesenteric artery, 5-HT= 4.021054 nglmg protein, 5-HIAA= 01310.05 nglmg protein, Figure 16). III. The Lack of 5-HT Storage in Peripheral Artery 1. Investigation of Whether Rat Aorta Has Ability to Store 5-HT Basal endogenous levels of 5-HT in rat aorta were reported as above. Here we tested whether a peripheral artery has the ability to store more than basal level of 5-HT. To test this idea, we incubated untreated-aorta and 5-HT- loaded aorta from normal and pargyline-treated rats in PSS and compared 5- HIAA and 5-HT content in rat aorta and PSS after 0, 4 or 8 hours. The 5-HIAA and 5-HT we measured in PSS were released during 4 or 8 hours incubation from rat aorta. The amount of 5-HIAA and 5-HT remaining in rat aorta after incubation was also measured. First we tested whether aorta would maintain endogenous 5-HT. We observed that in aorta from normal rats, the 5-HT content was maintained in aorta after 4 or 8 hours of PSS incubation (Figure 17 bottom, Table), while 5- HIAA was not and was released to PSS (Figure 17 top, Table 2). We next tested whether aorta could store more than basal level of 5-HT by comparing 5-HIAA and 5-HT levels in aorta after 4 or 8 hours PSS incubation in exogenous 5-HT (1 11M, 15 min) preloaded-aorta with those at time 0 and right after exogenous 5-HT 84 Figure 16. Quantification of 5-HIAA and 5-HT in aorta (top) and superior mesenteric artery (bottom) incubated with vehicle or 1 11M exogenous 5-HT for 15 min. Arteries were from rats treated with MAO A inhibitor pargyline (100 mg/Kg, i.p., 30 min). Bars represent means 1 SEM for the number of animals in parentheses. * p<0.05 compared to vehicle-incubated tissues- 5-HIAA = 5—hydroxyindoleacetic acid. 5-HT = 5-hydroxytryptamine. SMA= superior mesenteric artery. MAO A = monoamine oxidase. 85 Aorta From Pargyline-treated Rats "ESP DVehicle .5-HT1 11M 15 min (N=4) N 1 1"}: Concentration (nglmg protein) ‘1’ 1‘ ‘1" ‘P _ |'_'l 5-HIAA 5-i-IT O SMA From Pargyline-treated Rat ’E 35’ 91 EIVehicIe 33' .5-HT1 11M 15 min 237‘ (N=4) 36' 55- * .5 4' E 3- E, 2- E 1. <3 o- ——-'-_ . 5-HIAA 5-HT Figure 16. 86 Figure 17. Top: Quantification of 5-HIAA and 5-HT released into PSS during 0, 4 or 8 hours incubafion. Bottom: Quantification of 5-HIAA and 5-HT remaining in tissues after 0, 4 or 8 hours incubation in PSS. Time 0 represents no incubation (basal endogenous 5-HlAA and 5-HT levels). Bars represent means i SEM for the number of animals in parentheses. * p<0.05 compared to basal level (no incubation, time 0). 5-HlAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. PSS=physiological salt solution. Table 2. Quantification of 5-HlAA and 5-HT released into PSS and remained in tissue during 0, 4 or 8 hours incubation. Values reported as nglmg protein. * p<0.05 compared to basal level (no incubation, time 0). 87 PSS €1.4- Clo E 1.2- .4 hours a .8 hours E 1.0- (N=6-8) 2’ 0.8- ‘E .2 0.6- E ‘E' 0.4- a: 2 0.2- o 0 o.c 5 HT ...... Rat Aorta .s 0-5' :10 43 .4 hours 3. o_4. ”49:2." 8 hours as» (N=6-8) g: o-3 _T_ l ; ‘E , :2 0.2 / e * a , % 3 0.1 / 5 Z % 0 o.c . .5 5-HIAA 5-HT Table 2. Figure 17. 0 4 Hours 8 Hours PSS 5-HIAA 0 1.02:0.23* 1 .018:0.19* 5-HT 0 0.097:0.05* 0.05:0.006* Rat 5-HIAA 0.27:0.05 04210.03" 0.085=0.01* Aorta 5-HT 0.28:0.07 0.26:0.1 0.29:0.065 88 incubation. We rinsed and flushed aorta with fresh drug-free PSS to eliminate exogenous 5-HT attached on the aorta wall. As shown in Figure 18, bottom and Table 3, aortic 5-HIAA level was greatly increased by 15 minutes incubation of 1 uM exogenous 5-HT with a moderate increase of 5-HT in aorta. After 4 or 8 hours incubation with PSS, the majority of 5-HIAA was released to PSS with a slight though significant decrease of 5-HT in aorta with time (Figure 18, top and Table 3). We then took a further step to investigate whether aorta would store 5- HT when 5-HT metabolism was inhibited. We did a similar experiment in aorta from MAO A inhibitor, pargyline (100 mglKg, 30 min)-treated rats. After incubation with exogenous 5-HT (1 (M, 15 min), the aortic 5-HT content increased with no change of 5-HIAA (Figure 19, bottom, Table 4). However, this is an acute accumulation of 5-HT, after 4 or 8 hours, 5-HT concentration decreased by releasing both 5-HT and 5—HlAA from aorta to PSS (Figure 19, top, Table 4). 2. Investigation of “Serotonylated” 5-HT in Rat Aorta We tested whether 5-HT covalently bound to protein exists in peripheral arteries by Western analysis. We separated rat aorta protein on a 12% gel by molecular weight and blotted with a 5-HT antibody. Multiple bands were visualized (Figure 20), suggesting the presence of serotonylated proteins in rat aona. 89 Figure 18. Top: Quantification of 5-HIAA and 5-HT released into PSS during 4 or 8 hours incubation from 5-HT preloaded rat aorta (5-HT 1 (M, 15 min). Bottom: Quantification of 5-HIAA and 5-HT in naive aorta (basal level) and in 5- HT preloaded aorta after 0, 4 or 8 hours incubation in PSS. Time 0 represents no incubation (basal endogenous 5-HIAA and 5-HT levels). Bars represent means :l: SEM for the number of animals in parentheses. * p<0.05 compared to basal level. # p<0.05 compared to 5-HT 1 uM incubation. 5-HlAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. PSS= physiological salt solution. Table 3. Quantification of 5-HlAA and 5-HT in naive aorta and 5-HIAA and 5-HT released to PSS or remained in 5-HT preloaded aorta after 0, 4 or 8 hours PSS incubation. Values are reported as nglmg protein. * p<0.05 compared to basal level. # p<0.05 compared to 5-HT 1 uM incubation. 90 PSS 3515.0- * * :IBasal % , -5-HT1 uM 15 min 512'5' // -4 Hours 3100. .8 Hours 3 (N=6) c 7.5 .2 *5 5.0. E 8 2.5- C O U 0.0 5-HIAA 5-HT :5 Rat Aorta L__lo .5-HT1uM 15 min .4 hours -8 hours (N=6) r—1 - “it _# 5-HT Figure 18. 4 Hours 8 Hours 13.08:1.72* 12.93:1.71* 5-HT 0 0 O 0 Rat 5-HIAA 1.38:0.09 6.78:0.48* 20210.03" 0.991045" Aorta 5-HT 0.28:0.05 0.43:0.07 0.16:0.03” 0.14:0.01" 91 Figure 19. Top: Quantification of 5-HIAA and 5-HT released into PSS during 4 or 8 hours incubation from 5-HT preloaded (5-HT 1 (M, 15 min) aorta from pargyline-treated rat. Bottom: Quantification of 5-HlAA and 5-HT in aorta (basal level) and in 5-HT preloaded aorta from pargyline-treated rats after 0, 4 or 8 hours incubation in PSS. Time 0 represents no incubation (basal endogenous 5-HIAA and 5-HT levels). Bars represent means 1 SEM for the number of animals in parentheses. * p<0.05 compared to basal level. # p<0.05 compared to 5-HT 1 uM incubation. 5-HlAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. PSS: physiological salt solution. Points represent means i SEM. Table 4. Quantification of 5-HIAA and 5-HT in aorta and 5-HlAA and 5-HT released to P88 or remained in 5-HT preloaded aorta from pargyline-treated rats after 0, 4 or 8 hours PSS incubation. Values are reported as nglmg protein. * p<0.05 compared to basal level. # p<0.05 compared to 5-HT 1 uM incubation. 92 PSS ’E '53 0'77 ... L—JBasal g o 6- .5-HT 1 [1M 15 min or 0 5. -4 Hours % ' -8 Hours 5 0.4- 01:4) .5 0.31 . ‘E'é * b 0.2- * 8 c 0.1- O 0 0.0 5-HIAA 5-HT ? Rat Aorta '6 3.0! Do £25 .5-HT1uM15 min or ' -4 hours ’///. 8 h g 2'0! - OUI‘S g (N-4) : 1.5- .2 g 1.0- C 8 0.5- C O U o_c m 5-HIAA Table 4. Figure 19. 0 5414;- 1.“M 4 Hours 8 Hours mIn PSS 5-HIAA o o 0.15:0.03* 0.46:0.16* 5-HT 0 o 0.19:0.03* o.22=o.os* Rat 5-HIAA o 0.01:0.01 0.08:0.01 0.09:0.02 Acme 5-HT 0.92:0.07 2.49:0.37* 1.08:0.09" 0.47101" 93 No primary Figure 20. Western blot of 5-HT in homogenates from rat aorta. Representatives of three separate experiments, each with a different rat. Left: Blot incubated with secondary antibody with no exposure to primary antibody. Right: Blot incubated with rabbit anti-5-HT antibody (Serotec, Raleigh, NC). 94 IV. The Presence of Uptake and Release of 5-HT in Peripheral Arteries 1. The Presence of SERT in Peripheral Arteries The presence of SERT mRNA was demonstrated by real time RT-PCR. Figure 21 top shows that one PCR product, obtained using primers developed through Primer Express® software for the SERT encoding sequence, was identified in cDNA made from rat aorta. No product observed in the lanes with no reverse transcriptase suggesting no genomic contamination in our cDNA samples. In Western analyses, an antibody directed toward the carboxyterminus of the human SERT recognized one protein band of 74 kDa mass in homogenates of rat aorta, mesenteric resistance arteries, mouse aorta and mouse aorta sample immunoprecipitated with the same antibody (Figure 21, bottom). IHC experiments were performed in rat aorta paraffin embedded and mouse aorta frozen sections using an antibody that recognized the C-terminus (C-20) of the SERT. The SERT was expressed in the smooth muscle and in the endothelial cell layer and the black DAB precipitate did not appear in sections incubated with antibody plus competing peptide (Figure 22, compare top and middle panel). The adventitia of the vessels of rat aorta stained darkly, but this was not competed off by competing peptide; thus this staining is likely nonspecific. The bottom panel showed aorta sections incubated with Secondary antibody but no primary antibody. The presence of SERT in peripheral artery was further validated by immunocytochemistry. We double-stained freshly isolated rat aortic smooth muscle cell using SERT antibody (C-20) and a specific cell plasma membrane 95 Figure 21. Top: Final product of real time RT-PCR for detection of SERT and GAPDH mRNA in rat aorta. 1-4 are separate rats. RT=reverse transcriptase. Bottom: Western blot of SERT in homogenates from rat aorta, rat superior mesenteric arteries, mouse aorta homogenates and immunoprecipitated protein (by SERT C-20 antibody, Santa Cruz Biothechnology) from a mouse aorta. Representatives of at least four separate experiments, each with a different rat/mouse. SERT= serotonin transporter. SMA= superior mesenteric artery. 96 SERT mRNA Expression in Rat Aorta SERT 1 2 3 4 GAPDH 1 2 3 4 SERT Protein Expression in Arteries Rat Aorta Rat SMA 74”) mm~--- Mouse Aorta 74'kE) Ill-ll III-II Homog. IP Figure 21. 97 Figure 22. Immunohistochemical staining of the SERT (Santa Cruz Biotechnology, C-20) in smooth muscle between cables of elastin/collage in the rat aorta (left, paraffin embedded sections) or mouse aorta (right, frozen sections). Parallel sections were incubated with antibody alone (top), quenched with a 5X excess competing peptide (middle) or no primary (bottom). Arrows indicate the placement of staining. Representative of 4-6 different experiments (rats/mice). SERT: serotonin transporter. 98 SERT Protein Expression in Arteries Rat Aorta Mouse Aorta Figure 22. 99 marker pan-cadherin antibody. By using confocal microscopy, we scanned a single layer of smooth muscle cell. Overlaying the staining of the SERT antibody and the pan-cadherin antibody, we observed that most of the SERT was localized on plasma membrane (Figure 23). 2. The Characterization of Active 5-HT Uptake in Peripheral Arteries A. 5-HT Uptake Time Course Study Different tissues (conduit or small vessels) from normal or pargyline- treated rats were used in 5-HT uptake time course studies. Superior mesenteric arteries from normal rats were incubated in normal PSS with 5-HT (1 nM) for 0, 15, 30 or 45 minutes in room temperature (25 °C). Figure 24 demonstrates that arterial 5-HIAA content increased in a time- dependent manner with a concomitant minor increase of 5-HT. We also tested 5-HT uptake in a time course study using arteries from pargyline-treated rats. The merits of using pargyline-treated rats are that we can study 5-HT uptake directly and reduce changes in 5-HlAA. Rat aortae were incubated in PSS with 1 uM pargyline and 1 uM 5-HT for 0, 15,30, 45, 60 and 90 minutes at room temperature (25 °C) and 37 °C (Figure 25). At room temperature, aortae took up 5-HT in a time dependent manner and plateaued at 60 minutes with minimal changes of 5-HlAA. The maximal level of 5-HT in aorta was 8.48:0.71 nglmg protein. However, at 37 °C, uptake plateaued after 30 minutes of exogenous 5-HT (1 uM) incubation with maximal aortic 5-HT level at 5.25:0.27 nglmg protein. After 15 minutes 5-HT (1 uM) incubation, aortic 5-HT 100 Figure 23. Confocal image of freshly dissociated rat aortic smooth muscle cells double stained with pan-cadherin antibody (Sigma) and SERT antibody (C-20, Santa Cruz Biotechnology). Representative of cells from 6 different experiments (rats). SERT= serotonin transporter. 101 SERT Protein Expression in Freshly Isolated Rat Aortic Smooth Muscle Cells Pan-cadherin O Overlay Figure 23. 102 Rat SMA |:IVehicle .5-HT1pM 15min MS-HT 111M 30min 24:2: 5-HT 1p.M 45min (N=5-9) .3 .° * Concentration (nglmg protein) N P 5-HIAA 5-HT Figure 24. Time course for 5-HT uptake in the rat superior mesenteric artery incubated with 5-HT (1 HM) in room temperature. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared with time 0 (no incubation). 5-HlAA=5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SMA=superior mesenteric artery. 103 Figure 25. Time course for 5-HT uptake in the aorta from pargyline-treated rats incubated with exogenous 5-HT (1 uM) at 37 °C (Top) or at room temperature (bottom). Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared with time 0 (no incubation). 5-HIAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. 104 A h c» on O I I I I Concentration(nglmg protein) N Rat Aorta (37 °C) DVehicle .5-HT 1uM 15min m5-HT 115M 30min 33:2 5-HT 1uM 45min .5-HT 1.1M 60min . .. .5-HT 1uM 90min * * (N=4) O I A .h m on O J I I I Concentration(nglmg protein) N 5-HIAA 5-HT Rat Aorta (Room Temperature) EZIVehicle * .5-HT 1p.M 15min O I @5-HT 1uM 30min ,z, .5-HT 111M 45min * .5-HT 1uM 60min :3; .5-HT 1uM 90min (N=6-10) .1; £55 5-HIAA 5-HT Figure 25. 105 levels are similar at room temperature and 37 °C incubation groups (room temperate, 3.07:0.35 nglmg protein; 37 °C, 3.12:0.45 nglmg protein). Similarly, superior mesenteric artery from pargyline-treated rats time-dependently take up 5-HT, which did not increase further after 60 minutes of 5-HT incubation in room temperature (Figure 26). All of our following 5-HT uptake experiments incubated tissues with 5-HT (1 (M) for 15 minutes at room temperature. The real uptake of exogenous 5-HT into arteries was confirmed by using tph1-/- mice. The tph1-/- mice expressed normal amounts of 5-HT in brain but lack of 5-HT in periphery (Walther et al., 2003). The blank background of 5-HT in these mice makes them a good model for studying 5-HT uptake in peripheral arteries. Figure 27, table 5 compares the uptake of exogenous 5-HT in aorta from untreated wild type and tph1-/- mice. It is clear that basal 5-HT and 5-HIAA concentrations were significantly lower in aorta from tph1-/- mice. Upon incubation with exogenous 5-HT, the level of 5-HIAA increase in mouse aorta is much higher than the 5-HT increase in both wild type and tph1-/- mice, which is similar to what we observed in rat arteries. Thus, we concluded that 5-HT can be taken up by various peripheral arteries and 5-HT metabolized to 5-HIAA quickly after it gets into arteries. B. Investigation of SERT-dependent 5-HT Uptake We next investigated whether the active 5-HT uptake in peripheral arteries was mediated by SERT. We preincubated arteries (rat aorta and superior 106 Rat SMA (Room Temperature) [ZIVehicle V4 5-HT1uM 15min "T’T’T 5-HT 1p.M 30min A C I N I Concentration (nglmg protein) 7//_ 5-HT 1.1M 45min y 5.. 525252 5-HT 1.1M 60min é .5-HT 1.1M 90min 5% g 4- (N=4-12) : é % Z Z. 8 5+th 5-HT Figure 26. Time course for 5-HT uptake in the superior mesenteric artery from pargyline- treated rats incubated with 5-HT (1 (M) at room temperature. Bars represent means :i: SEM for the number of animals in parentheses. * p<0.05 compared to time 0 (no incubation). 5-HIAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SMA= superior mesenteric artery. 107 Figure 27. 5-HT uptake in the aorta from wild type (top) and tph1-l- mice (bottom) incubated with exogenous 5-HT (1 11M, 15 min) at room temperature. Bars represent means 1 SEM for the number of animals in parentheses. * p<0.05 compared to vehicle. 5-HIAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. TPH=tryptophan hydroxylase. Table 5. Quantification of 5-HIAA and 5-HT in aorta from wild type or tph1-l- mice incubated with vehicle or exogenous 5-HT (1 11M, 15 min) at room temperature. Values are reported as nglmg protein. Bars represent means a: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle. 108 Wild*Type Mouse Aorta €2.01 - ,9, DVehIcle g .5-HT 11M 15 min cents- (N=3-4) B: E. .. c 1.0" .2 .8 c 0.5- fl [1] o o c o o 0.0 , 5-HIAA 5-HT ’5 2 0 TPH1 -I- Mouse Aorta .g ' * EIVehicle a .5-HT 1 uM 15 min at 1 .5- (N=3-4) Bi 5 c 1 .0" .2 * E E 0.5. 1:1 1 o o 5 o 0.0- 5-HIAA 5-HT Table 5. Figure 27. Wild Type Mouse Aorta TPH 1-/- Mouse Aorta Vehicle 5-HT 111M 15min Vehicle 5-HT 111M 15min 5-HIAA 0.37:0.17 1.74:0.25* 0.0231002 1.5:0.40* 5-HT 0.49:0.10 0.85:0.19* 0.0831005 0.62:0.11" 109 mesenteric artery) with either vehicle or one of the SERT inhibitors fluvoxamine (1 (M) or fluoxetine (1 (M) for 30 minutes prior to exposure to 5-HT (1 (M) for 15 minutes. The 5-HT levels in arteries incubated with 5-HT and SERT inhibitors were significantly lower than in arteries only incubated with exogenous 5-HT suggesting a SERT dependent 5-HT uptake in arteries. Neither fluoxetine (1 uM) nor fluvoxamine (1 uM) inhibited the entire uptake of 5-HT (Figure 28). SERT-targeted mutation mice (SERT KO) were used in our study to test the dependence of SERT in 5-HT uptake in peripheral arteries. Basal level of 5- HlAA in aorta from pargyline-treated SERT KO mice were much lower compared with WT mice with a minor change of basal 5-HT level (Figure 29, vehicle incubation). Fluvoxamine inhibited 5-HT uptake in aorta from WT mice but not in SERT HET or SERT KO mice (Figure 29). Total 5-HT uptake reduced in aorta from SERT KO mice with no further inhibition of fluvoxamine pre-incubation. C. Investigation of SERT-independent 5-HT Uptake We tested whether there is a Na*-independent uptake because monoamine transporters (SERT, NET and DAT) are Na“-K*-Cl'—dependent transporters, which have the potential to take up 5-HT. The result showed that there was a 5-HT uptake in aorta and superior mesenteric artery from pargyline- treated rats in Na‘ free (Na+ was replaced isosmotically with N-methylglucamine) - PSS with 1 uM exogenous 5-HT (15 min), though the total uptake was significantly reduced compared to that in regular Na” containing PSS. SERT inhibitor, fluoxetine or fluvoxamine, did not further reduce 5-HT uptake in aorta 110 Figure 28. Effects of a 30-minute preincubation with SERT inhibitor fluvoxamine (1 11M) or fluoxetine (1 (M) on 5-HT uptake (1 uM 5-HT incubation for 15 min) in aorta (top) or superior mesenteric artery (bottom) from pargyline treated rats. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle. # p<0.05 compared to 5-HT (1 uM). 5-HlAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SERT= serotonin transporter. SMA=superior mesenteric artery. 111 Concentration(nglmg protein) Concentration (nglmg protein) 0) 5’5"}? ‘ L Rat Aorta DVehicle .Fluvoxamine 111M 'f’t’t‘i'm A L EFluoxetine 1p.M .5-HT 111M .5-HT+Fluvoxamine * E5-HT+Fluoxetine (N=3-10) *# 5-HIAA 5-HT Rat SMA ' |:|Vehicle * .Fluvoxamine 111M EFluoxetine 111M .5-HT 111M .5-HT+Fluvoxamine E5-HT+Fluoxetine (N=3-6) O I Figure 28. 112 Figure 29. Effects of a 30-minute preincubation with SERT inhibitor fluvoxamine (1 (M) on 5-HT uptake (1 uM 5-HT incubation for 15 min) in aorta from wild type (top), SERT HET (middle) and SERT KO mice (bottom). Bars represent means :i: SEM for the number of animals in parentheses. * p<0.05 compared with vehicle. 5-HIAA= 5-Hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SERT= serotonin transporter. 113 E" ? a? ‘1' Concentration (nglmg protein) O 15 Wild Type Mouse Aorta DVehicle .Fluvoxamine 1 uM .5-HT1 uM .5-HT-I-Fluvoxamine (N=3-4) P ‘P N .° .3 .5 ‘P ‘1' Concentration (nglmg protein) C 3r ......5 H5 5-HIAA 5-HT SERT HET Mouse Aorta CIVehicle .Fluvoxamine 1 uM .5-HT 1 uM .5-HT-i-Fiuvoxamine (N=3-4) * P ‘P N ? 1" '9 0 Concentration (nglmg protein) ir .° 1': 5-HT SERT KO Mouse Aorta DVehicle .Fluvoxamine 1 11M .5-HT 1 uM .5-HT+Fluvoxamine (N=3-4) Figure 29. 5-HiAA 114 Figure 30. Effects of a 30-minute preincubation with SERT inhibitor fluvoxamine (1 (M) or fluoxetine (1 pM) on 5-HT uptake (1 uM 5-HT incubation for 15 min) in Na“ -free PSS in aorta (top) or superior mesenteric artery (bottom) from pargyline treated rats. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle. # p<0.05 compared to 5-HT (1 uM) uptake in normal PSS (containing Na*). 5-HIAA= 5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SERT= serotonin transporter. SMA=superior mesenteric artery. 115 Concentration(nglmg protein) Concentration(nglmg protein) ‘3 w l |:|Vehicle Fluvoxamine 1 uM E Fluoxetine 111M .5-HT 1 uM 5-HT+Fluvoxamine g 5-HT+Fluoxetine .5-HT 111M with Na+ (N=6-10) Rat SMA 5- . I:IVehIcle 5_ .5-HT1uM .5-HT1 ”M with Na+ 4_ (N=4-6) .l 21 .II'_ 1- r—I 5-HIAA Rat Aorta 116 5-HT Ill|||llllllllllllllllllllllll Figure 30. incubated in Na+ free- PSS (Figure 30). We then tested whether this 5-HT uptake was SNS, NET and OCT- dependent in peripheral arteries. To investigate whether uptake of 5-HT by the artery was dependent on sympathetic nerves, we used 6-OHDA denervated rats. Rats were injected with 6-OHDA during an 8-day protocol, and arteries procured on day 8. Validation of denervation was verified by lack of glyoxylic acid-reacted norepinephrine (Figure 31, top). While basal levels of 5-HT and 5-HIAA were slightly but not significantly lower than in untreated animals, arteries were still capable of taking up and metabolizing 5-HT as the concentrations of 5-HT and 5- HIAA increased in the vessels exposed to 5-HT for 15 minutes. This was true for all 3 arteries examined (Figure 31). The NET inhibitor nisoxetine (100 nM) was unable to inhibit 5-HT uptake (Figure 32). The OCTs inhibitor, corticosterone (1 uM) did not inhibit 5-HT uptake in aorta but potentially could in superior mesenteric from pargyline-treated rats (Figure 33). Based on our observation, arterial 5-HT uptake is not dependent on SNS, NET or OCT. As the barrier between arterial smooth muscle and blood, endothelium could also take up 5-HT. Uptake of 5-HT, at least in the superior mesenteric artery appears to be largely independent of the endothelium as removal of this cell layer did not alter 5-HT uptake or metabolism (Figure 34). 117 Figure 31. A. Glyoxylic acid fluorescence in small mesenteric resistance arteries from vehicle or 6-OHDA-treated rats. The white network is sympathetic nerve fibers. B. 5-HT uptake in aorta, carotid artery and superior mesenteric artery from 6- OHDA treated rats. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared with vehicle. 5-HlAA=5-hydroxyindoleacetic acid. 5-HT= 5-hydroxytryptamine. SMA=superior mesenteric artery. 118 A' Mesenteric . - c0!" “ Artery Glyoxylic Acid Staining Vehicle " 6-OHDA, Day 8 E 3.5- * RatAorta % DVehicle a 3.0- .NS;HT1p.M a = g 2.5- ( l E 2.0- C ;§ 1.5- g 1.0- C § 0.54 i O U .c 5-HIAA 5-HT E 3.51 Rat SMA 33 DVehicle a 3-0‘ * .5-HT 1 ”M 2’ 2.5- (N=4) Bi 5 2 o- s g 1.5- * ‘6 i: 1.0- C 8 0 0.5 - ,_..., - 5-HIAA 5-HT 351 Rat Carotid Artery EIVehicle 3.01 .5-HT1uM Concentratioin (nglmg protein) 9 > 2.5- (N=4) 2.0- 1.5- 1.0- ' . 0.5- _ [:1 Figure 31. " 5-H'iAA 5-HT 1 19 Rat SMA .h J |:]Vehicle .Nisoxetine 100 nM .5-HT 1 (1M :1" , i 5-HT+Nisoxetine w I Concentration (nglmg protein) N Figure 32. Effects of a 30-minute preincubation with NET inhibitor nisoxetine (100 nM) on 5- HT uptake (1 uM 5-HT incubation for 15 min) in aorta from normal rats. Bars represent means 1: SEM for the number of animals in parentheses. * p<0.05 compared with vehicle incubation. 5-HlAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. NET=norepinepherin transporter. SMA=superior mesenteric artery. 120 Figure 33. Effects of a 30-minute preincubation with OCT inhibitor corticosterone (1 (M) on 5-HT uptake (1 uM 5-HT incubation for 15 min) in aorta (Top) and superior mesenteric artery (bottom) from pargyline-trated rats. Bars represent means 1 SEM for the number of animals in parentheses. * p<0.05 compared with vehicle incubation. #p<0.05 compared with 5- HT+fluvoxamine+corticosterone. 5-HlAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. OCT: organic cation transporter. SMA=superior mesenteric artery. 121 Concentration(nglmg protein) in) —‘ N I .3 O m J h J Concentration(nglmg prote N Rat Aorta *# 7l___IVehicle *# 6 |]]]]]Ifl]Corticosterone 111M Fluvoxamine-I-Corticosteron : 5.5-HT 1 uM u 5-HT+Corticosterone 4.5-HT+Fluvoxamine+ N w I I Corticosterone (N=7-8) * ill 5-HIAA 5-HT Rat SMA |_—_|Vehicle *# ||]I]]I[|Corticosterone 1p.M F luvoxamine+Corticosterone 5-HT1 M l M 5-HT+ orticosterone .5-HT+Fluvoxamine+ Corticosterone (N=4) 5-HIAA I , 5- HT L _ 0"" II: o L Figure 33. 122 Figure 34. 5-HT uptake (1 uM 5-HT incubation for 15 min) in endothelium-intact or endothelium denuded superior mesenteric artery from normal rats. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle incubation. 5-HIAA= 5-hydroxyindole acetic acid. 5-HT= 5-hydroxytryptamine. SMA=superior mesenteric artery. 123 Concentration (nglmg protein) A Concentration (nglmg protein) .° N N —‘L P P N N P Rat SMA (with Endothelium) 5. DVehicle .5-HT 0‘ (N=5-10) .5- o. 5. G Rat SMA (without Endothelium) 5' * DVehicle .5-HT 0- (N=5-10) .5- .0- 5.. l l 0 . I 1 . 5-HIAA 5-HT Figure 34. 124 3. The Presence of 5-HT Release in Peripheral Arteries (+)-Fenfluramine and (+)-norfenfluramine are SERT substrates and 5-HT releasers. We used these two drugs to test the release of 5-HT from arteries. To enhance the signal of 5-HT, we used pargyline-treated animals to study 5—HT release from arteries. Figure 35 shows that (+)-fenfluramine concentration (1 uM or 10 11M, 20 min) dependently released endogenous 5-HT from rat aorta into PSS. Similar amounts of 5-HT were released from rat aorta by incubation with 10 uM (+)- fenfluramine (0.37:0.07 nglmg protein) and 10 11M (+)-norfenfluramine (0.35:0.18 nglmg protein) for 20 minutes. The remaining 5-HT in aorta after fluramines incubation were 0.56:0.12 nglmg protein and 0.74:0.22 nglmg protein after (+)-fenfluramine or (+)-norfenfluramine-incubation, respectively. Thus, the released 5-HT during fluramines incubation accounts for about 30% of total endogenous 5-HT in rat aorta (Figure 35). The release of endogenous 5-HT is not a vessel specific effect as we observed fenfluramine (10 11M, 20 min) released endogenous 5-HT in superior mesenteric artery from pargyline-treated rats (Figure 36). Fluoxetine (1 uM, 30 min), which potentiated basal 5-HT release, did not inhibit (+)-fenfluramine (1 (M or 10 11M, 30 min)—induced 5-HT release from rat aorta (Figure 37). 125 Figure 35. (+)-Fenfluramine (1 11M or 10 (M) or (+)-norfenfluramine (10 11M) induced 5-HT release from aorta obtained from pargyline-treated rats. Top: released 5-HIAA and 5-HT during fluramine incubation. Bottom: remained 5-HIAA and 5-HT after fluramine incubation. Bars represent means :i: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle incubation. 5-HlAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. PSS: physiological saline solution. 126 PSS F 0.6 . 1 :IVehicle . 0 -(+)-Fenfluramine1uM -- °- '57 -(+)-Fenfluramine 1011M §§§(+)-Norfenfluramine 10uM (N=3-8) i323 i’ihi Fl 5-HIAA 5-HT Rat Aorta 2.0 - [:IVehicle .(+)-Fenfluramine 1 uM . 1%: (+)-Fenfluramine _T_ 10uM , . .(+)-Norfenfluramine ' 10pM (N=3-8) .1. 0| Concentration (nglmg protein) 0 0.5—MM 5-HIAA 127 Figure 36. (+)-Fenfluramine (1O uM)-induced 5-HT release in superior mesenteric artery from pargyline-treated rats. Top: released 5-HlAA/5-HT during (+)-fenfluramine incubation. Bottom: remaining 5-HlAA/5-HT after (+)-fenfluramine incubation. Bars represent means :1: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle incubation. 5-HIAA= 5-hydroxyindole acetic acid. 5-HT= 5-hydroxytryptamine. PSS: physiological saline solution. SMA=superior mesenteric artery. 128 Concentration (nglmg protein) .c .O h OI l J I 00 I .3 .3 N 0 0| O l l J Concentration (nglmg protein) C in P o PSS DVehicle * .Fenfluramine 10 uM 20 min (N=5-7) 5-HIAA 5-HT Rat SMA [:lVehicle .Fenfluramine 10uM 20 min (N=5-7) l r— , - , 5-HIAA 5-HT Figure 36. 129 Figure 37. Effects of 30 minutes SERT inhibitor fluoxetine (1 11M) preincubation on (+)- fenfluramine (10 (M) —induced 5-HT release in aorta from pargyline-treated rats. Top: released 5-HlAA and 5-HT during fluramine incubation. Bottom: remaining 5-HIAA and 5-HT after (+)-fenf|uramine incubation. Bars represent means :i: SEM for the number of animals in parentheses. * p<0.05 compared to vehicle incubation. 5-HIAA=5-hydroxyindoleacetic acid. 5-HT=5-hydroxytryptamine. PSS=physiological saline solution. SERT=serotonin transporter. 130 PSS DVehicle * .Fluoxetine1 uM .Fenfluramine1 uM A 2" Concentration(nglmg protein) 1,0. MFenfluramine1 0 uM f/ .Fenfluramine1 uM Z +Fluoxetine % . 2m Fenfluramine 1 0 uM % 0,5. +Fluoxetine % . (N=3-8) . é ' 2 w, / 0.0- we. .. A 5-HIAA 5-HT A Rat Aorta é, 2-5 ~- Vehicle 25 ' Fluoxetine 1 pM ozo Fenfluramine 1 (M 2’ 5:32 Fenfluramine 10 11M 3,15 l . Fenfluramine 111M g. +Fluoxetine , 3 1 0 .88 Fenfluramine 10 M r ‘5 ' +Fluoxetine : *5 (N=3-8) i 3 0.5- E = I 8 : 0.0- - 5-HIAA -HT Figure 37. 131 Hypothesis #2: Changes in the localized serotonergic system contribute to the increase of blood pressure in hypertension. We focused on comparing (1) SERT expression; (2) basal 5-HIAA and 5- HT levels; (3) peripheral arteries 5-HT uptake ability; and (4) function of SERT in modifying 5-HT—induced aortic contraction in normotensive and hypertensive rats. i. Measurement of SERT Expression in Aorta from Normotensive and Hypertensive rate We used two experimental hypertensive rats model (DOCA-salt hypertensive rats and LNNA-hypertensive rats) and one genetic hypertensive model (SHR). The blood pressure of the animals we used in our study were SBP [mmHg], DOCA= 197:6, SHAM(D)= 112:4, LNNA= 228:9, SHAM(L)= 128:2, SHR= 172:7 and WKY= 121:3. Western blotting analysis using a SERT antibody, which recognizes amino acids (516-630) mapping at the carboxy terminus of the SERT protein (H115 antibody, Santa Cruz, CA) showed a single band slightly lower than 75 kDa (Figure 38). The expression of SERT in aorta from DOCA-salt rats was significantly elevated compared to SHAM rats (SHAM(D), which paired with DOCA-salt hypertensive rats) (arbitrary unit, normalized to a-actin expression, SHAM(D)= 2774:469, DOCA= 4374:125, p<0.05). This upregulation of SERT protein expression was not caused by increased transcription but increased translation of mRNA, as we observed no 132 Figure 38. Western blotting analysis of SERT in aorta homogenates from SHAME», DOCA- salt hypertensive rats, SHAM“, LNNA hypertensive rats, WKY and SHR. Representative of three experiments/rats. SERT= serotonin transporter. SHAM“), = SHAM rats paired with DOCA-salt hypertensive rats. SHAM(L,= SHAM rats paired with LNNA hypertensive rats. 133 mzm >x>> <22._ 23% <08 éz 1" i: O x o '1‘ : (N=4-9) Percentage PE (105 M) Contraction 3 ? Put—i -1o -9 -8 7 -5 -5 $1 33. Log 5-HT [M] Rat Aorta 200' +Sham Vehicle -o- Sham Fluvoxamine +DOCA Vehicle 150- -v- DOCA Fluvoxamine (N=4) IOI'I Contract .8 O ‘P at ? Percentage PE (105 M) O L -1o -9 5. Log 5-HT [M] Figure 43. 148 °°“"°' SHAM DOCA-salt Threshold 6.92:0.07 8.00:0.16* £050 5.90:0.08 6.13:0.09* Fluoxetine SHAM DOCA-salt Threshold 7.06:0.25 8.83:0.16* £050 5.90:0.10 6.44:0.08* Fluvoxamine SHAM DOCA-salt Threshold 7.50:0.20" 8.50:0.28* £050 6.43:0.21” 6.85:0.08* Table 6. Quantification of the threshold and EC50 in 5-HT-induced contraction in aorta from normotensive and hypertensive rats. Values reported as the negative logarithm of the value [M]. *indicates statistically significant differences (p<0.05) when compared to SHAM. # indicates statistically significant differences (p<0.05) when compared to vehicle. ECso = concentration of 5-HT eliciting a half-maximal contraction. Threshold = lowest concentration at which contraction is observed. 149 Discussion: Originally discovered in intestinal tract and blood, multiple functions of 5- HT in physiological and pathological conditions have been revealed. The loss of precise serotonergic system regulation (including 5-HT synthesis or metabolism, reuptake or release and 5-HT receptor expression) in the brain leads to mental disease. 5-HT is related to many peripheral vascular diseases but the role of a local serotonergic system has not been studied. In this study, we investigated the existence of a local serotonergic system in peripheral arteries and whether this local regulation of 5-HT was altered in hypertension. l. Biochemical Proof of 5-HT and its Metabolite in Peripheral Normal Arteries It was considered that peripheral arteries are only exposed to 5-HT from blood. Whether peripheral arterial smooth muscle has endogenous 5-HT was not known. The existence of endogenous 5-HT and its metabolite 5-HlAA is the fundamental tenet of our hypothesis -- a local serotonergic system exists in peripheral arteries. Three different techniques were used in our experiments. All results suggested the existence of 5-HT in peripheral arteries. Use of HPLC (Figure 4), as opposed to a classic assay using measurement of [3H]5-HT uptake, was critical, because we could measure both 5-HIAA and 5-HT in these assays. The existence of 5-HT in smooth muscle is supported by 5-HT immunoreactivity in smooth muscle layers and cytosol of freshly isolated smooth muscle cells that 150 are not exposed to blood, a potential source of extraneous 5-HT through blood platelets. While arteries contain mast cells, it is unlikely that these cells make a significant contribution to the 5-HT concentrations measured as few mast cells could be identified. Mast cell populations have been reported as rich in the coronary adventitia (Wolf et al., 1998), a site where there was in fact strong staining for 5-HT in our study (Figure 6). Additionally, mast cells have been found in femoral arteries (Mathiau et al., 1993). it is important to note that measures of mast cells were not made in the carotid and superior mesenteric arteries, and thus we cannot exclude the possibility that some of the 5-HT and 5- HIAA measured came from mast cells in these particular arteries. The staining of 5-HT antibody in isolated aortic smooth muscle cells (Figure 7) further supported that at least part of the 5-HT we measured using HPLC came from arterial smooth muscle cells. Thus, we quantified basal arterial endogenous 5-HIAA and 5-HT concentration by HPLC measurements and localized 5-HT in endothelium and peripheral arteries smooth muscle cells by immunostaining. ll. Investigation of 5-HT Synthesis in Peripheral Normal Arteries The presence of TPH is considered a hallmark of 5-HT synthesis. in literature, studies investigating the serotonergic innervation of peripheral arterial blood vessels were performed before the advent of PCR and antibody-based IHC and Western analyses. 151 As depicted in Figure 1, TPH, AADC and MAO A are enzymes responsible for each step of 5-HT synthesis and metabolism. The existence of these enzymes is an essential requirement for synthesizing and metabolizing 5-HT in peripheral arteries. We tried to identify the existence of TPH protein in peripheral artery by Western analysis and lHC. However, due to technique limitations of the antibodies, we were not able to visualize TPH protein in homogenates from rat or mouse aorta in Western analysis. This could be due to degradation of TPH protein in the procedure of protein isolation. The results from IHC experiments were not convincing either. Similar results were observed in both frozen sections (Figure 9) and paraffin embedded sections (pictures not shown) with relatively darker staining in sections incubated with primary antibody compared to no primary incubation. Thus, another experimental technique was necessary to identify the existence of TPH protein. Pictures from immunocytochemistry experiments showed a positive TPH antibody staining in freshly isolated aortic smooth muscle cells. To lend support to this finding, we did real time RT-PCR experiments to investigate the existence of tph1 mRNA. The results using two different sets of tph1 primers suggested the presence of tph1 mRNA in peripheral artery, which reinforced the result of existence of TPH protein in our immunocytochemistry experiment. We followed these measures with an activity assay. The rationale of our TPH activity assay was that the measurement of the accumulated 5-HTP in arteries, when AADC was inhibited by NSD1015, would indicate that a functional TPH exists. The assumption of this experiment was 152 that peripheral arteries do not take up 5-HTP (the 5-HTP that we measured was synthesized in arteries). Though we did observe a 5-HTP peak in arteries from NSD1015 treated rats and an increased 5-HTP was observed in NSD1015 + 5- HT synthesis substrate, tryptophan-treated rats, we cannot make the conclusion that this 5-HTP was synthesized by the peripheral artery as we also observed that peripheral arteries are capable of taking up 5-HTP (Figure 11). Thus, it is important to do an in vitro experiment to test the function of TPH in arteries. As we mentioned in the introduction, TPH is an extremely unstable enzyme, which loses its function quickly due to exposure to oxygen. In our in vitro TPH activity assay (Figure 12), we incubated isolated rat arteries with incubation solution, in which we added the reducing agent ferrous ammonium sulfate and D'I'I' to recover oxygen reduced TPH activity, added catalase to stabilize TPH, added 5- HT synthesis cofactor BH4, added NSD1015 to inhibit the conversion of 5-HTP to 5-HT and in some of our experiments we added tryptophan. There was no detectable 5-HTP in artery samples, while very low amount of 5-HTP was observed in rat raphe samples. We have two explanations for our data. The first, there is TPH expression in peripheral arteries. However, this TPH is not functional, at least in normal animals. The presence of TPH, AADC and BH4 (Gross et al., 1993) in arteries suggests the possibility of 5-HT synthesis. In disease condition, TPH activity, BH4 production and Ca2+lca|modulin-dependent protein kinase could change and TPH might be activated by these changes. Evidence supporting this speculation is increased Ca2*/calmodulin-dependent protein kinase activity in SHR (Boknik et al., 2001) and increased vascular gene 153 and protein expression of PKA in two-kidney, one clip (2K-1C) renal hypertensive vs. normotensive two kidney (2K) rats (Callera et al., 2004), which might lead to increased TPH activity. However, it was reported that in DOCA-salt hypertensive rats, the arterial BH4 level was decreased. The reduction of BH4 minimized the possibility that a functional TPH exists in peripheral arteries. The second explanation for our data is that there is 5-HT synthesis in peripheral arteries from normal rats. However, we are limited by the sensitivity of HPLC to measure this level of 5-HTP. Ill. Investigation of Metabolism of 5-HT in Peripheral Arteries Uptake study in arteries from normal rats (Figure 15) showed that after 15 minutes of incubation with exogenous 5-HT, the concentration of 5-HIAA in arteries was significantly higher than 5-HT, suggesting that 5-HT was taken up into blood vessel and rapidly metabolized. This observation suggests that peripheral arteries might play an important role in clearance of free circulating 5-HT in blood. IV. Investigation of Storage of 5-HT in Peripheral Arteries We do not believe that peripheral arteries function like platelets as a “5-HT sink”, which store and release 5-HT, because even in an extreme condition, in which we preload 5-HT in aorta from pargyline-treated rats, we did not observe that 5-HT could be kept in aorta. Peripheral arteries take up and metabolize 5- HT but do not store 5-HT. 154 We also investigated the localization and the form of endogenous intracellular 5-HT. Western analysis showed serotonin-modified proteins in arteries. Moreover, numerous proteins were reported as the substrates of TC", such as actin, myosin and troponin (Griffin et al., 2002). Whether this modification is linked to a functional change is not known yet. Other 5-HT could be packed in vesicles, as we observed fluramines-induced 5-HT release, although we do not have direct evidence showing the existing of VMAT in peripheral arteries. V. Investigation of functional SERT in Peripheral Normal Arteries We employed a multi-faceted approach in determining that peripheral smooth muscle has the potential to synthesize SERT. Real time RT-PCR and gel analyses of the final product after 35 cycles indicated the presence of one product. Translation of this message was confirmed by successfully measuring SERT protein product. Importantly, we confirmed immunohistochemically that a majority of the SERT is located in smooth muscle. Endothelial cells were stained positive for the SERT and this is consistent with previous findings (Lee et al., 1986; Small et al., 1977; Brust et al., 2000). Having demonstrated the presence of the SERT protein in arteries, we determined if peripheral arteries had the ability to take up 5-HT. In a simple assay (Figure 24), we demonstrated that 5-HT was taken up in a time-dependent manner by observing increased 5-HIAA with time in arteries. Because MAO A is an enzyme classically found in mitochondria, the increased arterial 5-HIAA after 155 exogenous 5-HT incubation suggested the ability of peripheral arteries to taken up 5-HT. We proved that this uptake is SERT-dependent by pharmacologically inhibition (Figure 28) and genetic ablation of SERT (Figure 29). VI. Investigation of SERT-independent 5-HT Uptake Mechanisms in Peripheral Arteries We noticed that neither pharmacological inhibition nor genetic ablation of SERT could abolish all the 5-HT uptake in arteries, which suggested that other SERT-independent mechanisms are involved in 5-HT uptake. One of the vessels we have used is the aorta, which contains sparse innervation by the sympathetic nervous system. Uptake in this minimally innervated tissue lends indirect support that it is the smooth muscle cells rather than nerves, takes up 5-HT (Kawasaki and Takasaki, 1984; Kawasaki et al. 1987). Moreover, in arteries from animals that have been chemically denervated or exposed to the NET inhibitor nisoxetine (Figure 32), uptake in all vessels remained normal. While these experiments do not exclude the possibility that sensory nerves could take up 5-HT, these data strongly support the idea that uptake of 5-HT into the artery is nerve- independent. Smooth muscle is not, however, the only arterial cell type that can take up 5-HT. Cultured endothelial cells take up and metabolize 5-HT (Lee et al., 1986; Small et al., 1977; Brust et al., 2000). By contrast, human fibroblasts appear to have a significantly lower rate of 5-HT uptake and metabolism (Small et al., 1977). In our experiments, the endothelial cell appears to make little contribution to 5-HT uptake in the whole 156 artery, though the SERT was located immunohistochemically to these same cells. The lack of effect of endothelial cell and neuron removal on 5-HT uptake may be because these two cell types do not contribute significantly and/or sufficiently to the total mass and uptake capability of the artery. Thus, the sensitivity of our assay (2—5 pg) may be insufficiently low, such that we were unable to reveal uptake by the endothelial cell and sympathetic neuron. This is a noted limitation of these studies. NET, DAT and SERT are all Na+-dependent transporters and use the energy provided by ATP. The uptake of 5-HT in Na+-free PSS demonstrated that part of 5-HT that crosses the cell membrane does not depend on Na+ moving downhill of the electrochemical gradient, which was built by Na“-K+ ATPase. We do not know, whether all of the 5-HT that passes the cell membrane is energy dependent, as in our experiment, arteries were incubated in a high concentration (compared to intracellular 5-HT concentration) of exogenous 5-HT (1 nM, 15 minutes) solutions. Experiments depleting ATP would help us answer the question whether the SERT-independent 5-HT uptake is active transport. OCTs are non-specific cation transporters. Though it has been reported that OCTs are upregulated in the gastrointestinal system in SERT-deficient mice and they might play a role in 5-HT uptake, we did not observe the OCTs inhibitor reducing 5-HT uptake in arteries from normal animals. We did not have a successful experience in identifying OCTs in arteries by Western analysis. We visualized multiple bands in our positive control as well as in our samples by using the antibody and there was no competing peptide available to verify the 157 specificity. OCTs are not involved in 5-HT uptake in normal condition but we could not exclude the possibility that in diseases or situations when the function of SERT lost, OCTs might start to take up 5-HT as a redundant mechanism. VII. Investigation of 5-HT Release Mechanisms in Peripheral Arteries (+)-Fenfluramine and (+)-norfenfluramine (fluramines) are SERT substrates and release 5-HT. The mechanism behind this effect is that fluramines are taken up by SERT into cytoplasm and then act on VMAT to release vesicular 5-HT. With the increased cytoplasmic 5-HT, SERT is reversed and starts to transport 5-HT outside of cells (Rothman and Baumann, 2002). If this is the only mechanism by which fluramines release 5-HT, we may conclude that part of endogenous 5-HT is stored in vesicles in peripheral artery smooth muscle. However, we do not yet have evidence to support the existence of the vesicles. The release of 5-HT from arteries may have significant physiological or pathological effect by changing the local 5-HT concentration. Because fluramines need to be taken up into smooth muscle cells to cause 5-HT release, SERT inhibition should reduce this effect. _ In our experiment, fluoxetine itself (1 uM) caused increases of 5-HT in PSS (Figure 37) during incubation, which is much higher than the amount of 5-HT' released by fluramines. Thus, the inhibition effect of fluramines-induced 5-HT release by fluoxetine could be masked by fluoxetine action. The observation of increased 5-HT in PS8 by fluoxetine suggests that there is a spontaneous release of 5-HT in the peripheral artery. in normal conditions, there could be a balance between 158 5-HT release and uptake in peripheral arteries. The mechanism of this spontaneous release is not clear, nor is the source of the intracellular 5-HT. VIII. Physiological Relevance of 5-HT Uptake in Peripheral Arteries and Speculation It has been argued that 5-HT plays an insignificant role in the physiological control of blood vessel tone as uptake of 5-HT by platelets and/or adrenergic nerves causes circulating levels of 5-HT to be low [estimated between 15—120 nM, Martin, 1994] and out of a range that can directly cause constriction. One manner by which physiological, autocrine/paracrine concentrations of 5-HT may affect vascular tone is by potentiating arterial contraction and mitogenesis to other vasoactive substances (Szabo et al.,1991; Yildiz et al, 1998; Watanabe et al. 2001). Thus, a local 5-HT system may play a significant role in affecting local contractility and mitogenesis by regulating local 5-HT concentration. This could not be discerned by measuring plasma concentrations of 5-HT. Because we have observed 5-HT uptake and the physical presence of the SERT in multiple arteries, this finding may be global in the body. Peripheral, non-pulmonary arteries could then be added to the growing list of already recognized sites of 5-HT uptake, including the brain, cerebral arteries (Scatton et al., 1985; Amenta et al., 1985; Brust et al., 1996), pulmonary arteries (Eddahibi, 1999), gut (Chen et al., 2001), pituitary (Johns et al., 1982), airway smooth muscle (Dodson et al., 2004) and cardiac myocytes (Sari and Zhou, 2003). The uptake and metabolism of 5-HT in peripheral arteries could play a role in 159 reducing 5-HT concentrations to maintain a normal physiological 5-HT content in free circulation. One can speculate that, dysfunction of this 5-HT uptake and metabolism system in peripheral arteries would lead to increased free circulating 5-HT and diseases related to that. IX. Pathological Relevance of 5-HT Uptake in Peripheral Arteries and Speculation The role of 5-HT and, more recently, its regulator, SERT, in hypertension is controversial and intriguing. The enhancement of potency for 5-HT in inducing vascular contraction or pressor responses in hypertension is striking. Increases in reactivity to 5-HT have been observed in a number of different forms of hypertension models including DOCA-salt hypertensive rats (Watts, 1998), SHR (Nishimura and Suzuki, 1995), and in human patients (Golino et al., 1991). Many mechanisms may contribute to this hyperresponsiveness such as changes in the 5-HT receptor signaling or changes in the circulating levels of 5-HT. It makes logical sense that a change of the activity of the 5-HT regulator (SERT) would change circulating 5-HT concentrations and thus change the responsiveness of arteries to 5-HT. We compared SERT function by testing 5-HT uptake of aorta from normotensive and hypertensive rats and tested whether changed in SERT expression and function were common to hypertension by using three hypertension models, the DOCA-salt rats, LNNA rats and SHR. These three models of hypertension are different in terms of mechanisms by which animals became hypertensive and the human hypertension they 160 resemble. DOCA-salt hypertension or mineralocorticoid hypertension resembles the clinical situation of aldosterone excess. DOCA-salt hypertension is caused by a surgical uninephrectomy followed by administration of a DOCA pellet and excess salt (1% NaCl+ 0.2% KCI in water). Blood pressure then rises within a few weeks into the hypertensive range. This is a sodium-dependent, low-renin model. Non-sodium mechanisms have been suggested to play a role in the development of DOCA-hypertension, including activation of sympathetic nervous system and vasopressin activation. LNNA inhibits all isoforms (endothelial, neuronal and inducible) of nitric oxide synthase (NOS), with a moderate selectivity for the endothelial isoform (Takahashi et al., 1995). Established hypertension was developed in two weeks after rats were given water with LNNA (0.5 g/L). The loss of nitric oxide production directly leads to a reduced nitric oxide-mediated vasodilation and superoxide “quenching” ability in blood vessels. Other mechanisms involved in LNNA-induced hypertension include activation of angiotensin converting enzyme (Charpie et al., 1997) and the AT1 receptor (Verhagen et al., 2000) but not vasopressin (Loichot et al., 2000). The model of LNNA hypertensive rats resembles a reduced bioavailability of nitric oxide in hypertensive individuals (Kelm, 2003, review). SHR develops hypertension and target organ damage similar to those seen in human essential hypertension. The blood pressure of SHR is relatively sodium-independent (Griffin et al., 2001). Therefore, investigation of the serotonergic system in these three models would answer the question of whether there is a change of component of 161 serotonergic system in hypertension and whether this is a general change in all hypertension. X. Basal 5-HT Concentration in Aorta of DOCA-salt, LNNA Hypertensive Rats and SHR As we stated earlier the free circulating plasma 5-HT levels (15-120 nM) (Martin, 1994) could not activate the 5-HT2A receptor (Ki= 100-3000 nM, PDSP Ki Database), which is the major receptor mediating 5-HT-induced arterial contraction in normotensive animals. However, many reports showed that plasma 5-HT concentrations and arterial 5-HT receptors are changed in hypertension. in essential hypertensive humans (Fetkovska et al., 1990) and cyclosporine-induced hypertensive rats (Reis et al., 1999), reduced levels of platelet 5-HT and increased plasma 5-HT concentration were measured. Consistent with these reports, unpublished work from our lab determined that the DOCA-salt rat platelet 5-HT was reduced (ng/ml of whole blood, SHAM(D)= 215.6:35.4, DOCA= 150.4:36.7) while free circulating 5-HT concentration was increased (ng/ml whole blood, SHAM(D) =5.82:1.14, DOCA= 28.1:11.1). The current observation of reduced basal 5-HT content in aorta from DOCA-salt rats suggests that arteries function as a metabolism organ for 5-HT with decreased 5- HT uptake ability in DOCA-salt hypertension. We speculate that the increased free circulating 5-HT in DOCA-salt rats may result from decreased 5-HT storage in and uptake by platelets and arteries. The increased extracellular free circulating 5-HT in DOCA-salt hypertensive rats may have significant effects. 5- 162 HT has high affinity for the 5-HT23 receptor (Ki= 10 nM, Bonhaus et al., 1995), and the expression and function of 5-HT23 receptor is upregulated in arterial smooth muscle in DOCA-salt hypertensive rats (Banes and Watts, 2003; Watts, 2002). This receptor is activated endogenously to maintain the high blood pressure of DOCA rats (Watts and Fink, 1999). Moreover, nanomolar concentrations of 5-HT potentiate arterial contraction to ET-1 and NE (Watts, 2000). Thus, it is possible that increased free circulating 5-HT concentration induces vasoconstriction, increases total peripheral resistance and thus elevates blood pressure in DOCA-salt hypertensive rats. Unfortunately, there are no literature reports of measurements of platelet 5-HT content or free circulating 5-HT concentration in the LNNA hypertensive rats. Our observation of decreased basal aortic 5-HT and 5-HIAA concentration suggests a changed SERT activity and changed level of circulating 5-HT in LNNA rats. Similar to the DOCA-salt rat, 5-HT23 receptor expression and function is increased in LNNA rats and is necessary for maintaining elevated blood pressure (Russell et al., 2002). It is possible that these changed factors of the serotonergic system in LNNA rats play roles in increasing and maintaining blood pressure. Further studies need to be done to investigate the change of platelet and free circulating 5-HT in LNNA rats. Only two studies in 1985 investigated platelet 5-HT level in SHR. As opposed to a decreased 5-HT in platelet in DOCA-salt hypertensive rats, SHR have more platelets and similar platelet 5-HT levels in SHR compared to WKY (Guicheney et al., 1985, a, b). Free circulating 5-HT in SHR has not been 163 reported. Our observation of similar basal 5-HT levels in aorta from SHR and WKY was consistent with these reports in platelet. We are aware that the basal level of 5-HT concentration is lower in WKY compared to Sprague-Dawley normotensive rats. This may suggest an inherent difference in these strains. The serotonergic system may have different degree of impact on blood pressure regulation in WKY compared to Sprague-Dawley rats. it is important to understand arterial function of SERT in hypertension not only as a reflection of circulating 5-HT levels, but also because intracellular 5-HT may have a function. Changing intracellular 5-HT concentration would in turn change arterial contraction. Thus, our observation of an altered basal activity of SERT in DOCA-salt and LNNA hypertension may have consequent pathological results. XI. 5-HT Uptake Ability was Reduced in Aorta of DOCA-salt and LNNA Hypertensive Rats, but not in SHR By normalizing 5-HT content to protein concentration and comparing the change of 5-HT concentrations in nglmg protein, our results showed a decreased total 5-HT uptake and a decreased SERT-dependent 5-HT uptake (Figure 40, 41) in aorta from DOCA-salt and LNNA hypertensive rats exposed to exogenous 5-HT, compared to that of aorta from their normotensive SHAM rats. We did not observe differences in 5-HT uptake in aorta from SHR and WKY. Our results suggest that SERT function was impaired in DOCA-salt and LNNA hypertensive rats. The decreased total 5-HT uptake observed in DOCA-salt and LNNA 164 hypertensive rats may account for the decreased basal aortic 5-HT content in both models and the increased free circulating 5-HT (DOCA-salt hypertension). Possible explanations for the contradictory findings of an upregulated SERT protein expression and a decreased SERT function in DOCA-salt and LNNA rats includes different membrane/cytosol distribution of SERT in these SHAM and hypertensive animals as well as post translational modification of SERT proteins. Phosphorylation of SERT by p38 MAPK increases SERT function by stimulating the insertion of intracellular SERT into the cell membrane and increasing total SERT activity (Blakely et al., 2005). By contrast, PKC reduces SERT function by phosphorylating SERT protein and causing translocation of SERT to the cytosol (Jayanthi et al., 2005). Thus, it is possible that the total expression of SERT increases in DOCA and LNNA hypertensive animals while the membrane fraction or functional SERT actually decreases. We did not observe a difference in basal aortic 5-HT content, 5-HT uptake ability or SERT protein expression in aorta from SHR compared to WKY rats. There are several factors that may contribute to the differences we observed. First, as we discussed above, the strain is different (WKY vs. Sprague-Dawley). Second, the systolic blood pressures of SHR (172:7 mmHg) were lower than DOCA-salt rats (197:6 mmHg) and LNNA rats (228:9 mmHg). The change of SERT expression and function may only happen in severe hypertension. Third, it is possible that the deoxycorticosterone we used in our DOCA-salt hypertension model causes the different SERT expression and function. However, Kulikov reported that stimulation of mineralocorticoid receptors had no effect on SERT 165 radioligand binding density in rat midbrain (Kulikov et al., 1997). Furthermore, the observation of impaired SERT function was also obtained in LNNA hypertensive rats. Thus, it is unlikely that the change of SERT function in DOCA- salt hypertension is from effects of mineralocorticoid. More importantly, a correlation between 5-HT23 receptor expression and plasma 5-HT or SERT function was reported recently (Callebert et al., 2006). Callebert et al. observed significantly increased plasma serotonin levels in wild type mice with upregulated 5-HT23 receptor after exposure to chronic hypoxia but not in 5-HT23 receptor-l- mice. Acute treatment with 5-HT23 receptor agonist induced a rapid SERT-and 5-HT23 receptor-dependent increase of plasma serotonin levels, which suggests that 5-HT23 receptor activation inhibits SERT function and thus less 5-HT is taken up. Upregulated 5-HT23 receptor expression and function in DOCA and LNNA hypertension have been reported (Watts and Fink, 1999, Watts, 2000; Russell et al., 2002). On the other hand, the 5-HT23 receptor was not involved in 5-HT-induced vasoconstriction in the hindquarters of SHR (Calama et al., 2004), which suggests 5-HT23 receptor was not activated in SHR as it was in DOCA-salt and LNNA hypertensive rats. Our results are consistent with Calleberts’ report in that we observed an increased free circulating 5-HT with increased 5-HT23 receptor function while decreased SERT function in DOCA rats; increased 5-HT23 receptor function while decreased SERT function in LNNA rats; no change of 5-HT23 receptor and normal SERT activity in SHR. How the 5-HT23 receptor regulates SERT is not yet clear. 166 XII. Other Arterial 5-HT-uptake Mechanism in Hypertensive Animals Similar to what we observed in normal animals, in arteries from hypertensive rats, a maximal concentration of fluoxetine (1 [nM) and fluvoxamine (1 uM) did not block the entire uptake stimulated by exogenous 5-HT. This suggests that there are still other mechanisms by which 5-HT is transported into aona. it is important to note that we studied one concentration of exogenous 5- HT at one time point in our active uptake study. Studies that examine 5-HT uptake at various times and with a range of 5-HT concentrations need to be done to compare SERT Km and Vmax values in aorta from DOCA-salt, LNNA hypertensive rats and their SHAM control rats. However, this is difficult without determination of other mechanisms for 5-HT uptake. Thus, kinetic studies have to wait until these alternative mechanisms are discovered. XIII. The Effect of SERT Function on Arterial Contractility in Hypertension Fluoxetine and fluvoxamine differed in their effects in aorta from normal rats, where a potentiation of 5-HT—induced contraction was observed with fluvoxamine but not fluoxetine. Interestingly, Gruetter et al (Gruetter et al., 1992) demonstrated nearly 10 years ago that 5-HT-induced contraction in aorta from normal rats could be markedly potentiated by citalopram, another SERT inhibitor, but only modestly by fluoxetine. Moreover, Cohen and Wiley (Cohen and Wiley, 1997) observed similar results in the relative inability of fluoxetine to potentiate aortic contraction to 5-HT. 167 How SERT modifies arterial contraction is not clear. We may speculate that the active uptake and metabolism of 5-HT in peripheral arteries is a protective mechanism of the body to keep a normal level of free circulating 5-HT and prevent blood pressure increases. When this uptake was inhibited by fluoxetine/fluvoxamine, peripheral arteries lost the ability to clear environmental 5-HT, and were exposed to a relatively higher concentration of 5-HT. In normotensive animals, 5-HT2A receptor-mediated 5-HT-induced contraction would not be changed because of the relatively low affinity of 5-HT for 5-HT2A receptor. The change of the amount of 5-HT by fluoxetine inhibition was not enough to cause change in aortic contraction. In hypertensive animals, the expression and function of 5-HT25 receptors (for which 5-HT has much higher affinity) are upregulated. This change of local 5-HT concentration might be sufficient to activate the 5-HT23 receptors to cause the potentiated contraction as we observed. This speculation can also be applied to explain the potentiated response to 5-HT in aorta from DOCA-salt rats compared to SHAM rats. The impaired SERT function in aorta from DOCA-salt rats suggest a loss of an arterial 5-HT clearance mechanism in these animals, which might lead to elevated free circulating 5-HT, vascular tone and blood pressure. The alternative explanation of our observation is that fluoxetine and fluvoxamine do not selectively inhibit SERT. Fluoxetine has fairly high affinity for rat 5-HT2A receptor (Ki= 299:31 nM, Rothman et al., 2000) and might cause inhibition of (+)-norfenfluramine-induced contraction. This is not consistent with our observation of potentiation of 5-HT-induced contraction in aorta from DOCA- 168 salt rats. The affinity of fluvoxamine for 5-HT2A receptor is over 12,000 nM (no affinity of fluvoxamine for 5-HT23 receptor has been reported). It is unlikely that the concentration we used in our experiments (1 uM) inhibited 5-HT2A receptor and 5-HT23 receptor. The important message from these particular findings is that inhibition of the transporter can affect contractility, and likely does so in a local manner. One can also speculate that the change of serotonergic system could also happen in arteries in which 5-HT plays a role in other pathophysiological states such as migraine and coronary vasospasm, and this has certainly been supported by findings in cerebral arteries (Amenta et al., 1985; Brust et al., 1996). With respect to blood pressure regulation, the finding of a functional peripheral SERT is potentially supported by findings that fluoxetine causes a pressor response in conscious rats (Lazarigues et al., 2000). XIV. Future Research The finding of a local serotonergic system in peripheral arteries is exciting and opens a new area of serotonin research. More studies need to be done to further characterize this system. Western analysis is the best way to quantify and compare protein expression. Showing the direct evidence of the existence of TPH1 protein by Western analysis is very important. A new TPH1 antibody has been reported recently (Sakowski et al., 2006). This is the first antibody that allows the differentiation of TPH1 and TPH2 upon immunoblotting, immunoprecipitation, 169 and immunocytochemical staining of tissue sections from brain (TPH2) and gut (TPH1). Recombinant TPH1 and TPH2 protein should be used as positive control in these future experiments. The activity of tryptophan hydroxylase is enhanced by phosphorylation of Ser58, by cAMP dependent PKA and Ca2*/calmodulin kinase II. Tryptophan hydroxylase (phospho 858) antibody is also commercially available and worthy to try. It is not yet known which proteins could be modified by intracellular 5-HT. With the knowledge of actin and myosin as substrates for TGII and important proteins for vasoconstriction, we could choose these proteins as candidates and perform an immunoprecipitation experiment with 5-HT antibody. This experiment would further elucidate which kind of protein could be modified by 5-HT. The in vitro measurement of TPH activity in peripheral arteries is still an ongoing project in our lab. It is possible that we could detect 5-HTP if we extend the incubation time and/or adjust other conditions, such as increasing tryptophan concentration. it is also worthwhile to test TPH activity in arteries in the presence/absence of PKA or Ca2*/calmodulin-dependent protein kinase activator and compare TPH activity in arteries. Whether a change in the local 5-HT concentration, by the mechanisms that we discussed in our studies, is sufficient to alter vascular tone is not clear. To answer this question, we could use the SERT substrate and 5-HT releaser (+)-fenfluramine to actively release endogenous 5-HT and test whether this is sufficient to cause vasoconstriction or to potentiatie the effect of other vasoconstrictors. 170 It is necessary to investigate SERT-independent 5-HT uptake mechanisms. After elucidating these mechanisms, we should be able to compare Km and Vmax of peripheral arterial SERT and brain SERT. XV. Limitations The original intention of this study was to determine the presence and potential function of the SERT in peripheral arteries. We have not compared this transporter, either kinetically, biochemically, or pharmacologically, to the classic transporters in the brain and the lung. Such work will be important to determine if these proteins could be potentially separate pharmacological targets. In our studies we used the thoracic aorta in RT-PCR, Western, IHC, and contractility assays. We are aware that the aorta is not a resistance artery and thus we repeated some of these studies in the smaller artery, superior mesenteric artery, which could be more similar to real resistance arteries. 171 Conclusions Unlike previous serotonin research in vasculature, which focused on serotonin receptor functions, we investigated whether peripheral arteries have the ability to regulate 5-HT concentration locally, and possible roles of this regulation in physiological and pathological conditions. Overall, we demonstrated the presence of a functioning serotonergic system in normal rat peripheral arteries. We proved the presence of the essential components for 5-HT synthesis, such as TPH and AADC, but we were limited by our technique to directly prove or disprove an active local peripheral arterial 5-HT synthesis. The existence of uptake and metabolism of 5-HT was clearly shown by observing increased 5-HIAA in isolated arteries from normal animals incubated with exogenous 5-HT (1 (M, 15 min). Using arteries from pargyline-treated rats, we studied the 5-HT uptake and metabolism mechanisms and eliminated the change of 5-HT concentration caused by metabolism. A basal- and fluramine-induced endogenous 5-HT release was observed, which indicated the possibility of local increases of extracellular and, accordingly, decreases of intracellular 5-HT concentration and as a consequence, changed 5- HT function. The 5-HT uptake of peripheral arteries is partially mediated by SERT, while other mechanisms involved in this process need to be further elucidated. There is no 5-HT storage in peripheral arteries as we observed that 5-HT content reduced back to basal level in 5-HT-loaded aorta after 4 hours incubation in PSS. Studies in arteries from hypertensive animals revealed a decreased basal endogenous 5-HT and increased expression but impaired 172 function of the SERT in peripheral arteries. SERT modifies 5-HT-induced contraction, and inhibition of the transporter is significantly enhanced in mineralocorticoid hypertension. Understanding the functions of the local serotonergic system in peripheral arteries in physiological and pathological conditions will help us reveal the etiology of 5-HT related peripheral vesicular diseases and find new targets for drug development. 173 Reference: Abenhaim L, Moride Y, Brenot F, Rich S, Benichou J, Kurz X, Higenbottam T, Oakley C, Wouters E, Aubier M, Simonneau G, Begaud B. Appetite-suppressant drugs and the risk of primary pulmonary hypertension. N Engl J Med; 1996.355: 609-616. Aitken A, Jones D, Soneji Y, Howell S. 14-3-3 proteins: biological function and domain structure. Biochem Soc Trans. 1995;23(3):605-611. Amenta F, DeRossi M, Mione MC, Geppetti P. Characterization of [3H]5- hydroxytryptamine uptake within rat cerebrovascular tree. Eur J Pharmacol. 1985;112:181-186. Armando l, Tjurmina OA, Li Q, Murphy DL, Saavedra JM. The serotonin transporter is required for stress-evoked increases in adrenal catecholamine synthesis and angiotensin II AT(2) receptor expression. Neuroendocrinology. 2003;78:217-225. Azzadin A, Mysliwiec M, Wollny T, Mysliwiec M, Buczko W. Serotonin is involved in the pathogenesis of hypertension developing during erythropoietin treatment in uremic rats. Thrombosis Res. 1995, 77: 217—224. Balasubramaniam G, Lee HS, Mah SC. Differences in the chronic hypotensive mechanism of action of ketanserin in spontaneously hypertensive and Wistar- Kyoto rats. J Hypertens. 1994;12(1):7-14. Banes AK, Watts SW. Arterial expression of 5-HT2B and 5-HT1B receptors during development of DOCA-salt hypertension. BMC Pharmacol. 2003; 3: 12— 37. Banik U, Wang GA, Wagner PD, Kaufman S. Interaction of phosphorylated tryptophan hydroxylase with 14-3-3 proteins. J Biol Chem. 1997;272(42):26219- 26225. Barrnan SA, lsales CM. Fenfluramine potentiates canine pulmonary vasoreactivity to endothelin-1. Pulm Pharmacol Ther. 1999;11:183-7. 1 Battaglino R, Fu J, Spate U, Ersoy U, Joe M, Sedaghat L, Stashenko P. Serotonin regulates osteoclast differentiation through its transporter. Bone Miner Res. 2004;19(9):1420-1431. Berndt TJ, Liang M, Tyce GM, Knox FG. lntrarenal serotonin, dopamine, and phosphate handling in remnant kidneys. Kidney Int; 2001, 59: 625-630. 174 Blakely RD, Berson HE, Fremeau RT, Caron MG, Peek MM, Prince HK, Bradley CC. Cloning and expression of a functional serotonin transporter from rat brain. Nature. 1991;354:66-70. Blakely RD, Ramamoorthy S, Schroeter S, Qian Y, Apparsundaram SD, Galli A, DeFelice LJ. Regulated phosphorylation and trafficking of antidepressant- sensitive serotonin transporter proteins. Biol Psychiatry. 1998;44:169-178. Blakely RD, Defelice LJ, Galli A. Biogenic amine neurotransmitter transporters: just when you thought you knew them. Physiology (Bethesda). 2005; 20:225-231. Review. Bonhaus DW, Bach C, DeSouza A, Salazar FH, Matsuoka BD, Zuppan P, Chan HW, Eglen RM. 5-HT vs 5-HT2B affinity: The pharmacology and distribution of human 5-hydroxytryptamine2B (5-HT2B) receptor gene products: comparison with 5-HT2A and 5-HT2C receptors. Br J Pharmacol. 1995; 115: 622-628. Browning RA, Bramlet DG, Myers JH, Bundman MC, Smith ML. Failure to produce blood pressure changes following pharmacological or surgical depletion of brain serotonin in the spontaneously hypertensive rat. Clin. Exp. Hypertens. 1981, 3: 953—973. Brownfield MS, Poff BC, Holzwarth MA. Ultrastructural immunocytochemical co- localization of serotonin and PNMT in adrenal medullary vesicles. Histochemistry. 1985;83(1):41-46. Brust P, Bergmann R, Johannsen B. High-affinity binding of [3H]paroxetine to caudate nucleus and microvessels from porcine brain. Neuroreport. 1996;711405—1408. Brust P, Friedrich A, Krizbai IA, Bergmann R, Roux F, Ganapathy V, Johannsen B. Functional expression of the serotonin transporter in immortalized rat brain microvessel endothelial cells. J Neurochem. 2000;74(3):1241-1248 Buckingham RE, Hamilton TC, Moore RA. Prolonged effects of p- chlorophenylalanine on the blood pressure of conscious normotensive and DOCA/saline hypertensive rats. Br. J. Pharmacol. 1976; 56: 69—75. Buzzi MG, Moskowitz MA. The pathophysiology of migraine: year 2005. J Headache Pain. 2005 ;6(3):105-11 1. Review. Calama E, Moran A, Ortiz de Urbina AV, Martin ML, San Roman L. Vasoconstrictor responses to 5-hydroxytryptamine in the autoperfused hindquarters of spontaneously hypertensive rats. Pharmacology. 2004; 71: 66— 72. 175 Callebert J, Esteve JM, Herve P, Peoc’h K, Tournois C, Drouet L, Launay JM, Maroteaux L. Evidence for a control of plasma serotonin levels by 5-HT2B receptors in mice. J Pharmacol Exp Ther. 2006; 317: 724-731. Callera JC, Bonagamba LG, Sevoz C, Laguzzi R, Machado BH. Cardiovascular effects of microinjection of low doses of serotonin into the NTS of unanesthetized rats. Am J Physiol. 1997;272(4 Pt 2):R1135-1142. Callera GE, Yeh E, Tostes RC, Caperuto LC, Carvalho CR, Bendhack LM. Changes in the vascular beta-adrenoceptor-activated signalling pathway in 2Kidney-1Clip hypertensive rats. Br J Pharmacol. 2004;141(7):1151-1158. Carrasco G, Cruz MA, Gallardo V, Miguel P, Lagos M, Gonzalez C. Plasma and platelet concentration and platelet uptake of serotonin in normal and pre- eclamptic pregnancies. Life Sci. 1998;62(15):1323-1332. Chang AS, Chang SM, Starnes DM, Schroeter S, Bauman AL, Blakely RD. Cloning and expression of the mouse serotonin transporter. Mol. Br. Res 1996;43:185-192. Chapman ME, Wideman RF. Hemodynamic responses of broiler pulmonary vasculature to intravenously infused serotonin. Poultry Sci. 2002, 81:231—238. Charpie JR, Charpie PM, Goud C, Pitt B, Webb RC. Quinapril prevents hypertension and enhanced vascular reactivity in nitroarginine-treated rats. Blood Press. 1997;6(2):117-124. Chen JJ, Li 2. Pan H, Murphy DL, Tamir H, Koepsell H, Gershon MD. Maintenance of serotonin in the intestinal mucosa and ganglia of mice that lack the high affinity serotonin transporter: abnormal intestinal motility and the expression of cation transporters. J. Neurosci. 2001;21:6348-6361. Chen NH, Reith MEA. Structure-function relationships for biogenic amine neurotransmitter transporters. In: Reith MEA, editor. Contemporary Neuroscience: Neurotransmitter Transporters, Functional and Regulation, 2nd ed. New JerseyzHuman Press Inc., 2002; p. 53-109. Cohen ML, Wiley KS. Neuronal uptake inhibitors, nisoxetine and fluoxetine, on rat vascular contractions. Eur J Pharmacol. 1977;44:219—229. Cohen ML, Fuller RW, Kurz KD. Evidence that blood pressure reduction by serotonin antagonists is related to alpha receptor blockade in spontaneously hypertensive rats. Hypertension. 1983; 5: 676—681. 176 Cohen ML, Schenck KW, Kurz KD. 5-HT2-receptor antagonists: alpha 1- vs. 5- HT2-receptor blocking properties in blood vessels. J Cardiovasc Pharmacol. 1988;11 Suppl 12825-29. Cohen ML, Schenck KW, Hemrick-Luecke SH. 5-Hydroxytryptamine 1A receptor activation enhances norepinephrine release from nerves in the Rabbit saphenous vein. J Pharmacol Exp Ther. 1999;290(3):1195-1201. Collis MG, Vanhoutte PM. Vascular reactivity of isolated perfused kidneys from male and female spontaneously hypertensive rats. Circ Res. 1977;41(6):759- 767. Collis MG, Vanhoutte PM. Tachyphylaxis to 5-hydroxytryptamine in perfused kidneys from spontaneously hypertensive and normotensive rats. J. Cardiovasc. Pharmacol. 1981,3: 229—235. Connolly HM, Crary JL, McGoon MD, Hensrud DD, Edwards BS, Edwards WD Schaff HV. Valvular heart disease associated with fenfluramine-phentermine. N Engl J Med. 1997; 337: 581-588. Cooke JP, Marshall JM. Mechanisms of Raynaud's disease. Vasc Med. 2005;10(4):293-307. Review. Cooper JR, Bloom FE and Roth RH. Serotonin (5-Hydroxytryptamine), Histamine, and Adenosin. The Biochemical Basis of Neuropharrnacology, 8th ed. New York, Oxford University Press, 2002;p271-320. Cooper, JR, Bloom FE, Roth RH. Serotonin (5-hydroxytryptamine), histamine and adenosine. In: The Biochemical Basis of Neuropharmacology. Oxford University Press, 2003. p.271-320. Cote F, Thevenot E, Fligny C, Fromes Y, Darrnon M, Ripoche MA, Bayard E, Hanoun N, Saurini F, Lechat P, Dandolo L, Hamon M, Mallet J, Vodjdani G. Disruption of the nonneuronal tph1 gene demonstrates the importance of peripheral serotonin in cardiac function. Proc Natl Acad Sci U S A. 2003 11;100(23):13525-13530. Cote F, Fligney C, Fromes Y, Mallet J and Vodjdani G. Recent advances in understanding serotonin regulation of cardiovascular function. Trends Mol. Med. 2004,10: 232—238. Coto E, Reguero JR, Alvarez V, Morales B, Batalla A, Gonzalez P, Martin M, Garcia-Castro M, lglesias-Cubero G, Cortina A. 5-Hydroxytryptamine 5-HT2A receptor and 5-hydroxytryptamine transporter polymorphisms in acute myocardial infarction. Clin Sci (Lond). 2003;104(3):241-245. 177 Dalton DW, Feniuk W, Humphrey PP. An investigation into the mechanisms of the cardiovascular effects of 5-hydroxytryptamine in conscious normotensive and DOCA-salt hypertensive rats. J. Auton. Pharmacol. 1996,6: 219—228. Delarue C, Becquet D, Idres S, Hery F, Vaudry H. Serotonin synthesis in adrenochromaffin cells. Neuroscience. 1992;46:495-500. De Luca Sarobe V, Nowicki S, Carranza A, Levin G, Barontini M, Arrizurieta E, lbarra FR. Low sodium intake induces an increase in renal monoamine oxidase activity in the rat. Involvement of an angiotensin II dependent mechanism. Acta Physiol Scand. 2005;185(2):161-167. Desta B, Steed S, Ravel D, Laudignon N, Vanhoutte PM, Boulanger CM. Acute and chronic effects of dexfenfluramine on the porcine pulmonary artery. Gen Pharmacol. 1998;30:403-410. Ding YA, Chou TC, Huan R. Are platelet cytosolic free calcium, serotonin concentration and blood viscosity different between hypertensive and normotensive subjects? Cardiology. 1994;85:76-81. Docherty JR. An investigation of the effects of intravenous injection of the 5- hydroxytryptamine2 receptor antagonists ketanserin and LY53857 on blood pressure in anesthetized spontaneously hypertensive rats. J. Pharmacol. Exp. Ther. 1989, 248; 736—740. Dodson AM, Anderson GM, Rhoden KJ. Serotonin uptake and metabolism by cultured guinea pig airway smooth muscle cells. Pulm Pharmacol Ther. 2004;17:19—25. Doggrell SA. The role of 5-HT on the cardiovascular and renal systems and the clinical potential of 5-HT modulation. Expert Opin lnvestig Drugs. 2003;12(5):805-823. Eddahibi S, Fabre V, Boni C, Marteres MP, Reffesting B, Hamon M, Adnot S. Induction of serotonin transporter by hypoxia in pulmonary vascular smooth muscle cells. Relationship with the mitogenic action of serotonin. Circ. Res. 1999;84:329-336. Eddahibi S, Hanoun N, Lanfumey L, Lesch K, Raffestin B, Hamon M, Adnot S. Attenuated hypoxia pulmonary hypertension in mice lacking the 5- hydroxytryptamine transponer gene. J. Clin. Invest. 2000;105:1555-1562. Eddahibi S, Humbert M, Fadel E, Raffestin B, Darmon M, Capron F, Simonneau G, Dartevelle P, Hamon M, Adnot S. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin Invest. 2001;108(8):1141-1150. 178 Eddahibi E, Adnot S. Anorexigen-induced pulmonary hypertension and the serotonin (5-HT) hypothesis: lessons for the future in pathogenesis. Respir. Res. 2002;3z9-12. Eddahibi S, Chaouat A, Morrell N, Fadel E, Fuhnnan C, Bugnet AS, Dartevelle P, Housset B, Hamon M, Weitzenblum E, Adnot S. Polymorphism of the serotonin transporter gene and pulmonary hypertension in chronic obstructive pulmonary disease. Circulation. 2003;108(15):1839-1844. Fetkovska N, Amstein R, Ferracin F, Regenass M, Buhler FR, Pletscher A. 5- Hydroxytryptamine kinetics and activation of blood platelets in patients with essential hypertension. Hypertension. 1990; 15: 267—273. Fregly MJ, Lockley OE, van der Voort J, Sumners C and Henley WN. Chronic dietary administration of tryptophan prevents the development of deoxycorticosterone acetate induced hypertension in rats. Can. J. Pharmacol. Physiol. 1987, 65: 753—764. Fumeron F, Betoulle D, Nicaud V, Evans A, Kee F, Ruidavets JB, Arveiler D, Luc G, Cambien F. Serotonin transporter gene polymorphism and myocardial infarction: Etude Cas-TAmoins de l'lnfarctus du Myocarde (ECTIM). Circulation. 2002;105(25):2943-2945. Fusegawa Y, Hashizume H, Okumura T, Deguchi Y, Shina Y, Ikari Y, Tanabe T. Hypertensive patients with carotid artery plaque exhibit increased platelet aggregability. Thromb Res. 2006;117(6):615-622. Galligan JJ, Furness JB, Costa M. Effects of cholinergic blockade, adrenergic blockade and sympathetic denervation on gastrointestinal myoelectric activity in guinea pig. J Pharmacol Exp Ther. 1986;238:1114-1125. Garattini S. Biological actions of drugs affecting serotonin and eating. Obes Res. 1995;3 Suppl 4:463S-47OS. Gershon MD, Liu KP, Karpiak SE, Tamir H. Storage of serotonin in vivo as a complex with serotonin-binding protein in central and peripheral serotonergic neurons. J Neurosci. 1983;3(10):1901-1911. Gillis CN, Pitt BR. The fate of circulating amines within the pulmonary circulation. Annu Rev Physiol. 1982;44:269-281. Glusa E, Pertz HH. Further evidence that 5-HT-induced relaxation of pig pulmonary artery is mediated by endothelial 5-HT(2B) receptors. Br J Pharmacol. 2000;130(3):692-698. 179 Golino P, Piscione F, Willerson JT, Cappelli-Bigazzi M, Focaccio A, Villari B, Indolfi C, Russolillo E, Condorelli M, Chiariello M. Divergent effects of serotonin on coronary-artery dimensions and blood flow in patients with coronary atherosclerosis and control patients. N Engl J Med. 1991;324(10):641-648. Gonzalez AM, Smith AP, Emery CJ, Higenbottam TW. The pulmonary hypertensive fawn-hooded rat has a normal serotonin transporter coding sequence. Am J Respir Cell Mol Biol. 1998;19:245-249. Gorman JM, Kent JM. SSRIs and SMRls. broad spectrum of efficacy beyond major depression. Clin Psychiatry. 1999;60 Suppl 4:33-38; discussion 39. Gradin K, Pettersson A, Hedner T and Persson B. Chronic 5-HT2 receptor blockade with ritanserin does not reduce blood pressure in the spontaneously hypertensive rat. J. Neural Transm. 1985, 64: 145—149. Greenberg BD, Tolliver TJ, Huang S-J, Li Q, Bengel D, Murphy DL. Genetic variation in the serotonin transporter promoter region affects serotonin uptake in human blood platelets. Am J Med Genet. 1999;88:83-87. Griffin KA, Churchill PC, Picken M, Webb RC, Kurtz TW, Bidani AK. Differential salt-sensitivity in the pathogenesis of renal damage in SHR and stroke prone SHR. Am J Hypertens. 2001;14(4 Pt 1):311-320. Griffin M, Casadio R, Bergamini CM. Transglutaminases: Nature’s biological glues. Biochem J. 2002;368:377-396. Gross SS, Levi R, Madera A, Park KH, Vane J, Hattori Y. Tetrahydrobiopterin synthesis is induced by LPS in vascular smooth muscle and is rate-limiting for nitric oxide production. Adv Exp Med Biol. 1993;338:295-300. Review. Gruetter CA, Lemke SM, Anestis DK, Szarek JL, Valentovic MA. Potentiation of 5-hydroxytryptamine-induced contraction in rat aorta by chlorpheniramine, citalopram and fluoxetine. Eur J Pharmacol. 1992;217:109—118. Gu H, Wall SC, Rudnick G. Stable expression of biogenic amine transporters reveals differences in inhibitor sensitivity, kinetics, and ion dependence. J Biol Chem. 1994;269(10):7124-7130. a. Guicheney P, Baudouin-Legros M, Garnier JP, Roques P, Dreux C, Meyer P. Platelet serotonin and blood tryptophan in spontaneously hypertensive and normotensive Wistar-Kyoto rats. J Cardiovasc Pharmacol. 1985; 7: $15-$17. b. Guicheney P, Legros M, Marcel D, Kamal L, Meyer P. Platelet serotonin content and uptake in spontaneously hypertensive rats. Life Sci. 1985; 36: 679- 685. 180 Hafdi Z, Couette S, Comoy E, Prie D, Amiel C and Friedlander G. Locally formed 5-hydroxytryptamine stimulates phosphate transporte in cultured opossum kidney cells and in rat kidney. Biochem. J. 1996, 320: 615—621. Hara K, Hirowatari Y, Yoshika M, Komiyama Y, Tsuka Y, Takahashi H. The ratio of plasma to whole-blood serotonin may be a novel marker of atherosclerotic cardiovascular disease. J Lab Clin Med. 2004;144(1):31-37. Hayashi M, Haga M, Yatsushiro S, Yamamoto A, Moriyama Y. Vesicular monoamine transporter 1 is responsible for storage of 5-hydroxytryptamine in rat pinealocytes. J Neurochem. 1999;73(6):2538-2545. Heils A, Teufel A, Petri S, Stobeer G, Riederer P, Bengel D, Lesch KP. Allelic variation of human serotonin transporter gene expression. J. Neurochem. 1996;66:2621-2624. Hergovich N, Aigner M, Eichler HG, Entlicher J, Drucker C, Jilma B. Paroxetine decreases platelet serotonin storage and platelet function in human beings. Clin Pharmacol Ther. 2000;68(4):435-442. Herve P, Launay JM, Scrobohaci ML, Brenot F, Simonneau G, Petitpretz P, Poubeau P, Cerrina J, Duroux P, Drouet L. Increased plasma serotonin in primary pulmonary hypertension. Am J Med. 1995;99(3):249-254. Hoffman BJ, Mezey E, Brownstein MJ. Cloning of a serotonin transporter affected by antidepressants. Science 1991;254:579-580. Holmsen H, Weiss HJ. Secretable storage pools in platelets. Annu Rev Med. 1979;30:119-134. Review. Holtje M, VWnter S, Walther D, Pahner I, Hortnagl H, Ottersen OP, Bader M, Ahnert-Hilger G. The vesicular monoamine content regulates VMAT2 activity through Galphaq in mouse platelets. Evidence for autoregulation of vesicular transmitter uptake. J Biol Chem. 2003;278(18):15850-15858. Horvath G, Sutto Z, Torbati A, Conner GE, Salathe M. Wanner A. Norepinephrine transport by the extraneuronal monoamine transporter in human bronchial arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2003;285zL829-837. Howe PR, Rogers PF, King RA, Smith RM. Elevation of blood pressure in hypertensive rats after lesioning serotonin nerves in the dorsomedial medulla oblongata. Clin. Exp. Pharmacol. Physiol. 2003, 10: 273—277. 181 lkeda K, Tojo K, Otsubo C, Udagawa T, Kumazawa K, lshikawa M, Tokudome G, Hosoya T, Tajima N, Claycomb WC, Nakao K, Kawamura M. 5- hydroxytryptamine synthesis in HL-1 cells and neonatal rat cardiocytes. Biochem Biophys Res Commun. 2005;328:522-525. lsbister GK, Bowe SJ, Dawson A, Whyte IM. Relative toxicity of selective serotonin reuptake inhibitors (SSRIs) in overdose. J Toxicol Clin Toxicol 2004;42:277-285. Ito H, Shiokawa H, Torii M, Suzuki, T. Effects of tryptophane on SHRSP offspring growth. Clin. Exp. Hypertens. 1991, 13: 971—979. Jayanthi LD, Samuvel DJ, Blakely RD, Ramamoorthy S. Evidence for biphasic effects of protein kinase C on serotonin transporter function, endocytosis, and phosphorylation. Mol Pharmacol. 2005; 67: 2077—2087. Jiang GC, Yohrling GJ 4th, Schmitt JD, Vrana KE. Identification of substrate orienting and phosphorylation sites within tryptophan hydroxylase using homology-based molecular modeling. J Mol Biol. 2000 ;302(4):1005-1017. Johns MA, Azmitia EC, Krieger DT. Specific in vitro uptake of serotonin by cells in the anterior pituitary of the rat. Endocrinology. 1982;110:754—760. Kamal LA, Le Quan-Bui KH, Meyer P. Decreased uptake of 3H-serotonin and endogenous content of serotonin in blood platelets in hypertensive patients. Hypertension. 1984;62568-573. Kawasaki H, Takasaki K. Vasoconstrictor response induced by 5- hydroxytryptamine released from vascular adrenergic nerves by periarterial nerve stimulation. J Pharmacol Exp Ther. 1984;229(3):816-822. Kawasaki H, Urabe M, Takasaki K. Enhanced 5-hydroxytryptamine release from vascular adrenergic nerves in spontaneously hypertensive rats. Hypertension. 1987;10(3):321-327. Kelly CA, Dhaun N, Laing WJ, Strachan FE, Good AM, Bateman DN. Comparative toxicity of citalopram and the newer antidepressants after overdose. J Toxicol Clin Toxicol. 2004;42:67-71. Kelm M. The L-arginine-nitric oxide pathway in hypertension. Curr Hypertens Rep. 2003;5(1):80-86. Review. Kereveur A, Callebert J, Humbert M, Herve P, Simonneau G, Launay JM, Drouet L. High plasma serotonin levels in primary pulmonary hypertension. Effect of long-term epoprostenol (prostacyclin) therapy. Arterioscler Thromb Vasc Biol. 2000;20(10):2233-2239. 182 Kilic F, Rudnick G. Oligomerization of the serotonin transporter and its functional consequences. Proc. Natl. Acad. Sci. USA, 2000;97:3106-3111. Kocabas AM, Rudnick G, Kilic F. Functional consequences of homo- but not hetero-oligomerization between transporters for the biogenic amine neurotransmitters. J Neurochem 2003;85:1513-1520. Kowlessur D, Kaufman S. Cloning and expression of recombinant human pineal tryptophan hydroxylase in Escherichia coli: purification and characterization of the cloned enzyme. Biochim Biophys Acta. 1999;1434(2):317-330. Krygicz D, Azzadin A, Pawlak R, Malyszko JS, Pawlak D, Mysliwiec M, Buczko W. Cyclosporine A affects serotonergic mechanisms in uremic rats. Pol. J. Pharmacol. 1996,48: 351—354. Kuhn DM, Rosenberg RC, Lovenberg W. Determination of some molecular parameters of tryptophan hydroxylase from rat midbrain and murine mast cell. J Neurochem. 1979;33(1):15-21. Kuhn DM, Arthur R Jr. Molecular mechanism of the inactivation of tryptophan hydroxylase by nitric oxide: attack on critical sulfhydryls that spare the enzyme iron center. J Neurosci. 1997;17(19):7245-7251. Kuhn DM, Arthur R Jr, States JC. Phosphorylation and activation of brain tryptophan hydroxylase: identification of serine-58 as a substrate site for protein kinase A. J Neurochem. 1997;68(5):2220-2223. Kuhn DM, Geddes TJ. Molecular footprints of neurotoxic amphetamine action. Ann N Y Acad Sci. 2000;914:92-103. Kulikov A, Mormede P, Chaouloff F. Effects of adrenalectomy and corticosterone replacement on diurnal [3H]citalopram binding in rat midbrain. Neurosci Lett. 1997; 222: 127-131. Kurne E, Ertugrul A, Anil Yagcioglu AE, Yazici KM. Venous thromobembolism and escilatolopram. Gen Hosp Psychiatry. 2004;26:481-483. Launay JM, Herve P, Peoc'h K, Tournois C, Callebert J, Nebigil CG, Etienne N, Drouet L, Humbert M, Simonneau G, Maroteaux L. Function of the serotonin 5- hydroxytryptamine 23 receptor in pulmonary hypertension. Nat Med. 2002;8(10):1129-1135. Launay JM, Schneider B, Loric S, Da Prada M, Kellermann O. Serotonin transport and serotonin transporter-mediated antidepressant recognition are 183 controlled by 5-HT2B receptor signaling in serotonergic neuronal cells. FASEB J. 2006 ;20(11):1843-1854. Lazartigues E, Brefel-Courbon C, Bagheri H, Costes S, Gharib C, Tran MA, Senard JM, Montastruc JL. Fluoxetine-induced pressor response in freely moving rats: a role for vasopressin and sympathetic tone. Fundam Clin Pharmacol. 2000;14:443—451. Lee SL, Fanburg BL. Serotonin uptake by bovine pulmonary artery endothelial cells in culture. I. Characterization. Am J Physiol. 1986;250(5 Pt 1):C761-765. Lee SL, Wang W, Fanburg BL. Dexfenfluramine as a mitogen signal via the formation of superoxide anion. FASEB J. 2001;15:1324-1325. Linden A, Desmecht D, Amory H, Lekeux P. Cardiovascular response to intravenous administration of 5-hydroxytryptamine after type-2 receptor blockade, by metrenperone, in healthy calves. Vet. J. 1999, 157: 31—37. Loichot C, Cazaubon C, Grima M, De Jong W, Nisato D, lmbs JL, Barthelmebs M. Vasopressin does not effect hypertension caused by long-term nitric oxide inhibition. Hypertension. 2000;35(2):602-608. MacLean MR, Deuchar GA, Hicks MN, Morecroft I, Sheri S, Sheward J, Colston J, Loughlin L, Nilsen M, Dempsie Y, Harmar A. Overexpression of the 5- hydroxytryptamine transporter gene. Effect on pulmonary hemodynamics and hypoxia-induced pulmonary hypertension. Circ. 2004;109:2150-2155. Magnani F, Tate CG, Wynne S, Williams C, Haase J. Partitioning of the serotonin transporter into lipid microdomains modulates transport of serotonin. J Biol Chem. 2004;279(37):38770-38778. Manoli I, Le H, Alesci S, McFann KK, Su YA, Kino T, Chrousos GP, Blackman MR. Monoamine oxidase-A is a major target gene for glucocorticoids in human skeletal muscle cells. FASEB J. 2005;19(10):1359-1361. Marcos E, Adnot S, Pham MH, Nosjean A, Raffestin B, Hamon M, Eddahibi S. Serotonin transporter inhibitors protect against hypoxic pulmonary hypertension. Am J Respir Crit Care Med. 2003;168:487-493. Martel F, Azevedo I. An update on the extraneuronal monoamine transporter (EMT) characteristics, distribution and regulation. Curr Drug Metab. 2003;42313- 318. Martin GR. Vascular receptors for 5-hydroxytryptamine: distribution, function and classification. Pharmacol Ther. 1994,62(3): 283-324. 184 Mathiau P, Reynier-Rebuffel AM, lssertial O, Callebert J, Decreme C, Aubineau P. Absence of serotonergic innervation from raphe nuclei in rat cerebral blood vessels-4|. Lack of tryptophan hydroxylase activity in vitro. Neuroscience. 1993;52(3):657-665. Maurer-Spurej E. Serotonin reuptake inhibitors and cardiovascular disease: a platelet connection. Cell Mol Life Sci. 2005; 62:159-170. McCafferty DM, Wallace JL, and Sharkey KA. Effects of chemical sympathectomy and sensory nerve ablation on experimental colitis in the rat. Am J Physiol. 1997, 272: G272-6280. McCall RB, Harris LT. Characterization of the central sympathoinhibitory action of ketanserin. J Pharmacol Exp Ther. 1987 May;241(2):736-740. McDuffie JE, Motley ED, Limbird LE, Maleque MA. 5-hydroxytryptamine stimulates phosphorylation of p44/p42 mitogen-activated protein kinase activation in bovine aortic endothelial cell cultures. J Cardiovasc. Pharmacol. 2000;35:398-402. McGregor DD, Smirk FH. Vascular responses to 5-hydroxytryptamine in genetic and renal hypertensive rats. Am J Physiol. 1970;219(3):687-690. MacLennan SJ, Bolofo ML, Martin GR. Amplifying interactions between spasmogens in vascular smooth muscle. Biochem. Soc. Trans. 1993, 21: 1145— 1 150. Marcos E, Adnot S, Pham MH, Nosjean A, Raffestin B, Hamon M, Eddahibi S. Serotonin transporter inhibitors protect against hypoxic pulmonary hypertension. Am J Respir Crit Care Med. 2003 Aug 15;168(4):487-493. Mene P, Pugliese F and Cinott GA. Serotonin and the glomerular mesangium. Mechanisms of intracellular signaling. Hypertension. 1991,17z151-160. Mlinar B, Corradetti R. Endogenous 5-HT, released by MDMA through serotonin transporter- and secretory vesicle-dependent mechanisms, reduces hippocampal excitatory synaptic transmission by preferential activation of 5-HT1B receptors located on CA1 pyramidal neurons. Eur J Neurosci. 2003;18:1559-1571. Morecroft I, Heeley RP, Prentice HM, Kirk A, MacLean MR. 5-hydroxytryptamine receptors mediating contraction in human small muscular pulmonary arteries: importance of the 5-HT1 B receptor. Br J Pharmacol. 1999;128:730-734. Morecroft l, Loughlin L, Nilsen M, Colston J, Dempsie Y, Sheward J, Harmar A, MacLean MR. Functional interactions between 5-hydroxytryptamine receptors 185 and the serotonin transporter in pulmonary arteries. J Pharmacol Exp Ther. 2005;313:539-548. Murata R, Hamada N, Nakamura N, Kobayashi A, Fukueda M, Taira A, Sakata R. Serotonin activity and liver dysfunction following hepatic ischemia and reperfusion. In Vivo. 2003;17(6):567-572. Murphy DL, Lerner A, Rudnick G, Lesch KP. Serotonin transporter: gene, genetic disorders, and pharmacogenetics. Mol. lnterv. 2004;4z109-123. Myers MG, Reid JL, Lewis, P. J. The effect of central serotonin depletion on DOCA-saline hypertension in the rat. Cardiovasc. Res. 1974, 8: 806—810. Nebigil CG, Maroteaux L. A novel role for serotonin in heart. Trends Cardiovasc. Med. 2001, 11: 329-335. Nelson M, Coghlan JP, Denton DA, Tresham JJ, Whitworth JA, Scoggins BA. Ritanserin and serotonergic mechanisms in blood pressure and fluid regulation in sheep. Clin. Exp. Pharmacol. Physiol. 1987, 14: 555-563. Nemecek GM, Coughlin SR, Handley DA, Moskowitz MD. Stimulation of aortic smooth muscle cell mitogenesis by serotonin. Proc Natl Acad Sci USA 1986;83:674-678. Nilsson O, Ericson LE, Dahlstrom A, Ekholm R, Steinbusch HW, Ahlman H. Subcellular localization of serotonin immunoreactivity in rat enterochromaffin cells. Histochemistry. 1985;82(4):351-355. Nishimura Y, Suzuki A. Enhanced contractile responses mediated by different 5- HT receptor subtypes in basilar arteries, superior mesenteric arteries and thoracic aortas from stroke-prone spontaneously hypertensive rats. Clin Exp Pharmacol Physiol Suppl. 1995;22(1):S99-101. Otani K, Tybring G, Mihara K, Yasui N, Kaneko S, Ohkubo T, Nagasaki T, Sugawara K. Correlation between steady-state plasma concentrations of mianserin and trazodone in depressed patients. Eur J Clin Pharmacol. 1998;53(5):347-349. ‘ Ozaslan D, Wang S, Ahmed BA, Kocabas AM, McCastlain JC, Bene A, Kilic F. Glycosyl modification facilitates homo- and hetero-oligomerization of the serotonin transporter. A specific role for sialic acid residues. J. Biol Chem 2003;278:43991-44000. Pacher P, Ungvari Z, Kecskemeti V, F urst S. Review of cardiovascular effects of fluoxetine, a selective serotonin reuptake inhibitor, compared to tricyclic antidepressants. Curr Med Chem. 1998;52381-390. 186 Pacher P, Ungvari Z, Nanasi PP, Furst S, Kecskemeti V. Speculations on difference between tricyclic and selective serotonin reuptake inhibitor antidepressants on their cardiac effects. Is there any? Curr Med Chem. 1999;62469-480. Ramage AG. Examination of the effects of some 5-HT2 receptor antagonists on central sympathetic outflow and blood pressure in anaesthetized cats. Naunyn- Schmiedeberg’s Arch Pharmacol. 1988, 338: 601-607. Pandey A, Habibulla M, Singh R. Tryptophan hydroxylase and 5-HTP- decarboxylase activity in cockroach brain and the effects of p- chlorophenylalanine and 3-hydroxybenzylhydrazine (NSD-1015). Brain Res. 1983;273(1):67-70. Ramammoorthy S. Regulation of Monoamine transporters: regulated phosphorylation, dephosphorylation and trafficking. In: Reith MEA, editor. Contemporary Neuroscience: Neurotransmitter Transporters, Function and Regulation, 2nd ed. New JerseyzHuman Press Inc., 2002, pp. 1-23. Ramamoorthy S, Bauman AL, Moore KR, Han H, Yang-Feng T, Chang AS, Ganapathy V, Blakely RD. Antidepressant and cocaine-sensitive human serotonin transporter: molecular cloning, expression and chromosomal localization. Proc Natl Acad Sci USA. 1993;90:2542-2546. Ramamoorthy S, Giovanetti E, Qian Y, Blakely RD. Phosphorylation and regulation of antidepressant-sensitive serotonin transporters. J. Biol. Chem. 1998;273:2458-2466. Ramamoorthy S, Blakely RD. Phosphorylation and sequestration of serotonin transporters differentially modulated by psychostimulants. Science. 1999 ;285(5428):763-766. Reis F, Tavares P, Fontes Ribeiro CA, Antunes F, Teixeira F. The peripheral serotonergic system and platelet aggregation in cyclosporin A-induced hypertensive rats. Thromb Res. 1999; 96: 365—372. PDSP Ki Database. Available at: http://kidb.cwru.edu/pdsp.php. Accessed April 20,2006. Peter D, Jimenez J, Liu Y, Kim J, Edwards RH. The chromaffin granule and synaptic vesicle amine transporters differ in substrate recognition and sensitivity to inhibitors. J Biol Chem. 1994;269(10):7231-7237. 187 Pihel K, Hsieh S, Jorgenson JW, Wightman RM. Quantal corelease of histamine and 5-hydroxytryptamine from mast cells and the effects of prior incubation. Biochemistry. 1998;37(4):1046-1052. Rothman RB, Ayestas MA, Dersch CM, Baumann MH. Aminorex, fenfluramine, chlorphentermine are serotonin transporter substrates; implications for primary pulmonary hypertension. Circ. 1999;100:869-875. Rothman RB, Baumann MH, Savage JE, Rauser L, McBride A, Hufeisen SJ, Roth BL. Evidence for possible involvement of 5-HT(2B) receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medications. Circulation. 2000;102(23):2836-2841. Rothman RB, Baumann MH. Therapeutic and adverse actions of serotonin transporter substrates. Pharmacol Ther. 2002;95(1):73-88. Russell A, Banes A, Berlin H, Fink GD, Watts SW. 5-Hydroxytryptamine(2B) receptor function is enhanced in the N(omega)-nitro-L-arginine hypertensive rat. J Pharmacol Exp Ther. 2002;303(1):179-187. Sakowski SA, Geddes TJ, Thomas DM, Levi E, Hatfield JS, Kuhn DM. Differential tissue distribution of tryptophan hydroxylase isoforms 1 and 2 as revealed with monospecific antibodies. Brain Res. 2006;1085(1):11-18. Sari Y, Zhou FC. Serotonin and its transporter on proliferation of fetal heart cells. Int J Dev Neurosci. 2003;21:417-424. Sauer WH, Berlin JA, Kimmel SE. Effect of antidepressants and their relative affinity for the serotonin transporter on the risk of myocardial infarction. Circulation. 2003 Jul 8;108(1):32-36. Saxena PR, Villalon CM. 5-Hydroxytryptamine: a chameleon in the heart. Trends Pharmacol. Sci. 1991, 12: 223—227. Scanlon SM, Williams DC, Schloss P. Membrane cholesterol modulates serotonin transporter activity. Biochemistry. 2001 ;40(35):10507-10513. Scatton B, Duverger D, L'Heureux R, Serrano A, Fage D, Nowicki JP, MacKenzie ET. Neurochemical studies on the existence, origin and characteristics of the serotonergic innervation of small pial vessels. Brain Res. 1985;345:219—229. a. Schmid JA, Just H, Sitte HH. Impact of oligomerization on the function of the human serotonin receptor. Biochm Soc Trans. 2001;29:732-736. b. Schmid JA, Scholze, Kudlacek O, Freissmuth M, Singer EA, Sitte HH. Oligomerization of the human serotonin transporter and of the rat GABA 188 transporter 1 visualized by fluorescence resonance energy transfer microscopy in living cells. J Biol Chem. 2001;276; 6:3805-3810. Serebruany VL, O'Connor CM, Gurbel PA. Effect of selective serotonin reuptake inhibitors on platelets in patients with coronary artery disease. Am J Cardiol. 2001;87(12):1398-400.. J Biol Chem. 2001;276:3805-3810. Shingala JR, Balaraman R. Antihypertensive Effect of 5-HT(1A) Agonist Buspirone and 5-HT(ZB) Antagonists in Experimentally Induced Hypertension in Rats. Pharmacology. 2004;73(3):129-139. Slominski A, Pisarchik A, Semak I, Sweatman T, Szczesniewski A, Wortsman J. Serotoninergic system in hamster skin. J Invest Dennatol. 2002;119:934-942. Slominski A, Pisarchik A, Semak l, Sweatman T, Wortsman J. Characterization of the serotoninergic system in the C57BU6 mouse skin. Eur J Biochem. 2003;270(16):3335-3344. Small R, Macarak E, Fisher AB. Production of 5-hydroxyindoleacetic acid from serotonin by cultured endothelial cells. J Cell Physiol. 1977;90(2):225-231. Stier CT Jr, McKendall G, ltskovitz HD. Serotonin formation in nonblood-perfused rat kidneys. J Pharmacol Exp Ther. 1984;228(1):53-56. Stier CT, ltskovitz HD. Formation of serotonin by rat kidneys in vivo. Proc Soc. Exp. Biol. Med. 1985;180:550-557. Stott DJ, Saniabadi AR, Hosie J, Lowe GD, Ball S6. The effects of the 5-HT2 antagonist ritanserin on blood pressure and serotonin-induced platelet aggregation in patients with untreated essential hypertension. Eur J Clin Pharmacol. 1988, 35: 123-129. Sved AF, Van ltalli CM, Fernstrom JD. Studies on the antihypertensive action of L-tryptophan. J Pharmacol Exp Ther. 1982, 221: 329—333. Szabo C, Hardebo JE, mean C. An amplifying effect of exogenous and neurally stored 5-hydroxytryptamine on the neurogenic contraction in rat tail artery. Br J Pharmacol. 1991;102:401—407. Takahashi H, Hara K, Komiyama Y, Masuda M, Murakami T, Nishimura M, Nambu A, Yoshimura M. Mechanism of hypertension induced by chronic inhibition of nitric oxide in rats. Hypertens Res. 1995;18(4):319-324. Tamir H, Theoharides TC, Gershon MD, Askenase PW. Serotonin storage pools in basophil leukemia and mast cells: characterization of two types of serotonin 189 binding protein and radioautographic analysis of the intracellular distribution of [3H]serotonin. J Cell Biol. 1982;93(3):638-647. Taylor MA, Ayers CR, Gear AR. Platelet calcium and quenched-flow aggregation kinetics in essential hypertension. Hypertension. 1989;13(6 Pt 1):558-566. Tian RX, Kimura S and Kondou N Fujisawa Y, Zhou MS, Yoneyama H, Kosaka H, Rahman M, Nishiyama A, Abe Y. DOI, a 5-HT2 receptor agonist, induces renal vasodilation via nitric oxide in anesthetized dogs. Eur. J. Pharmacol. 2002,37: 79-84. Tierney AJ. Structure and function of invertebrate 5-HT receptors: a review. Comp Biochem Physiol A Mol Integr Physiol. 2001;128(4):791-804. Review. Tomita T, Umegaki K, Hayashi E. Hypoaggregability of washed platelets from stroke-prone spontaneously hypertensive rats (SHRSP). Stroke. 1984;15:70-75. Turla MB, Webb RC. Vascular responsiveness to 5-hydroxytryptamine in experimental hypertension. In The Peripheral Actions of 5-Hydroxytryptamine. Oxford University Press, Oxford.1989; p327—353. Umegaki K, lchikawa T. Vitamin E did not prevent platelet activation, but prevented increase of lipid peroxides and renal dysfunction in DOCA-salt hypertensive rats. J Nutr Sci Vitaminol (Tokyo) 1993;39:437-449. Vanhoutte PM. Platelet-derived serotonin, the endothelium, and cardiovascular disease. J. Cardiovasc. Pharmacol. 1991;17:86-812. van Schie DL, de Jeu RM, Steyn DW, Odendaal HJ, van Geijn HP. The optimal dosage of ketanserin for patients with severe hypertension in pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2002, 102: 161—166. Verhagen AM, Hohbach J, Joles JA, Braam B, Boer P, Koomans HA, Grone H. Unchanged cardiac angiotensin ll levels accompany losartan-sensitive cardiac injury due to nitric oxide synthase inhibition. Eur J Pharmacol. 2000;400(2- 3):239-247. Villalon CM, Heiligers JP, Centurion D, De Bries P and Saxena PR. Characterization of putative 5-HT7 receptors mediating tachycardia in the cat. Br. J. Pharmacol. 1997,121, 1187-1195. Villazon M, Padin JF, Cadavid MI, Enguix MJ, Tristan H, Orallo F, Loza Ml. Functional characterization of serotonin receptors in rat isolated aorta. Biol Pharm Bull. 2002;25(5):584-590. 190 Wainscott DB, Lucaites VL, Kursar JD, Baez M, Nelson DL. Phamiacologic characterization of the human 5-hydroxytryptamine2B receptor: evidence for species differences. J Pharmacol Exp Ther. 1996;276(2):720-727. Walther DJ, Bader MA. A unique central tryptophan hydroxylase isoform. Biochem. Pharmacol. 2003;66:1673-1680. a. Walther DJ, Peter JU, Bashammakh S, Hortnagl H, Voits M, Fink H, Bader M. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science. 2003;299(5603):76. b. Walther DJ, Peter JU, Winter S, Holtje M, Paulmann N, Grohmann M, Vowinckel J, Alamo-Bethencourt V, Wilhelm CS, Ahnert—Hilger G, Bader M. Serotonylation of small GTPases is a signal transduction pathway that triggers platelet alpha-granule release. Cell. 2003;115(7):851-862. Wanstall JC, Fiore SA Gambino A, Chess-VWliams R. Potentiation of 5- hydroxytryptamine (5-HT) responses by a 5-HT uptake inhibitor in pulmonary and systemic vessels: effevts of exposing rats to hypoxia. Naunyn Schmiedebergs Arch Pharmacol. 2003;368:520-527. Watanabe T, Pakala R, Katagiri T, Benedict CR. Serotonin potentiates angiotensin ll-induced vascular smooth muscle cell proliferation. Atherosclerosis. 2001 ;159:269—279. Watts SW. The development of enhanced arterial serotonergic hyperresponsiveness in mineralocorticoid hypertension. J Hypertens. 1998;16(6):811-822. Watts SW. 5-HT in systemic hypertension: foe, friend or fantasy? Clin Sci (Lond). 2005;108(5):399-412. Review. Watts SW. 5-Hydroxytryptamine-induced potentiation of endothelin-1- and norepinephrine-induced contraction is mitogen-activated protein kinase pathway dependent. Hypertension. 2000; 35(1 Pt 2):244-248. Watts SW, Baez M, Webb RC. The 5-hydroxytryptamine2B receptor and 5-HT receptor signal transduction in mesenteric arteries from deoxycorticosterone acetate-salt hypertensive rats. J Pharmacol Exp Ther. 1996;277(2):1103-1113. Watts SW, Fink GD. 5-HT2B-receptor antagonist LY-272015 is antihypertensive in DOCA-salt-hypertensive rats. Am J Physiol. 1999;276(3 Pt 2):H944-452. Weir EK, Reeve HL, Huang JM, Michelakis E, Nelson DP, Hampl V, Archer SL. Anorexic agents aminorex, fenfluramine, and dexfenfluramine inhibit potassium 191 current in rat pulmonary vascular smooth muscle and cause pulmonary vasoconstrction. Circulation. 1996;94:2216-2220. Wenting GJ, Woittiez AJ, Man in't Vel AJ, Schalekamp MA. 5-HT, a- adrenoceptors, and blood pressure. Effects of ketanserin in essential hypertension and autonomic insufficiency. Hypertension 1984, 6: 100—109. Woittiez AJ, Wenting GJ, Man in't Veld AJ, Boomsma F, Schalekamp MA. Ketanserin: a possible tool for studying the role of serotonin in hypertension. J. Cardiovasc. Pharmacol. 1995, 7 (Suppl. 7): $130—$136. Wolf C, Cai WJ, Vosschulte R, Koltai S, Mousavipour D, Scholz D, Afsah-Hedjri A, Schaper W, Schaper J. Vascular remodeling and altered protein expression during growth of coronary collateral arteries. J Mol Cell Cardiol. 1998;30(11):2291-2305. Wrona MZ, Dryhurst G. A putative metabolite of serotonin, tryptamine-4,5-dione, is an irreversible inhibitor of tryptophan hydroxylase: possible relevance to the serotonergic neurotoxicity of methamphetamine. Chem Res Toxicol. 2001;14(9):1184-1192. Yildiz O, Smith JR and Purdy RE. Serotonin and vasoconstrictor synergism. Life Sci. 1998, 62: 1723—1732. Zhang J, Lesort M, Guttmann R, Johnson G. Modulation of the in situ activity of tissue transglutaminase by calcium and GTP. J Biol Chem. 1998;273(4):2288- 2295. Zhu CB, Hewlett WA, Feoktistov I, Biaggioni I, Blakely RD. Adenosine receptor, protein kinase G, and p38 mitogen-activated protein kinase-dependent up- regulation of serotonin transporters involves both transporter trafficking and activation. Mol Pharmacol. 2004 ;65(6):1462-1474. Zhu CB, Carneiro AM, Dostmann WR, Hewlett WA, Blakely RD. p38 MAPK activation elevates serotonin transport activity via a trafficking-independent, protein phosphatase 2A-dependent process. J Biol Chem. 2005;280(16):15649- 15658. Zohar J, Westenberg HG. Anxiety disorders: a review of tricyclic antidepressants and selective serotonin reuptake inhibitors. Acta Psychiatr Scand Suppl. 2000;403:39-49. 192 BIBLIOGRAPHY Personal Data Name: Wei Ni B445 Life Science Building Born: 1/13/1978 Department of Pharmacology and Toxicology Michigan State University East Lansing, MI. 48824-1317 Phone number: (517) 353-3900 Fax number: (517) 353-8915 Educational Background 1996-2001 Peking University Health Science Center Beijing, P. R. China B.S. (Pharmacology) 2002-present Michigan State University, East Lansing, Michigan Doctoral candidate (Pharmacology and Toxicology) Teaching Activities 2005-2006 Lecturer for Pharmacology (PHM) 450 class Research Training 2001-2002 Hepatoprotective role of Ganoderma lucidum polysaccharide against BCG-induced immune liver injury in rats. Research with Dr. Zhibin Lin and Dr. Guoliang Zhang 2002-present The presence of a local serotonergic system in peripheral arteries. Doctoral thesis research with Dr. Stephanie W. Watts, Michigan State Unversity Membership in Academic Societies American Society of Pharmacology and Experimental Therapeutics American Heart Association American Physiological Society Society for Experimental Biology and Medicine Awards American Heart Association Predoctoral Fellowship-July,2004 to June,2006 ASPET Graduate Student Travel Award- Experimental Biology, 2005 ASPET Graduate Student Travel Award- Experimental Biology, 2006 MSU College of Veterinary Medicine Fellowship (Fall,2002; Fall,2003; Spring,2004; Fall,2004) MSU Graduate School Incentive Fellowship-2004 193 MSU Phi Zeta Research Day Best Poster Presentation by a Ph.D. Student- 2004 MSU the Graduate Office Fellowship- Fall, 2005 MSU the Graduate School Dissertation Completion Fellowship- 2006 Papers Zhang GL, Wang YH, Ni W, Teng HL and Lin ZB: Hepatoprotective role of Ganoderma lucidum polysaccharide against BOG-induced immune liver injury in mice. World J Gastroenterol. 2002 Aug; 8(4): 728-33. Ni W, Li MW, Thakali K, Fink GD and Watts SW: The fenfluramine metabolite (+)-norfenfluramine is vasoactive. J. Pharmacol. Exp. Ther., 2004, 309(2):845- 52. Ni W, Thompson JM, Northcott CA, Lookingland K and Watt SW: The serotonin transporter is present and functional in peripheral arterial smooth muscle. J Cardiovasc Pharmacol., 2004, 43(6):770-781. Ni W, Wilhelm CS, Bader M, Murphy DL, Lookingland K, Watts SW: (+)- Norfenfluramine-induced arterial contraction is not dependent on endogenous 5- hydroxytryptamine or 5-hydroxytryptamine transporter. J Pharmacol Exp Ther., 2005, 314(3):953-60. Ni W and Watt SW: 5-Hydroxytryptamine in the cardiovascular system: focus on the serotonin transport (SERT). In press, Clin. Exp. Pharmacol. Physiol, 2006, 33(7):575-83. Ni W, Lookingland, Watts SW: Arterial 5-HT transporter function is impaired in DOCA and LNNA but not spontaneously hypertensive rats. Hypertension. 2006, 48(1):134-40. Ni W, Lookingland K, Watts SW. Response to blood pressure in mutant rats lacking the 5-hydroxytryptamine transporter. Hypertension. 2006 Abstract Ni W, Zhan GL, Lu J, Teng HL, Bu XY, Lin ZB: Effect of ganoderma lucidum extracts on the immune liver injury induced by BCG and the possible mechanism. 3rd International Symposium on Hepatology, Hangzhou, China, October 2001 Thakali K, Ni W, Li MW, Fink GD and Watts SW: The anorexigen (+)- norfenfluramine as a pressor; enhanced response in hypertension. (Abstract) 57th Annual Fall Conference of the Conference of the Council for High Blood Pressure Research, Washington DC, September 2003, Hypertension, 42(3),425. 194 Ni W, Li MW and Watts SW: Dual role of 5-HT2A receptor and 5-HT transporter in (+)-norfenfluramine-induced aortic contraction. Experimental Biology Meeting, Washington DC, 2004. Ni W, Li MW, Thakali K, Fink GD and Watts SW: The fenfluramine metabolite (+)-norfenfluramine is vasoactive. Fund. Clin Pharmacol 18(S1):p.139,A 3.7,2004 Watts SW, Ni W, Northcot CA, Lookingland K, Thompson J.: The serotonin transporter in peripheral arteries. Fund. Clin Pharmacol 18(S1):p.139,A 3.7,2004 Serotonin club symposium-EPHAR Satellite in Porto 2004. Ni W, Wilhelm C, Bader M, Lookingland K, Watts SW: (+)-Norfenfluramine- induced arterial contraction is not dependent on 5-HT release: use of tryptophan hydroxylase (TPH) 1 deficent mice. 58th Annual Fall Conference of the Conference of the Council for High Blood Pressure Research, Chicago,lL, October 2004, Hypertension, 44(4), 518. Ni W and Watts SW: Peripheral arterial uptake and release of 5-HT. Experimental Biology Meeting, San Diego, 2005. Ni W, Lookingland K and Watts SW: 5-HT transporter (5-HTT) function is impaired in DOCA-salt hypertensive rats but not spontaneously hypertensive rats (SHR). 59th Annual Fall Conference of the Conference of the Council for High Blood Pressure Research, Washington DC, September, 2005, Hypertension, 46(4), 843. Ni W, Lookingland K and Watts SW: Peripheral arteries take up but do not concentrate 5-HT. Experimental Biology Meeting, San Francisco, 2006 NM, Szasz T, Lookingland K and Watts SW: The existence of a 5-HT synthetic and metabolism system in rat peripheral arteries. 60th Annual Fall Conference of the Conference of the Council for High Blood Pressure Research, San Antonio, TX, October, 2006, Hypertension, 48(4), e81. 195 lljllllljjllljjljjlllllllllgjl