a. a luv u 3...! . 3w u cw. .«flfléfimfifl «mgr: ufifini Janna 4w.“ Ravfiw. :. , ibfimw #4,: .w . $1va 1. V , "mm; 31.: .hfiultnmu I. dinnilsJHHWn 14 3 , . .mfiumnsnm M.“ www.mfi .cc .0}. .. Lhfinq a... 2x; .2. £an tufsis L‘BRAIEI tej / c/ ichigan . a WW7 M Universfiy This is to certify that the dissertation entitled CHARACTERIZATION OF DELTA-9- TETRAHYDROCANNABINOL-MEDIATED ALTERATIONS IN LEUKOCYTE AND PULMONARY AIRWAY EPITHELIAL CELL RESPONSES TO A PRIMARY CHALLENGE WITH INFLUENZA NPR/8/34 IN C57BL/6 WILD-TYPE AND CB1/CBZ RECEPTOR- NULL MICE presented by JOHN PHILIP BUCHWEITZ has been accepted towards fulfillment of the requirements for the Ph.D. degree in Pharmacology and Toxicology A WWT Major Professor’s Signature 3/2/ 0 7 Date MSU is an Affirmative Action/Equal Opportunity Institution 4 » .—-—.-—-.—.---I—¢-Q-.-_g-'-a-0---Q-O-O-g-C-c-O-o-n-a-n-n- a---0-o-I-O-I—l-D-O-.-,-l-.-.-n—~— 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 1 1 L1 j) 8 l l f1. v v NOV 1 (l 2008 6/07 p:/ClRC/DateDue.indd-p.1 CHARACTERIZATION OF DELTA-9-TETRAHYDROCANNABINOL-MEDIATED ALTERATIONS IN LEUKOCYTE AND PULMONARY AIRWAY EPITHELIAL CELL RESPONSES TO A PRIMARY CHALLENGE WITH INFLUENZA A/PR/8/34 IN C57BL/6 WILD-TYPE AND CB1/CB2 RECEPTOR-NULL MICE By John Philip Buchweitz 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 2007 ABSTRACT CHARACTERIZATION OF DELTA-9-TETRAHYDROCANNABINOL-MEDIATED ALTERATIONS IN LEUKOCYTE AND PULMONARY AIRWAY EPITHELIAL CELL RESPONSES TO A PRIMARY CHALLENGE WITH INFLUENZA A/PR/8/34 IN C57BL/6 WILD-TYPE AND CBl/CBZ RECEPTOR-NULL MICE By John Philip Buchweitz Delta-9-tetrahydrocannabinol (Ag-THC) is a well-established immunosuppressive agent derived from the plant Cannabis Sativa. The biological activity of Ag-THC is presumed to result from interactions of Ag-THC with the cannabinoid receptors CB; and C82. Ag-THC has been demonstrated to effect innate, cell-mediated, and humoral immune responses. Accordingly, A9-THC has been implicated as both a therapeutic agent toward unwarranted humoral immune responses and as a determinant of susceptibility toward infectious disease. Influenza is a common respiratory pathogen that affects millions of people world-wide each year. Viral clearance from the respiratory tract occurs through a combination of innate, cell-mediated, and humoral immune responses in the immunocompetent host. Therefore, it was hypothesized that the intrinsic immunosuppressive properties of Ag-THC could lead to decreased host resistance in mice resulting in a greater viral burden when compared with uninfected mice. It was further hypothesized that decreased host resistance to influenza would enhance morphologic features consistent with cellular injury and repair along the bronchiolar epithelium lining the airways of mouse lungs. Moreover, since clonal expansion of T cell populations critical to viral clearance are dependent on IL-2, and IL-2 expression is modulated by cannabinoids through a mechanism that is independent of CBl/CBZ receptors, the proposed increased susceptibility to influenza in the current studies might also arise from CB; and CB2 receptor-independent mechanisms. In the current studies, challenging mice with influenza alone resulted in robust inflammatory responses that were comprised of a diverse array of transient immune cells and their secretory chemical mediators in the airways. In addition to the inflammatory response, there were concomitant alterations in epithelial morphology that included cellular apoptosis and influenza virus-induced mucous cell metaplasia (MCM). Treatment of influenza infected mice with Ag-THC led to a dose-dependent increase in viral burden as compared to mice infected with influenza alone. More importantly, Ag-THC treated mice exhibited decreased recruitment of critical populations of CD4+ and CD8+ T cells and macrophages necessary for the clearance of influenza as compared to mice infected with influenza alone. Lastly, there were identifiable CB; and CB2 receptor-dependent and -independent mechanisms involved in host immune responses to influenza infection. More specifically, the profile of viral burden included an increase in H1 mRNA amongst mice treated with A9-THC that was similar between CBf”/CB2'/' and wild type mice suggesting a CB; and CB2 receptor- independent finding. Conversely, the magnitude of viral burden was strikingly less in CBl'/’/CBz"‘ mice than in wild type mice which suggested the involvement of CB; and/or CB; receptors in mediating immune homeostasis. In addition to viral burden, there were other unique findings exhibited by CBl'/'/CB2'/' mice in response to Ag-THC treatment alone as compared to Ag-THC treated wild type mice. Most notably, Ag-THC induced MCM of the bronchiolar epithelium independent of PR8 challenge in CBl'/'/CBz'/' mice. In conclusion, these findings suggest that Ag-THC is a determinant of susceptibility to increased viral burden and that alternative receptors for Ag-THC are, in part, responsible. This dissertation is dedicated to the men and women of our armed services who have fought so bravely in the defense of my family’s freedoms. God Bless all of you. iv ACKNOWLEDGMENTS First, I would like to extend my gratitude to my mentor, Norbert E. Kaminski, whose guidance and encouragement has not gone unnoticed in my tireless pursuit of the doctorate degree. In addition, I would like to thank my committee members, Dr. Alex Chen, Dr. Jane Maddox, and Dr. Jack Harkema for their support and assistance. My research was made all the more enjoyable by the camaraderie of my fellow colleagues including Dr. Courtney Sulentic, Dr. Cheryl Rockwell, Dr. gautham Rao, Dr. Jim Wagner, Dr. Aimen Farraj, Dr. Ammie Bachman, Dr. Barb Faubert Kaplan, Dr. Steve Carey, Dina Schneider, Colin North, and Peer Karmaus. The research was also made possible by the valuable assistance and skills of Lori Bramble, Bob Crawford, Kimberly Hambleton and the histology group (Amy, Joselyn, Kathy, and Rick)... you have all been a blessing. Lastly, I would like to thank my family, Cynthia, Nathan, Olivia, for being supportive and making each day more fulfilling. TABLE OF CONTENTS LIST OF TABLES ................................................................................................... LIST OF FIGURES ................................................................................................. LIST OF ABBREVIATIONS ................................................................................. INTRODUCTION ................................................................................................... I. II. III. Respiratory toxicology ................................................................................. A) Basic anatomy and physiology ............................................................... B) Respiratory clearance of inhaled particles and pathogens ...................... 1. Mucociliary clearance ...................................................................... 2. Alveolar clearance ............................................................................ C) Epithelial cell death and regeneration .................................................... 1. Clara cells ......................................................................................... 2. Type II cells ..................................................................................... Influenza Virus ............................................................................................. A) Classification .......................................................................................... B) Viral Entry .............................................................................................. C) Viral Clearance ....................................................................................... D) Chemokine and cytokine responses to influenza virus ........................... E) Models of susceptibility ........................................................................... Cannabinoids ................................................................................................ A) Chemistry of delta-(9)-tetrahydrocannabinol ......................................... B) Pharmacokinetics .................................................................................... 1. Absorption ........................................................................................ 2. Distribution ....................................................................................... 3. Metabolism ....................................................................................... C) Biological receptors and activity ............................................................ 1. CB1 and CB2 receptors .................................................................... 2. Alternative receptors ........................................................................ D) Effects of cannabinoids on the immune system ..................................... 13) 1. Effects on macrophage function ....................................................... 2. Effects on neutrophil function .......................................................... 3. Effects on natural killer cell function ............................................... 4. Effects on THI cells .......................................................................... 5. Effects on CD8+ T cells .................................................................... 6. Effects on immune cell-derived chemokines and cytokines .............. Models of host-resistance ....................................................................... 1. Model of A9-THC and Listeria monocytogenes ............................... vi X xi xiv OOVGM-h-bt—‘t—t —‘ p—A wt-‘OOOOOO p—i 15 15 15 16 16 18 18 18 19 2O 20 21 22 23 24 24 25 25 2. Model of Ag-THC and Herpes Simplex Virus ................................. 3. Model of Ag-THC and Legionella pneumophila .............................. IV. Objectives ..................................................................................................... MATERIALS AND METHODS ............................................................................ I. Cannabinoid compounds ...................................................................... 11. Animals ................................................................................................ III. Experimental designs ........................................................................... IV. Influenza A/PR/8/34 instillation .......................................................... V. Steady state blood serum levels of Ag-THC and its metabolites ........... VI. Necropsy, lavage collection, and tissue preparation ............................. VII. Immunocytochemistry ......................................................................... VIII. Total and PCNA positve epithelial numeric cell density and labeling index ........................................................................................................ IX. CAS-3 and alcian blue numeric cell densities ...................................... X. Morphometry of stored intraepithelial mucosubstances ...................... XI. Bronchoalveolar lavage cellularity ...................................................... XII. Histopathology scores for inflammation .............................................. XIII. Total protein ......................................................................................... XIV. Neutrophil elastase ............................................................................... XV. Inflammatory and THI/TH2 cytokines ................................................ XVI. T-lymphocyte flow cytometry .............................................................. XVII. RNA isolation ...................................................................................... XVIII. Statistical Analysis ............................................................................... EXPERIMENTAL RESULTS .............................................................. 1. Model developmentzEstablishing the dose ................................................... II. A) Concentration-dependent inflammatory responses to PR8 in pulmonary airways .................................................................................................... B) Steady state blood serum levels of A9-THC and its metabolites ............ Time-dependent airway epithelial and inflammatory cell responses induced by influenza virus A/PR/8/34 in C57BL/6 mice ............................. A) PR8 induces time-dependent alterations in epithelial morphology .......... B) Temporal analysis of stored intraepithelial mucosubstances following PR8 challenge ...................................................................... C) Expression levels of whole lung MUCSAC mRNA at 7 days post Infection D) Time-dependent differences in total and PCNA immunopositive epithelial numeric cell densrties E) Bronchoalveolar lavage fluid analysis .................................................... vii 26 27 29 30 30 3O 30 31 31 31 34 34 34 35 36 36 37 37 38 39 39 40 41 41 41 41 45 45 49 49 49 54 1. Protein ............................................................................................... 54 2. Total and differential inflammatory cell counts ................................ 54 3. inflammatory chemokines and cytokines .......................................... 54 4. TH2 cytokines IL-4, IL-5, IL-9 and IL-13 ........................................ 58 5. Elastase .............................................................................................. 58 III. Modulation of airway responses to influenza A/PR/8/34 by A9- tetrahydrocannabinol in C57BL/6 mice ......................................................... 62 A) Ag-THC increases whole lung H1 mRNA following PR8 challenge... 62 B) A 9-THC does not affect total protein levels in BALFM ........................... 62 C) A 9-THC decreases leukocyte populations retrieved in BALF ................ 62 D) Ag-THC decreases CD4+ and CD8+ T cells in BALF ............................ 66 E) Ag-THC modestly affects chemokines and cytokines in retrieved in BALF ...................................................................................................... 66 F) A9—THC treatment reduces inflammation scores for mouse lung sections ............................................................................ 70 G) Effect of A9-THC on the observed pulmonary histopathology to PR8.... 70 H) Caspase-3 mRNA expression levels and G5 numeric cell densities ....... 75 I) MUCSAC mRNA expression levels and G5 numeric cell densities ....... 76 J) A9-THC modestly enhances neutrophil-derived elastase levels in IV. Pulmonary airway responses to influenza A/PR/8/34 in CB1'/'/CB;'/' mice exposed to A9-tetrahydrocannabinol: An examination for the role of cannabinoid receptors .................................................................................... 81 A) Qualitative health assessment ................................................................. 81 B) The viral load of PR8 in the pulmonary airways of CB.'/'/CB;' ' and wild type mice measured 7 days after challenge ............................. 83 C) A 9-THC affects vascular permeability induced by Pr8 infection of the airways in CB] "/CB;' and wild 9type mice ............................................. 83 D) CB1 / "/CB;' mice treated with A 9-THC exhibit a distinctly different composition of leukocytes recruited to the airways in response to PR8 infection when compared to wild type mice. . . . . . .. ................................. 85 E) BALF-associated CD4+ and CD8+ T cell levels following PR8 challenge in CB|'/'/CB;'/° and wild type mice .......................................... 90 F) Cannabinoid receptor deficient mice exhibit unique differences in epithelial and leukocytic Chemokine and cytokine secretion in the pulmonary airways... 93 G) Cytokines and chemokines detected 1n blood serum .............................. 98 H) The absence of cannabinoid receptors CB. and CB; enhances the observed pulmonary histopathology ....................................................... 101 I) A9-THC affects the magnitude of the inflammatory response to PR8 1n wild type and CB; and CB; deficient mice .............................................. 104 J) Effects of C81 and CB; deficiency on the numeric cell densities of apoptotic cells and metaplastic goblet cells ............................................. 104 viii DISCUSSION .......................................................................................................... 109 1. Animals utilized in these studies ................................................................ 109 II. The method of delivery and the dose of PR8 utilized .. ............................. 109 III. The method of delivery and dose of A9-THC utilized in these studies ..... 109 IV. Effect of A9-THC on viral H1 mRNA levels in wild type and CBI'I' /CB;'/' mice ..................................................................................................... 1 l 1 V. Effect of PR8 on inflammatory cell recruitment to the pulmonary airways in the presence or absence of A9-THC in wild type and CBl'/' /CB;'/'mice ..................................................................................................... l 13 VI. Effect of PR8 on the release of soluble mediators into the pulmonary airways in the presence or absence of A9-THC in wild type and CBI'I' /CB;"'mice ..................................................................................................... 1 15 VII. Effect of a single PR8 instillation on pulmonary histopathology in the presence or absence of Ag-THC in wild type and CB1'/'/CB;"' mice ............ 119 VIII. Effect of a single PR8 instillation on epithelial cell apoptosis and MCM in the presence or absence of A9-THC in wild type and CB1'/'/CB;'/' mice.. 121 IX. Significance and relevance ....................................................................... 123 X. LITERATURE CITED ............................................................................................. 128 APPENDIX .............................................................................................................. 140 ix LIST OF TABLES . Analysis of A9-THC, 9-COOH-A9-THC, and ll-OH- A9-THC concentrations in mouse blood serum ............................................................ 44 . Total and differential cell counts from BALF 67 . CD4+ and CD8+ T cell populations observed in BALF by flow cytometry ................................................................................. 68 . Inflammatory cytokines quantified in BALF by cytometric bead array ....................................................................................... 69 . Summary of the effects of corn oil and A9-THC on BALF and histochemistry measurements taken for epithelial and inflammatory cells in wild type and CB;"’/CB;'/' mice ................................................ 110 10. 11. 12. 13. 14. 15. 16. LIST OF FIGURES Diagram of human (A) and mouse (B) lungs ........................... Structure of A9-tetrahydrocannabinol .................................... Diagram of the microdissection of mouse right lung lobes (R1, R2, R3, R4) and left lung lobe at generations 5 (G5) and 11 (G11) ...... Increasing concentrations of influenza virus result in increasing inflammatory responses in the hilar region of the left lung lobe at generation 5 of the main axial airway .................................... Bronchiolar epithelial morphometry following influenza infection. Time-to-onset of mucous cell metaplasia following influenza infection ...................................................................... MU C5AC gene induction by influenza at 7 days post infection. . . Time-dependent proliferation of epithelial cells lining the main axial airway following influenza infection .............................. Time-dependent detection of total protein in bronchoalveolar lavage fluid following influenza infection ............................... Time-dependent recruitment of leukocytes to the pulmonary airways following influenza infection .................................... Secretion of chemokines and cytokines into the airways in response to influenza infection ............................................ Detection of TH2 cytokines secreted into the airways in response to influenza infection .......................................................... Secretion of neutrophil-derived elastase into the airways in response to influenza infection ............................................ A9-THC treatment enhances hemagluttinin 1 mRN A levels .......... A9-THC does not alter total protein levels in BALF ................... A9-THC decreases inflammation scores in a time-dependent manner ........................................................................ xi 17 33 43 47 50 51 53 55 57 60 61 63 64 65 72 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. Examination of the effects of Ag-THC on the inflammatory response to influenza challenge by histopathology .................... 74 A9-THC has no effect on Caspase-3 mRNA levels and epithelial numeric cell densities ...................................................... 78 A9-THC increases MUCSAC mRNA levels and decreases epithelial numeric cell densities for acidic mucosubstances .......... 80 A9-THC modestly increases neutrophil-derived elastase levels secreted in the pulmonary airways ....................................... 82 Effects of A9-THC treatment on the expression levels of H1 mRNA in CB.""/CB;'/' and wild type mice ....................................... 84 Effects of A9-THC treatment on total protein in BALF in CB1" ’ /CBz"' and wild type mice ................................................. 86 Effects of A9-THC treatment on total and differential leukocyte counts in BALF in CB1'/'/CB;‘/' and wild type mice ......................... 89 Effects of A9-THC treatment on CD4)r and CD8+ T cells retrieved in BALF in CB1'/'/CB;"' and wild type mice ............................ 92 Effects of A9-THC treatment on inflammatory chemokines and cytokines in secreted into the pulmonary airways in CB1'/'/CB;'/' and wild type mice .......................................................... 95 Effects of Ag-THC treatment on TH2 cytokines secreted into the pulmonary airways in CBt'/'/CB;'/' and wild type mice ................ 97 Effects of A9-THC treatment on inflammatory chemokines and cytokines released into blood serum in CBl'/'/CB;'/' and wild type mice ........................................................................... 100 Inflammatory res onse to PR8 in the proximal section of the left lung lobe in CB1' '/CB;'/' and wild type mice ........................... 103 Effects of A9-THC treatment on histopathology-based inflammation scores in CB1’/'/CB;'/' and wild type mice ............. 105 Effects of Aq-THC treatment on the numeric cell density of caspase-3 positive epithelial cells in CB,'/'/CB;"' and wild type mice ........................................................................... 1 06 xii 31. Effects of A9-THC treatment on the numeric cell density of mucosubstances in airway epithelial cells in CB1'/’/CB;'/' and wild type mice ..................................................................... 108 xiii AB/H AB/PAS ABS AIDS ANOVA AP-l B-l 13-2 BALF BCA CAS-3 CBl CB1" CB; CB;'/' CBi"'/CB;"’ CD cDNA C/EBPB CNS CO CTL LIST OF ABBREVIATIONS Alcian blue/hematoxylin Alcian blue/periodic acid schiff Automation buffer solution Acquired Immune Deficiency Syndrome Analysis of variance Activating protein-1 Non-antigen stimulated B cell Antigen stimulated B cell Bronchoalveolar lavage fluid Bicinchoninic acid Caspase-3 Cannabinoid receptor type 1 Cannabinoid receptor type 1 null Cannabinoid receptor type 2 Cannabinoid receptor type 2 null Cannabinoid receptors type 1 and 2 null Cluster of differentiation copy-deoxynucleic acid Nuclear factor-IL-6 Central nervous system Corn oil Cytotoxic T lymphocytes xiv AKTHC DNA dpi F ACS F asL FCM GS G11 H&E HSVl HSV2 icv IFN or/[i IFN-y IL IP-10 IRF KO LAK LD50 M1 MCM Delta-9-tetrahydrocannabinol Deoxynucleic acid Days post-infection Flourescence assisted cell sorting F as ligand Flow cytometer Airway generation 5 Airway generation 11 Hemagglutinin Hematoxylin and eosin Herpes simplex virus type 1 Herpes simplex virus type 2 intracerebroventricularly Interferons -alpha and -beta Interferon-gamma Interleukin Interferon-gamma—inducible protein Interferon regulatory factors Knockout Lymphocyte activated killer cells Lethal dose to 50 percent of the population Matrix protein 1 Mucous cell metaplasia XV MCP-l MCP-3 MDCK MHC MIP 1 -0l MIP 1 -|3 MIP-2 MIP3-a mRNA NFAT NF -1 v R3 \ 611% ‘i\ Figure 3. Diagram of the microdissection of mouse right lung lobes (R1, R2, R3, R4) and left lung lobes at generations 5 (G5) and 11 (G11). 33 intraepithelial mucosubstances. VII. Immunocytochemistry. Hydrated paraffin sections (5—6 um thick) from formalin-fixed lung tissues were treated with 0.05% proteinase K for 2 min and washed with l N HCI for 1 h. Sections were then treated with 3% H;0; (in methanol) to block endogenous peroxide and were incubated with a monoclonal antibody (PC10) cocktail to PCNA (Biogenex, San Ramon, CA) consisting of the primary antibody to PCNA 1:50, a secondary antibody to Immunoglobulin 1:500 and mouse serum 1:50 for 1 h. Immunoreactive PCNA was visualized with the Vectastain Elite ABC kit (Vectastain Laboratories Inc., Burlingame, CA) using 3‘,3‘-diaminobenzidine (DAB) tetrahydrochloride (Sigma Chemical Co., St. Louis, MO) as a chromagen. VIII. Total and PCNA positive epithelial numeric cell density and labeling index. The total number of epithelia lining the luminal surface of the main axial airway at generation 5 was enumerated per length of basal lamina. Likewise, cells with nuclei staining positive for PCNA were also enumerated per length of basal lamina. A labeling index for PCNA was determined by dividing the number of PCNA positive cells per unit length of basal lamina by the total number of epithelial cells per unit length of basal lamina. IX. CAS-3 and alcian blue numeric cell densities. Slides of lung sections either stained immunohistochemically for CAS-3 or stained for alcian blue (acidic mucosubstances) were examined. Numeric cell densities were determined for epithelial cells 34 immunohistochemically reactive to CAS-3 via light microscopy by counting the number of nuclear profiles of these immunoreactive epithelial cells lining the bronchiolar epithelium at generation 5 and dividing by the length of the underlying basal lamina. Numeric cell densities for CAS-3 were expressed as the number of immunoreactive cells per mm basal lamina. In a Similar manner, numeric cell densities were determined for epithelial cells staining with alcian blue (acidic mucosubstances). The numeric cell density of epithelial cells staining for alcian blue (acidic mucosubstances) was expressed as the number of alcian blue reactive epithelial cells per mm basal lamina. X. Morphometry of stored intraepithelial mucosubstances. The volume density (Vs) of AB/PAS-Stained mucosubstances in the respiratory epithelium lining the main axial airway at 3, 7, 10, 15, and 21 dpi was quantified using computerized image analysis and standard morphometric techniques. The area of AB/PAS stained mucosubstance was calculated from the automatically circumscribed perimeter of stained material using a Power Macintosh 7100/66 computer and the public domain NIH Image program (written by Wayne Rasband, US. National Institutes of Health and available on the Internet at http://rsb.info.nih.gov/nih-image/l The length of the basal lamina underlying the surface epithelium was calculated from the contour length of the digitized image of the basal lamina. The volume of stored mucosubstances per unit of surface area of epithelial basal lamina was estimated using the method described in detail by Harkema et al., (1987) (112). The Vs of intraepithelial 2 mucosubstances was expressed as nanoliters of intraepithelial mucosubstances per mm of basal lamina. 35 XI. Bronchoalveolar lavage cellularity. The total number of leukocytes in BALF was counted with a hemacytometer. In brief, 10 ul of BALF was added to a hemacytometer and the number of leukocytes in the four etched comer quadrants were counted and multiplied by 2500 to yield the number of leukocytes per ml of sample. The percent of total leukocytes consisting of eosinophils, lymphocytes, macrophages, and neutrophils were determined from counts of 200 cells in a cytospin sample stained with Diff-Quick (Dade Behring, Newark, DE). The percentage of eosinophils, lymphocytes, macrophages, and neutrophils were multiplied by the total number of leukocytes determined from the hemacytometer to yield the respective number of each cell type per ml of sample. XH. Histopathology scores for inflammation. A histopathologic score was established based upon the numbers and distribution of inflammatory cells within the tissues, as well as non-inflammatory changes such as evidence of bronchiolar epithelial injury and repair. The scores assigned were: 0 = no inflammation, 1 = mild, inflammatory cell infiltrate of the perivascular/peribronchiolar compartment, 2 = moderate, inflammatory cell infiltrate of the perivascular/peribronchiolar space with modest extension into the alveolar parenchyma, and 3 = severe, inflammatory cell infiltrate of the perivascular/peribronchiolar space with a greater magnitude of inflammatory foci found in the alveolar parenchyma. A certified pathologist scored each lung section independently and a mean score with the standard error of the mean was calculated for each treatment group. 36 XIII. Total protein. Total protein was quantified in BALF using the bicinchoninic acid (BCA) method as provided by the manufacturer (Pierce, Rockford, IL). In brief, BALF samples were centrifuged at 600 rpm for 5 min to pellet cellular debris. Supernatants were removed and stored at -20°C until testing was performed. 25 31 of BALF supernatant or bovine serum albumin protein standard (25-2000 [jg/ml) was incubated in a 96-well microplate with 200 131 of a 50:1 mixture of reagents A (sodium carbonate, sodium bicarbonate, bicinchoninic acid and sodium taltrate in 0.1M sodium hydroxide) and B (4% cupric sulfate) for 30 minutes at 37°C. Samples were read on a Biotek microplate reader at 562 nm. XIV. Neutrophil elastase. Airway elastase recovered in BALF was determined by an ELISA for elastase using a rabbit monoclonal antibody to the human elastase (Calbiochem, La Jolla, CA). 50 ul aliquots of BALF were applied to a 96-well microtiter plate (Microfluor 2 Black, Dynex Technologies, Chantilly, VA) and dried overnight at 40°C. Plates were blocked with a solution of 1.5% goat serum in automation buffer solution (ABS, pH 7.5; Biomeda Corp., Foster City, CA) for 30 min at 37°C. Plates were then incubated with anti-elastase antibody (1:400 in ABS containing 1.5% goat serum) for l h at 37°C and then washed three times with ABS. Bound primary antibody was detected with a biotinylated goat anti-rabbit secondary antibody and quantitated using horseradiSh-peroxidase-conjugated avidin/biotin complex (ABC Reagent; Vector Laboratories, Burlingame, CA) and a fluorescent substrate (QuantaBlue; Pierce Chemical, Rockford, IL) using a fluorescence 37 microplate reader (SpectraMax Gemini; Molecular Devices; 318 nm excitation/410 nm emission). Readings were taken at 3 min intervals for 24 min. Duplicate samples were averaged and the group data is represented as mean Vmax units/S. XV. Inflammatory and T;;1/T;;2 cytokines. BALF samples were centrifuged at 600 rpm for 5 min to pellet cellular debris. Supernatants were removed and stored at -20°C until testing was performed. The cytokines IL-6, IL-10, MCP-l, IFN-y, TNF-a, and IL- l2p70 were detected Simultaneously by using the cytometric bead array (CBA) mouse inflammation kit (BD Pharmingen, San Diego, CA) and the THl/I‘H2 cytokines IL-2, IL- 4, IL-5, IFN-y, and TNF-a were detected simultaneously by using the mouse THl/THZ kit. In brief, 50 ul of BALF or serum from each sample was incubated individually with a mixture of capture beads and 50 ul of PE detection reagent consisting of PE-conjugated anti-mouse IL-6, IL-10, MCP-l, IFN-y, TNF-a, and IL-12p70 or 50 ul of PE detection reagent consisting of PE-conjugated anti-mouse IL-2, IL-4, IL-5, IFN-y, and TNF-a. The samples were incubated at room temperature for 2 h in the dark. After incubation, samples were washed once and resuspended in 300 pl of wash buffer before acquisition on a BD FACSCalibur flow cytometer. Data were analyzed using CBA software (BD Pharmingen, San Diego, CA). Standard curves were generated for each cytokine using the mixed cytokine standard provided with the respective kits. The concentration of each cytokine was determined by interpolation from the corresponding standard curve. The range of detection was 20 — 5000 pg/ml for each cytokine measured. Serum was analyzed for inflammatory cytokines only. In a similar manner, the cytokines IL-9 and IL-13 were analyzed using BD flex- 38 sets. Samples and standards were analyzed according to manufacturer-based instructions. XVI. T-Iymphocyte flow cytometry. After enumerating the retrieved cells in BALF, the cells were pelleted by centrifugation and reconstituted in 150 pl flow cytometer (FCM) buffer (PBS supplemented with 2% (w/v) bovine serum albumin and 0.09% (w/v) sodium azide) with purified anti-mouse CD16/CD32 (Fey III/II Receptor) antibody (BD Biosciences, San Jose, CA) to block for 30 min. The samples were then Split into two groups of equal volume, washed and reconstituted in FCM buffer. One group received antibodies for CD3 (APC anti-mouse CD38), CD4 [PE anti-mouse CD4(L3T4)] and CD8 [FITC anti-mouse CD8a(Ly-2)], while the other group received the cognate isotype control antibodies. Samples were allowed to incubate l h at 4°C, washed twice and then fixed for 10 min. with Cytofix. Samples were then washed again and reconstituted to a volume of 300 pl for analysis on the BD FACSCalibur flow cytometer. The total number of events taken per each sample was 10,000. XVII. RNA isolation. Total RNA was isolated from the lung lobes by using the TRI-reagent method (Sigma Chemical, St Louis, MO). The evaluation of the relative expression levels of H1, Caspase-3 and MUCSAC messenger ribonucleic acid (mRNA) were determined using the TaqMan real-time multiplex reverse transcriptase-polymerase chain reaction (RT-PCR) with custom designed TaqMan primers and probe to the target gene and the manufacturer’s pre-developed primers and probe to 183 (Applied Biosystems, Foster 39 City, CA). The primers and probe to both the target gene and endogenous reference gene were Specifically designed to exclude detection of genomic deoxynucleic acid (DNA). Aliquots of isolated tissue RNA (1 ug total RNA) were converted to copy DNA (cDNA) using random primers. The resultant cDNA (2 pl) was added to a reaction mixture that consisted of the target gene primers and probe, endogenous reference primers and probe (188 ribosomal RNA), and Taqman universal master mix to a final volume of 30 ul. Following PCR, amplification plots (change in dye fluorescence versus cycle number) were examined and a dye fluorescence threshold within the exponential phase of the reaction was set separately for the target gene and the endogenous reference (188). The cycle number at which each amplified product crosses the set threshold represents the CT value. The amount of target gene normalized to its endogenous reference was calculated by subtracting the endogenous reference C; from the target gene CT (ACT). Relative mRNA expression was calculated by subtracting the mean AC; of the control samples from the AC; of the treated samples (AACT). The amount of target mRNA, normalized to the endogenous reference and relative to the calibrator (i.e., RNA from control) is calculated by using the formula Z‘MCT. XVIII. Statistical Analysis. Data are expressed as mean is standard error of the mean (SEM). The differences between treatment groups were determined by either a Student's t-test, one-way or two- way analysis of variance (ANOVA) with multiple comparisons made by the Student- Newman-Keuls post hoc test using SigmaStat software from Jandel Scientific (San Rafael, CA). The criterion for significance was taken to be p <0.05. 40 EXPERIMENTAL RESULTS 1. Model development: Establishing the dose. A) Concentration-dependent inflammatory responses to PR8 in pulmonary airways The inflammatory response to PR8 was confined to the hilar region of the transverse left lung lobe sections (Figure 4). The inflammatory response was most notable at generation 5 with occasional evidence of inflammation observed at generation 11. The magnitude and severity of the inflammatory response was concentration- dependent, increasing with increasing concentrations of PR8. The inflammatory response consisted primarily of mononuclear cells that included lymphocytes, monocytes/macrophages, and neutrophils. B) Steady state blood serum levels of A9-THC and its metabolites. The steady state blood serum levels for A9-THC and its primary metabolites, 9- COOH-Ag-THC, and ll-OH-A9-THC were determined on the fifth day of treatment, 4 h after the final dose of Ag-THC (Table 1). At a dose of 5 mg/kg A9-THC, blood serum levels reached a mean concentration of 84.6 ng/ml Ag-THC. In addition, there was evidence of modest levels of the 9-COOH-A9-THC metabolite present in blood serum with no detectable levels of the ll-OH-A9-THC metabolite. At a dose of 75 mg/kg A9- THC, the mean blood serum levels for the parent compound were similar to those observed with the 5 mg/kg A9-THC dose. However, levels of the 9-COOH-A9-THC metabolite in blood serum were augmented when compared to the blood serum levels for mice receiving 5 mg/kg A9-THC with concentrations reaching 446 ng/ml 9-COOH-A9- 41 Figure 4. Increasing concentrations of influenza virus result in increasing inflammatory responses in the hilar region of the left lung lobe at generation 5 of the main axial airway. Light photomicrographs of the respiratory epithelium lining the luminal surface of the main axial airway (MAA) (generation 5) from mice intranasally instilled with PR8 at concentrations of 50 pfu (A), 300 pfu (B), and 500 pfu (C) at 13 days post infection. There was marked inflammatory cell infiltration of the perivascular/peribronchiolar submucosa (sm) extending into the alveolar parenchyma (p) in all of the sections representing each concentration of virus. There was a concentration- dependent increase in the magnitude and severity of the inflammatory response observed. Artery = (a). bar = 100 microns. Images in this dissertation are presented in color. 42 Figure 4. Increasing concentrations of influenza virus result in increasing inflammatory responses in the hilar region of the left lung lobe at generation 5 of the main axial airway. 43 Sample # A9-THC 9-coon-A9- ll-OH-Ag-THC Vehicle (ng/ml) THC (ng/ml) (ng/ml) 1 3.6 ND ND 2 2.5 ND ND 3 3.1 ND ND 4 42 ND ND mean 3.4 Sample # A9-THC 9-COOH-A9- ll-oH-A9-THC 5mg/kg THC (ng/ml) THC (ng/ml) (rig/ml) 1 55.2 6.8 ND 2 48.4 24.6 ND 3 133.5 30.0 ND 4 101.4 41.0 ND mean 84.6 25.6 Sample # A9-Tnc 9-cooH-A9- 11-0H-A9-Tllc 75mg/kg THC (ng/ml) THC (ng/ml) (ng/ml) 1 29.9 136.8 2.7 2 64.5 873.3 193 3 117.9 317.0 10.1 4 52.4 459.0 9.9 mean 66.2 446.5 10.5 Table 1. Analysis of A9-THC, 9-C00H-A9-THC, and ll-OH-A9-THC concentrations in mouse blood serum. C57BL/6 mice were treated with vehicle (corn oil), 5 mg/kg or 75 mg/kg A9-THC for 5 consecutive days. Four hours after the last treatment, mice were anesthetized and whole blood was collected by cardiac puncture. A9-THC and its metabolites were chemically extracted from whole blood and analyzed by GC/MS. ND = not detected. 44 THC. Lastly, the ll-OH-Ag-THC metabolite was modestly elevated with the 75 mg/kg A9-THC treatment group with concentrations averaging 10 ng/ml. H. Time-dependent airway epithelial and inflammatory cell responses induced by influenza virus A/PR/8/34 in C57BL/6 mice. A) PR8 induces time-dependent alterations in epithelial morphology No microscopic alterations were present in the examined proximal (at GS of the main axial airway) and distal (at G11 of the main axial airway) tissue sections from the left lung lobe of mice that were intranasally instilled with vehicle alone (controls) (Figure 5A). The principal pulmonary alteration in mice intranasally instilled with influenza virus and sacrificed 3 dpi (Figure 5 A-D) was an acute necrotizing bronchiolitis characterized by multiple focal areas of necrosis and luminal shedding (exfoliation) of the surface epithelial cells lining the main axial airway (Figure 5C) and smaller diameter pre- terminal and occasional terminal bronchioles. Similar but much less severe lesions were observed in the distal tissue section of some of these exposed mice. Airway epithelial lesions were accompanied by a mild intramural inflammatory cell infiltrate composed principally of mononuclear cells (lymphocytes and monocytes) and lesser numbers of neutrophils. A Similar inflammatory cell infiltrate was present in adjacent peribronchiolar and perivascular regions. Virus instilled mice that were sacrificed 7 dpi had a marked bronchiolitis and alveolitis again restricted mainly to the hilar region of the lung lobe (proximal tissue section). Necrosis and exfoliation of the bronchiolar ciliated epithelium observed at 3 dpi was replaced by a hyperplastic/hypertrophic, nonciliated, cuboidal and basophilic 45 Figure 5. Bronchiolar epithelial morphometry following influenza infection. Light photomicrograph of the respiratory epithelium (e) lining the luminal surface of the main axial airway (generation 5) from mice intranasally instilled with SAL (A, B, E, F, I, J) and PR8 (C, D, G, H, K, L) at 3, 10, and 21 dpi, respectively. A = H&E; SAL instilled mouse 3 dpi with no alterations to the respiratory epithelium lining the airway or to the peribronchiolar tissue (a). B = AB/PAS; SAL instilled mouse 3 dpi with no microscopic evidence of MCM. C = H&E; PR8 instilled mouse 3 dpi with degenerative respiratory epithelium lining the airway and peribronchiolar inflammatory cell infiltrate in the submucosa (sm). D = AB/PAS; PR8 instilled mouse 3 dpi with exfoliation of the epithelium lining the luminal surface of the main axial airway and no microscopic evidence of MCM. E = H&E; SAL instilled mouse 10 dpi with no alterations to the respiratory epithelium lining the airway or to the peribronchiolar tissue. F = AB/PAS; SAL instilled mouse 10 dpi with no microscopic evidence of MCM. G = H&E; PR8 instilled mouse 10 dpi with regenerative epithelium and mucous cell metaplasia (arrows) with marked peribronchiolar inflammatory cell infiltrate (asterisk). H = AB/PAS; PR8 instilled mouse 10 dpi with marked accumulation of mucous-containing epithelial cells. I = H&E; SAL instilled mouse 21 dpi with no alterations to the respiratory epithelium lining the airway or to the peribronchiolar tissue. J = AB/PAS; SAL instilled mouse 21 dpi with no microscopic evidence of MCM. K = H&E; PR8 instilled mouse 21 dpi with cuboidal epithelium and mucous cell metaplasia with decreased peribronchiolar inflammation. L = AB/PAS; PR8 instilled mouse 21 dpi with marked accumulation of mucous-containing epithelial cells lining the airway. Bar = 50 microns. Images in this dissertation are presented in color. 46 ' .\ e a; #19- - ‘ V sm Quad} f J‘EEI' ..x- 74;: ..- < 3;}:“2; . ' '- ' ”a 5? a:- ,. ”lei? .‘t : C: 4' "T ‘ ,_—,.,- ,, -9 - ‘*“-\W- * «Wm-«4‘1" ‘4) ark“ a T '4 ‘ fih‘ LN a '/‘I. ’- -.I \/."__§ m If a V’- - o - - 7 f- ...?" sm ,3"? v ' ' “-5’.l " T " «0"... , I~ I a ‘—o. ’ e, F . . M -- v. -- : smrc ,7; -kMid-bfifiu‘rfij .. _ I. \'l _ " - sm ,3 a v' / - x 7 sm V - d \y — . — ., /—— - ~ ~.(_ I . I Figure 5. Bronchiolar epithelial morphometry following influenza infection. 47 epithelium accompanied by a marked lymphocytic inflammatory cell infiltrate in the affected bronchioles and surrounding alveolar parenchyma (interstitial pneumonia). Lesser, but conspicuous, numbers of eosinophils were also intermixed with the mononuclear inflammatory cells. There was also mild to moderate alveolar type II cell hyperplasia and hypertrophy in these affected parenchyma] regions along with numerous large highly vacuolated alveolar macrophages, smaller monocytes, and lymphocytes within alveolar air Spaces. Various amounts of proteinacious material were also present in some of the alveolar lumens in the affected regions of the pulmonary parenchyma. Mice that were instilled with virus and sacrificed at 10 dpi (Figure 5 E-H) had a chronic bronchiolitis and alveolitis, again restricted mainly to the proximal tissue section (hilar aspect of the lung lobe). At 10 days post-infection, the affected bronchiolar epithelium was composed of tall cuboidal to columnar ciliated and nonciliated cells (Figure 5G). Many of the nonciliated epithelial cells were mucous cells with conspicuous amounts of AB/PAS stained (Figure 5H), intracytoplasmic mucosubstances (i.e. Mucous cell metaplasia). The associated inflammatory cell infiltrate in and around the bronchiolar walls, adjacent blood vessels and alveolar parenchyma was similar in composition to that observed in mice sacrificed at 7 dpi. However, the most conspicuous change in the affected regions of the alveolar parenchyma at 10 dpi compared to that at 7 dpi was the addition of coalescing regions of alveolar fibrosis accompanying the type 11 cell hyperplasia and the mainly lymphocytic inflammatory cell infiltrate (chronic alveolitis). Mice instilled with virus and sacrificed at 15 and 21 dpi (Figure 5 I-L) had Similar but less severe airway and parenchyma] lesions as compared to those lungs of the mice sacrificed at 10 dpi. 48 B) Temporal analysis of stored intraepithelial mucosubstances following PR8 challenge The Vs of intracytoplasmic acidic and neutral mucosubstances (Figure 6) in the airway epithelium was increased by approximately 4-fold in lung lobe sections obtained from PR8 treated mice as compared to respective time-matched SAL controls starting as early as 10 dpi. The marked elevation in mucosubstance Vs was maintained through 21 dpi. C) Expression levels of whole lung MUCSAC mRNA at 7 days post infection Recognizing that the time to onset of MCM was established at 10 dpi, RNA from whole lung homogenates at 7 dpi was analyzed for MUCSAC mRNA levels (Figure 7). MUCSAC levels were nearly 3-fold greater in lung homogenates from mice treated with PR8 than those treated with SAL. D) Time-dependent differences in total and PCNA immunopositive epithelial numeric cell densities By numeric cell density counts, the total airway epithelial cell counts (Figure 8A) observed at 10 dpi were mildly elevated with respect to counts enumerated in SAL controls. In addition to total cell counts, cells with nuclei staining positive for PCNA (Figure 8B) were also enumerated. PCNA numeric cell density and labeling index (Figure 8C) were Significantly elevated as compared to time-matched SAL controls by four to six-fold in the PR8 treated group at 7 and 10 dpi, respectively. 49 1-5‘ 1:1 saline x - Influenza 3g 1.2- * * 3 E g g 0 8- * 3 .8 S E 0.4- g E 0.0- 3 7 1O 15 21 Time (Days) Figure 6. Time-to-onset of mucous cell metaplasia following influenza infection. Effects of influenza instillation on the volume density of intraepithelial mucosubstances in the epithelium lining the main axial pulmonary airways at generation 5 of the left lung lobe. Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and tissues collected and processed as described in Materials and Methods. Data is expressed as mean 3: SEM. * = significantly different from respective control instilled with SAL. 50 8 c: 4 «1 fi 1', 3 £ 2 2 3 1 D E 0 Saline Influenza Treatment Figure 7. MUCSAC gene induction by influenza at 7 days post infection. Effects of influenza instillation on the levels of MUCSAC expression at 7 dpi in the lungs of mice treated with either SAL or PR8. Data is expressed as mean 5: SEM. * = significantly different from respective control instilled with SAL. 51 Figure 8. Time-dependent proliferation of epithelial cells lining the main axial airway following influenza infection. Effects of influenza instillation on the numeric cell density of total epithelial cells (A), PCNA positive cells (B), and the labeling index for PCNA (C) in the epithelium lining the main axial pulmonary airways at generation 5 of the left lung lobe. Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and tissues collected and processed as described in Materials and Methods. Data is expressed as mean i SEM. * = significantly different from respective control instilled with SAL. 52 13° [:1 Saline I" a 150 - Influenza 3 .E 2 e 120 a a g; 90 3' 60 35 e 30 o — _ — 3 7 1O 15 21 Time (Days) 25 . I: Saline \ - Influenza Q (U 20 8 g . ,g 2 15 .2 To in in as 10 < E 5 E 5 n. o — — — 3 7 10 15 21 Time (Days) 15 * * D saline 1:0 - Influenza 8 1: 2 10 O .1: .2 < z 0 n. a! 3 7 10 15 21 Time (Days) Figure 8. Time-dependent proliferation of epithelial cells lining the main axial airway following influenza infection. E) Bronchoalveolar lavage fluid analysis 1. Protein BALF-associated protein levels were significantly increased in samples collected from mice infected with PR8 as compared with SAL instilled mice (Figure 9). Increased protein levels were observed between 7 and 21 dpi with an apparent apex at 10 dpi. 2. Total and differential inflammatory cell counts To further characterize the inflammatory cell milieu that was present in the airways during these epithelial changes, differential cell counts were performed. There was a significant rise in the total BALF-associated leukocytes (Figure 10A) in PR8 infected mice when compared to the respective SAL controls occurring as early as 3 dpi and lasting through 15 dpi with an apparent apex at 10 dpi. The early innate immune response was marked by significant increases in neutrophils (Figure 10B) by 3 dpi that tapered by 10 dpi. Macrophages and other monocytic cells (Figure 10C) were Significantly elevated between 7 and 15 dpi. The adaptive immune response was characterized by marked increases in lymphocytes (Figure 10D) between 7 and 21 dpi with peak numbers observed at 10 dpi. Eosinophils (Figure 10E) were also abundant between 7 and 15 dpi. 3. Inflammatory chemokines and cytokines With ongoing inflammation there are a host of chemokines and cytokines released by activated leukocytes. To characterize the pro-inflammatory chemical mediators retrieved in BALF, a mouse-specific cytometric bead array analysis of the inflammatory 54 1500' I: Saline - Influenza r_.1200- % 3 900‘ C 39'; 600- 2 “- 300- 0 1" 3 7 1O 15 21 Time (Days) Figure 9. Time-dependent detection of total protein in bronchoalveolar lavage fluid following influenza infection. Effects of influenza instillation on total protein detected in BALF supematants. Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and concentrations of total protein in BALF were determined as described in Materials and Methods. Data is expressed as mean i SEM. * = Significantly different from respective control instilled with SAL. 55 Figure 10. Time-dependent recruitment of leukocytes to the pulmonary airways following influenza infection. Effects of influenza instillation on total cells (A), neutrophil (B), macrophage (C), lymphocyte (D) and eosinophil (E) retrieved in bronchoalveolar lavage fluid (BALF). Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and inflammatory cells were enumerated in BALF as described in Materials and Methods. Data is expressed as mean i SEM. * = significantly different from respective control instilled with SAL. 56 Total number of cells I ml (x1o‘) Number of neutrophils / ml (x103) [:1 Saline - lnfluenm 3 7 10 15 21 Time (Days) \ D Saline 3 so I: Saline g - Influenza g. - Influenza C? t a g A t g g 30 E-vo ,5 5 n 2 E _ ~— _ 204 .. E 3 E .8 1 .8 10‘ a * E e . - z _ __ _ _ _ g - _ __ __ _ _ 3 7 10 15 21 3 7 10 15 21 Time (Days) Time (Days) D Saline - lnfluenm Number of eosinophils / ml (x103) Time (Days) Figure 10. Time-dependent recruitment of leukocytes to the pulmonary airways following influenza infection. 57 cytokines TNF-a, IFN-y, lL-6, MCP-l, 1L-10, and IL-12p70 was employed. The BALF- associated concentrations of TNF-or (Figure 11A), IFN-y (Figure 11B), IL-6 (Figure 11C), and MCP-l (Figure 11D), were significantly elevated in influenza infected mice when compared to SAL controls at 7 dpi. In addition, concentrations of IL-10 (Figure 11E) were significantly decreased in PR8 infected mice as compared to SAL controls at 21 dpi. Levels of TNF-a and IL-6 remained Significantly elevated through 10 dpi with similar trends observed with MCP-l. There were no changes in the BALF-associated concentrations reported for IL-l2p70 (Figure 11F) with either PR8 or SAL instillation. 4. T;;2 cytokines IL-4, IL-5, IL-9 and IL-13 The TH2 cytokines IL-4 (Figure 12A), IL-5 (Figure 128), IL-9 (Figure 12C) and IL-13 (Figure 12D) have been implicated in the development of MCM. By employing cytometric bead array kits and flex sets for these cytokines, we observed marked increases in the levels of IL-5 detected at 7 dpi in the BALF of PR8-infected mice as compared to SAL instilled mice. Furthermore, there were significant, albeit mild increases in the BALF levels of IL-4 and IL-9 detected at 7 dpi in PR8-infected mice as compared to SAL instilled mice. The concentration of IL-13 in BALF was low in all treatment groups with no detectable differences observed between PR8 and SAL instilled mice. 5. Elastase In addition to factors derived from lymphocytes, neutrophil-derived elastase has also been implicated as a factor known to induce MCM. In the current study, neutrophil- 58 Figure 11. Secretion of chemokines and cytokines into the airways in response to influenza infection. Effects of influenza instillation on TNF-a (A), IFN-y (B), IL-6 (C), MCP-l (D), IL-10 (E), and IL-12p70 (F) retrieved in bronchoalveolar lavage fluid (BALF). Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and inflammatory chemokines and cytokines were enumerated in BALF by flow cytometry as described in Materials and Methods. Data is expressed as mean i SEM. * = significantly different from respective control instilled with SAL. 59 300 D Saline E g t - Influenza e .. i E g 100 3 a ‘ e V ‘ it 5 E L'- 3 7 10 15 21 Time (Days) Time (Days) 250 a D Saline A 400 D Saline if 200 - Influenza I: 350 * - Influenza A < 150 s g e 100 E 200 E 15 * E g 10 T.’ 5° 3 E.) 25 2 0 3 7 10 15 21 3 7 10 15 21 Time (Days) Time (Days) A 1°° D Saline G: 1° I: Saline LL - Influenza 2’ - Influenza 2' ‘° 0 m _ _ .0 E E 8 8 40 * 5’ [x 2. .. a _J ‘- — =1' 3 7 10 15 21 3 7 10 15 21 Time (Days) Time (Days) Figure 11. Secretion of chemokines and cytokines into the airways in response to influenza infection. 60 [:3 Same - Influenza ’i lL-4(pglmlBALF) 'i’ '1’ f 1' It 3 7 10 15 21 Time (Days) C3 Saline - Influenza lL-9 (pg/ml BALF) asrr‘ (a) I 7 10 15 21 Time (Days) IL-5 (pg/ml BALF) IL-13 (pg/ml BALF) 3 7 1O 15 21 Time (Days) 1 :3 Saline in - Influenza g... 4.1 w Time (Days) Figure 12. Detection of T32 cytokines secreted into the airways in response to influenza infection. Effects of influenza instillation on the TH2 cytokines IL-4 (A), IL-5 (B), IL-9 (C), and IL-13 (D) retrieved in bronchoalveolar lavage fluid (BALF). Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and TH2 cytokines were enumerated in BALF by flow cytometry as described in Materials and Methods. Data is expressed as mean :t SEM. * = significantly different from respective control instilled with SAL. N.D. = not detected. 61 derived elastase (Figure 13) was significantly elevated between 7 and 15 dpi in PR8 infected mice as compared to SAL control mice. III. Modulation of airway responses to influenza A/PR/8/34 by delta-9- tetrahydrocannabinol in C57BL/6 mice A) A9-THC increases whole lung H1 mRNA following PR8 challenge H1 mRNA levels in the lungs of mice challenged with PR8 in the absence of A9- THC treatment were Significantly elevated at 7 dpi when compared to SAL control mice (Figure 14). Levels of H1 mRNA detected in the lungs of mice challenged with PR8 and treated with A9-THC at a dose of 25 mg/kg were also mildly attenuated when compared to mice challenged with PR8 alone. The levels of H1 mRNA increased with increasing doses of Ag-THC and were significantly elevated in mice administered 75 mg/kg A9-THC when compared to PR8 challenged mice in the absence of A9-THC treatment. B) A9-THC does not affect total protein levels in BALF As a measure of alveolar/capillary membrane integrity, total protein in BALF was assayed. There were marked increases in BALF-associated total protein observed in mice challenged with PR8 at 7 dpi when compared to SAL control mice. However, there were no differences observed in total BALF-associated protein recovered in PR8 challenged mice treated with any dose of A9-THC (Figure 15). C) A9-THC decreases leukocyte populations retrieved in BALF. Consistent with an immune response to viral challenge, the total number of 62 10' * E Saline - Influenza s: 3' * * < 2 2. 6" 0 fl, .9 4' In L“ Ill 2. o. — — — — 3 7 1O 15 21 Time (Days) Figure 13. Secretion of neutrophil-derived elastase into the airways in response to influenza infection. Effects of influenza instillation on neutrophil elastase detected in BALF supematants. Mice were instilled with PR8 or SAL and sacrificed 3, 7, 10, 15, and 21 dpi and neutrophil elastase in BALF were determined as described in Materials and Methods. Data is expressed as mean 1 SEM. * = significantly different from respective control instilled with SAL. 63 7 days post infection 8,1000- :3 Saline g Influenza if; 800- # E Q 600- ,_ '2 400- * .3 2 g 200- (I e, . a m V I I I I I I 0 0 25 50 75 Ag-THC (mg/kg) Figure 14. A9-THC treatment enhances hemagluttinin 1 mRNA levels. Effects of A9- THC on Hemagluttinin 1 mRNA levels from influenza challenge in whole lung homogenates at 7 dpi in mice treated with SAL or Ag-THC (0, 25, 50, or 75 mg/kg) with PR8. Data is expressed as mean i SEM; * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. 7 days post infection 30007 E Saline Influenza 25001 2000- 1500- Proteln (pig/ml) S ‘3 \\\\\\\\\‘l -k\\\\\\\\\i-I <3 awn . I I 0 25 50 75 A9-THC (mg/kg) Figure 15. A9-THC does not alter total protein levels in BALF. Effects of Ag-THC on total protein detected in BALF supernatants from mice challenged with influenza. Total protein in BALF was determined for mice treated with SAL or Ag-THC (0, 25, 50, or 75 mg/kg) with PR8 and sacrificed at 7 dpi. Data is expressed as mean i SEM; * — Significantly different from respective control instilled with SAL. 65 leukocytes retrieved in BALF was Significantly elevated in PR8 infected mice at 7 dpi (Table 2). There was a trend toward dose-dependent effects of A9-THC treatment on the total number of BALF-associated leukocytes. Treatment of mice with A9-THC at all dose levels led to significant reductions in the number of BALF-associated lymphocytes. Additionally, treatment of mice with A9-THC at doses of 25 mg/kg and 50 mg/kg led to a reduction in the number of macrophages retrieved in BALF. D) A9-THC decreases CD4+ and CD8+ T cells in BALF. Since treatment of PR8 infected mice with A9-THC led to decreases in the number of lymphocytes, we evaluated differences in T cell subsets by flow cytometry. The absolute values of BALF-associated CD4+ and CD8+ T cells were determined (Table 3). Treatment of PR8 infected mice with Ag-THC led to significant decreases in the number of CD8+ T cells at all dose levels with respect to mice challenged with PR8 alone. A similar effect on the number of CD4+ T cells was observed in PR infected mice administered 50 and 75 mg/kg A9-THC as compared to mice challenged with PR8 in the absence of A9-THC treatment. E) A9-THC modestly affects chemokines and cytokines retrieved in BALF. Inflammatory chemokines and cytokines secreted into the airways were measured as an indicator of immune cell function in response to PR8 challenge. Mice challenged with PR8 alone exhibited increased BALF concentrations of TNF-a , IFN-y, IL-6, MCP- 1, and IL-10 at 7 dpi as compared to the SAL control (Table 4). In PR8 infected mice treated with A9-THC, an increase in MCP-l at a dose of 50 mg/kg A9-THC was observed, 66 [gal C0 217- THC 219- THC A”. THC pe 25mg/kg 50mg/kg 75mg/kg SAL PR8 PR8 PR8 PR8 Total 3.1:1.0 68.2:12.7 * 34.3:9.1 31.9:6.7 92:12.5 (1:10“) p = 0.020 = 0.170 r; = 0.058 E = 0.501 (4) 4) 10 ) p= 0.006 = 0.005 1p = 0.118 (4) F:utrophils 2.9:2.0 1552.5:386. 9 E5)1.6:324.3 F586.3:412.5 2757.0:829.5 * Fiaerophages 2.9:0.9 18.8:3.6 * F031. .3 # t.6i1.0 # 13.3:3.4 @; x102) 0. 391 = 0.961 '1’ = 0.210 4) = 0.010 4 ymsphocytes 1.1:0.2 334.0:63.4 * 178. 8:50. 8 # 09:20.7 # 178.2:41.2 # x10) p = 0.002 =0 019 = 0.004 1p = 0.047 (4) osinophils 0 46.41163 * 3). 1:3. 0 33:21.3 48.617.86 x102) = 0.041 = 0.436 = 0.298 I = 0.654 (4) 4) Table 2. Total and differential cell counts from BALF. The effects of A9-THC on the recruitment of inflammatory cells to the pulmonary airways of mice challenged with influenza. Mice were treated with A9-THC (25, 50, or 75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Total leukocyte counts were enumerated by hemacytometer. Differential cell counts were assessed by counting 200 cells from cytospins of the BALF stained with Diff-quick. Data is expressed as mean i SEM; n = 5 except where noted by (n); * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. 67 T-cell to A’— THC A’— THC ’- THC Itype 25mg/kg 50mg/kg 75mg4kg SAL PR8 PR8 PR8 PR8 CD4+ 154.6:25.4 883.7:96.8 * 38.2:77.4 304.4:15.5 # 475.4:64.0 # (x104) p < 0.001 F = 0.626 < 0.001 < 0.001 (4) lo I0 CD8+ 66.7:265 4360.5:133.4 261.9:76.2 483.4:52.1 460.3:74.0 # (x104) I: * E E F < 0.001 4) in < 0.001 < 0.001 < 0.001 Table 3. CD4+ and CD8+ T cell populations observed in BALF by flow cytometry. The effects of A9-THC on lymphocyte populations of CD4+ and CD8+ T cells recruited to the airways of mice challenged with PR8. Mice were treated with A9-THC (25, 50, or 75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. CD4+ and CD8+ T cells in BALF were quantified by flow cytometric analysis. Data is expressed as mean 3: SEM; n = 5 except where noted by (n); * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. 68 hemokine/ Ico AT-THC AZTHC AT- THC ytokine 25mg/kg 50mg/kg 75mg/kg SAL PR8 PR8 PR8 PR8 IIL-6 09:05 497.5:862 * 32.711207 9549:2724 1120.0i168.9 (4) p= 0 001 = )0. 830 lp= 0.125 ip = 0.054 TNF-a. 6.7:4.7 145. 3+9. 9 * 129. 3+2. 9 145. 3+28. 0 155.8:262 (4)t14<)0. 001 k; 0.663 1p= 0.732 r= 0.977 FMCP-l 7.411. 1 1643. 0+296. 2 308. 0+70. 0 3301. 0+728. 4 5239017900 (4) * = )0 589 = 0.143 = 0.013 E4: 0.044 FN-y 1.5:0. 1 E4:11.0+263. 2 E2801+160 2 F415. .0+405. 0 l300.0_-L-489.4 (4) = 0.372 0.893 Ip = 0.815 0.048 4) [IL-10 46.212.91.51 0:52. 0 L123. .0+22. 4 4. 2+11 .3 43.8:223 (4) =0 119 0.477 (4) =0 067 p = 0.100 L-12p70 35:04 .08+0. 6 .3+0. 7 .0+0. 4 5:1.2 (3);: 0.051 E= 0.884 F= 0.922 F= 0.855 4) Table 4. Inflammatory cytokines quantified in BALF by cytometric bead array. The effects of A9-Tl-IC on soluble mediators released by immune and epithelial cells in the pulmonary airways during an inflammatory response to PR8 challenge. Mice were treated with A9-THC (25, 50, or 75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Concentrations of inflammatory cytokines in BALF were enumerated by cytometric bead array analysis on the flow cytometer. Data is expressed as mean i SEM; n = 5 except where noted by (n); * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. 69 Treatment of PR8 infected mice with Ag-THC at any dose, however, did not significantly alter the BALF concentrations of TNF-a, IFN-y, IL-6, IL-10 or IL-12p70 when compared to mice infected with PR8 alone. F) A9-THC treatment reduces inflammation scores for mouse lung sections. The magnitude and severity of inflammation observed in histological sections of lung isolated from the right and left lobes were independently scored (0 to 3, with O = no inflammation and 3 = severe inflammation) and compared between treatment groups at 7 dpi (Figure 16A) and 10 dpi (Figure 16B). There was no inflammation observed in the lungs of mice intranasally instilled with SAL. In contrast, there was a marked increase in the inflammation score noted for lung sections obtained from mice challenged with PR8 alone at both 7 and 10 dpi, representing a moderate to severe inflammatory response. There was a trend toward decreased inflammation scores for sections obtained from A9- THC treated mice challenged with PR8 at 7 dpi. The inflammation scores for sections obtained from Ag-THC treated mice challenged with PR8 were, however, significantly attenuated at 10 dpi, representing mild to moderate levels of inflammation. G) Effect of A9-THC on the observed pulmonary histopathology to PR8. Exposure of mice to the corn oil vehicle or A9-THC alone did not result in significant histologic changes within the control mice (Figure 17A). Infection of mice with influenza induced a significant cellular and inflammatory reaction 7 dpi in all lung regions examined. The inflammatory infiltrate was centered upon the bronchiolo- alveolar duct junction, and extended out into the surrounding alveolar parenchyma. The 70 Figure 16. A9-THC decreases inflammation scores in a time-dependent manner. Effects of A9-THC on the inflammatory response gathered within the subepithelial interstitium and alveolar parenchyma following influenza challenge. Inflammation scores were recorded from lung sections taken at 7 dpi (A) and 10 dpi (B). Scores were tabulated as discussed in Materials and Methods. Data is expressed as mean i SEM; N.D. = not detectable. * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. fl sum. t O O. y d 7 m 6 af 3. V///////////An /7///////%6 ///////////////.o A9-rHc (mg/kg) N.-o “ d 3 2 1 200m coast—bacc— 10 da ays post infection E..///////////..,. ...////////m ..////////.2. .///////////////O 119-THC (mg/kg) :BV 0.60m cow—«Epcot... Figure 17. Examination of the effects of A9-THC on the inflammatory response to influenza challenge by histopathology. Effects of A9-THC on the inflammatory response observed at generation 5 of the main axial airway on day 10 post infection. Mice were treated with Ag-THC (25, 50, or 75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 10 post infection. Lungs were fixed with 10% neutral- buffered formalin for 24 hours, then sectioned and stained with hematoxylin and eosin (H&E). Light photomicrographs were taken for lung sections at generation 5 of the main axial airway (MAA) to include the bronchiolar artery (a) and alveolar parenchyma (p). A = lung section from a mouse receiving corn oil gavage surrounding a SAL instillation. There was no evidence of inflammation in the perivascular/peribronchiolar submucosal compartment (sm) or alveolar parenchyma. B = lung section from a mouse receiving corn oil gavage surrounding an influenza instillation. There was severe inflammation of the perivascular/peribronchiolar submucosal compartment with marked extension into the alveolar parenchyma. C = lung section from a mouse receiving THC (75 mg/kg) gavage surrounding an influenza instillation. There was severe inflammation of the perivascular/peribronchiolar submucosal compartment with modest extension into the alveolar parenchyma. Bar = 100 microns. Images in this dissertation are presented in color. 73 Dc .1 . a l U .(1‘. a Y... . ff .0 .n ..., Lg“: ‘ rs..fls a an ...... . . .. ... 41.4.6-0... .. . .. . .1 “Nut-'4‘.» . ...«W’... . .. l "‘8'.‘ ‘ n We _. H . . ‘ MT! 0 1' .wmv . V M13 . £1. E. . ”WW1 v Pitt 4. . ... . an". . .. .: h .. ......flw. 4&8“... . .. . .. ......aflu. v .. .....w . ...... .. . Jr N‘.F$wavw. t 1.... x . 1‘7\ ,1: . . $.th u -THC on the inflammatory response to Figure 17. Examination of the effects of A9 histopathology. influenza challenge by 74 inflammatory cells were a mix of primarily lymphocytes and neutrophils, with fewer macrophages and plasma cells. The lymphocytic and neutrophilic population often filled the alveoli, and there was moderate alveolar interstitial infiltration with similar inflammatory cells. The lymphocytes often formed well-organized perivascular and peribronchiolar aggregates. Acute epithelial necrosis was present in small numbers of bronchioles, and the remaining epithelial cells were moderately attenuated. At 10 dpi with influenza the inflammation was more severe, and often obscured the alveolar parenchyma (Figure 17B). The inflammatory cells 10 dpi were primarily lymphocytes, with smaller numbers of neutrophils. The bronchiolar epithelium 10 dpi was moderately hyperplastic and hypertrophied, and there were scattered foci of alveolar bronchiolarization (extension of bronchiolar epithelium into the adjacent alveolar spaces). PR8-infected mice treated with Ag-THC resulted in no observed decreases in inflammation 7 dpi at each A9-THC dose. At 10 dpi, exposure of PR8-infected mice with all dose levels of Ag-THC resulted in a mild to moderate decrease in histologically apparent inflammation within the lungs (Figure 17C). The inflammation within the mice was not unifome distributed throughout all lung regions, as found in the control PR8- infected mice. The decreases in inflammation included both decreases in inflammatory cell numbers, as well as extent of distribution within the tissue. The bronchiolar epithelial changes noted above were still present 10 dpi in the A9-THC co-exposed mice. H) Caspase—3 mRNA expression levels and G5 numeric cell densities CAS-3 is a cellular biochemical marker of committed activation of signaling pathways that lead to cell death by apoptosis. In the current study, there were markedly 75 higher CAS-3 mRNA levels observed at 7 dpi in total lung homogenates from mice challenged with PR8 alone as compared to mice instilled with SAL alone. The increase in CAS-3 mRNA suggests an increased commitment to apoptotic cell death by cells present in the lung in response to PR8 infection (Figure 18A). Ag-THC treated mice infected with PR8 exhibited a trend toward increasing levels of CAS-3 mRNA with increasing doses of Ag-THC when compared to mice infected with PR8 alone. In addition, immunohistochemical staining for CAS-3 revealed marked increases in the number of CAS-3 positive cells in lung sections obtained from mice infected with PR8 alone at 7 dpi (Figure 18B). There was a marked decrease (p = 0.056) in CAS-3 immunoreactive epithelial cells at 7 dpi in PR8 infected mice treated with 25 mg/kg A9-THC when compared to mice challenged with PR8 alone (Figure 18B). At 10 dpi, there were no significant differences observed in the numeric cell densities for CAS-3 with any of the treatments. I) MUC5AC mRNA expression levels and G5 numeric cell densities Increases in M UC5AC gene transcription might be an early indicator of increased mucin production and possibly MCM in the bronchiolar epithelium. In the current study, the levels of M UC5AC mRNA were increased in mice challenged with PR8 alone when compared to mice instilled with SAL alone (Figure 19A). In Ag-THC treated mice challenged with PR8, a dose of 25 mg/kg Ag-THC led to a four-fold increase in MUC5AC mRNA levels as compared to mice infected with PR8 alone. In addition to changes in MUC5AC gene expression, there were marked increases in the number of epithelial cells staining for alcian blue (acidic mucosubstances) in lung sections obtained from mice 76 Figure 18. A9-THC has no effect on Caspase-3 mRNA levels and epithelial numeric cell densities. The effects of Ag-THC on apoptotic cell death in response to influenza challenge were measured by the expression of CAS-3 mRNA (A) and immunohistochemical staining of CAS-3 in the epithelium lining the main axial airway at generation 5 of the left lung lobe at 7 dpi (B) and 10 dpi (C). Mice were treated with SAL or Ag-THC (O, 25, 50, or 75 mg/kg) with PR8. Data is expressed as mean :t SEM; N.D. = not detectable. * = significantly different from respective control instilled with SAL. 77 7 days posr Infection .. 10 D Saline 3, u Influenza 5 .C ‘1’ 2 «3. i a 8 o Ag-Tl-IC (mglkg) 3 7 days post infection E 4,0 D Saline 3 n Influenza 5% 3.01 * 8.9 o _ g g 2.0T ég 1.0+ 3. 00- ND. 3 o o 25 so 15 ° A’JHC (mg/kg) 3 10 days post infection '5 20 D Saline 3 n Influenza gg 1.5- 8% g; ml ”3 E 0.5‘ % a 0.0.) ND. Z NtD. I g o o 25 so 75 119-THC (mglkg) Figure 18. A9-THC has no effect on Caspase-3 mRNA levels and epithelial numeric cell densities. 78 Figure 19. Ag-THC increases MUC5AC mRNA levels and decreases epithelial numeric cell densities for acidic mucosubstances. The effects of Ag-THC on the development of mucous cell metaplasia in the epithelium lining the main axial airway following influenza challenge and subsequent inflammatory cell responses. Expression levels of MUC5AC mRNA (A) and G5 main axial airway labeling indexes of alcian blue were determined at 7 dpi (B) and 10 dpi for mice treated with SAL or Ag-THC (O, 25, 50, or 75 mg/kg) with PR8. Data is expressed as mean i SEM; * = significantly different from respective control instilled with SAL. # = significantly different from corn oil with PR8 challenge. 79 7 days post infection 12 D Saline a; # m lnfluenza c 10 G if, 8 3 g 6 2 4 * It, ‘5’ 2 E o o o 25 so 75 Ag-THC (mg/kg) 7 days post infection E Saline m lnfluenza Alcian blue positive epithelial cells / mm basal lamina All-THC (mglkg) 10 days post infection E Saline lnfluenza 25 15 10 Alcian blue positive epithelial cells I rnm basal lamina A9-THC (mg/kg) Figure 19. Ag-THC increases MUC5AC mRNA levels and decreases epithelial numeric cell densities for acidic mucosubstances. 80 infected with PR8 alone at 10 dpi but not at 7 dpi (Figures 19B and C). Ag-THC treatment of PR8 infected mice did not affect the number of alcian blue stained epithelial cells observed along the main axial airway at 7 dpi (Figure 198), but did attenuate the number of alcian blue-positive cells observed at 10 dpi in mice receiving 75 mg/kg A9- THC (Figure 19C) when compared to mice challenged with PR8 alone. .1) A9-THC modestly enhances neutrophil-derived elastase levels in BALF Neutrophil-derived elastase is another soluble mediator secreted into the airways during ongoing inflammation that has been shown to have an influence on the development of MCM. Neutrophil-derived elastase was not detectable in BALF from SAL instilled mice (Figure 20). However, BALF-associated elastase was markedly increased in mice infected with PR8 alone. Ag-THC treated mice infected with PR8 exhibited a modest increase in elastase levels at a dose of 75 mg/kg Ag-THC when compared to mice challenged with PR8 alone. IV. Pulmonary airway responses to influenza A/PR/8/34 in CBI'I'lCBz’l' mice exposed to A9-tetrahydrocannabinol: An examination for the role of cannabinoid receptors A) Qualitative health assessment of CBf’VCBf” and wild type mice challenged with PR. CBl‘l’lCBg'l' and wild type mice treated with corn oil or Ag-THC and intranasally instilled with SAL were normal in appearance and activity level. Infection with PR8 in the presence or absence of A9-THC treatment, however, led to marked differences in the 81 7 days post infection Saline 25- El IZZZ Influenza # 20- 154 10- Elastase (V MAX) 5- 0- Ag-THC (mg/kg) Figure 20. Ao-THC modestly increases neutrophil-derived elastase levels secreted in the pulmonary airways. Effects of Ag-THC on the secretion of elastase by neutrophils in response to influenza infection within the pulmonary airways at 7 dpi. Neutrophil-derived elastase in BALF was determined for mice treated with SAL or Ag-THC (0, 25, 50, or 75 mg/kg) with PR8. Data is expressed as mean 2: SEM; * = significantly different from respective control instilled with SAL. N.D. = not detectable. # = significantly different from corn oil with PR8 challenge. 82 gross appearance of CB1'/'/CB2'/’ and wild type mice. Specifically, CB1'/'/CB2'/' mice were notably more gaunt in comparison to wild type mice, suggesting that the mice were dehydrated. In addition, the CBl'/'/CB2'/' mice were lethargic and displayed unkempt fur. In contrast, wild type mice infected with PR8 in the presence or absence of Ag-THC treatment maintained normal grooming habits and exhibited similar activity levels as the SAL instilled controls. Upon gross examination of the lungs, CBl'/'/CB2'/' mice had extensive hemorrhaging across all lung lobes. Wild type mice also displayed evidence of hemorrhaging. however, to a much lesser extent than CB|'/'/CB2'/' mice. B) The viral load of PR8 in the pulmonary airways of CB;"’/CB2"' and wild type mice measured 7 days after challenge. The levels of mRNA for the highly antigenic viral surface protein, hemagglutinin 1, were assessed by quantitative real time PCR. For both CB1'/'/CB2'/' and wild type mice, viral H1 expression was markedly elevated above SAL controls at 7 dpi in the lungs of all mice challenged with PR8 (Figure 21). Infection of A9-THC treated cert/€132"- mice with PR8 resulted in an increase (p = 0.056) in H1 mRNA levels when compared to mice instilled with PR8 alone. In A9-THC treated wild type mice infected with PR8, there was a significant increase in viral H] mRNA levels when compared to wild type mice instilled with PR8 alone. The overall level of expression of H1 mRNA, though, was greatly reduced in CB.'/'/CB2'/' mice when compared to wild type mice. C) A9-THC affects vascular permeability induced by PR8 infection of the airways in CBf’VCBf’“ and wild type mice. 83 *#:| Wild type - c314'lc324' * *+ *+ Com oil A’-THC Corn oll A9-THC Saflne lnfluenza Figure 21. Effects of A9-THC treatment on the expression levels of H1 mRNA in CBl‘/‘/CB2'/' and wild type mice. The effects of Ag-THC on Hemagglutinin 1 mRNA levels in lungs from CB1'/'/CB2'/' and wild type mice challenged with PR8. Mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. Whole lungs were immersed in TRI reagent and homogenized prior to the isolation of total RNA. The mRNA levels for Hemagglutinin 1 were determined by real-time PCR with 18S utilized as an internal loading control. The fold-change in gene expression is normalized to mice instilled with SAL. Data is expressed as mean i SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 84 The detection of increased levels of total BALF-associated protein is indicative of increased vascular, permeability at the alveolar/capillary interface during the inflammatory response to PR8. The total BALF-associated protein levels in PR8 infected CB1'/'/CB2'/' and wild type mice, in the absence of A9-THC treatment, were 2-fold greater than SAL controls (Figure 22). A9-THC treated CB1'/'/CB2'/' mice infected with PR8 exhibited a 33% increase in total protein when compared to CB1'/'/CB2'/' mice challenged with PR8 alone. Conversely, Ag-THC treated wild type mice infected with PR8 had a 50% decrease in total protein when compared to wild type mice challenged with PR8 alone. When comparing CBI"'/CB2'" and wild type mice, the amount of total protein detected in mice treated with Ag-THC and infected with PR8 was 2-fold greater in CBl'l' /CB2'/' mice than amounts of total protein observed in wild type mice receiving the same treatment. D) CBl'l'lCBz'I' mice treated with A9-THC exhibit a distinctly different composition of leukocytes recruited to the airways in response to PR8 infection when compared to wild type. Primary influenza infection elicits an inflammatory response consisting of a mixed population of leukocytes that infiltrate the pulmonary airways. In particular, there is a marked influx of neutrophils and lymphocytes into the airways with monocytes and eosinophils representing a smaller portion of the total population of BALF-associated leukocytes. In the current study, the total number of BALF-associated leukocytes was 3- fold greater in PR8-infected wild type mice and 4-fold greater in CB 14 '/CB2'/ ' mice when compared to their respective non-infected controls (Figure 23A). A9-THC treated mice infected with PR8 exhibited a 2-fold increase above SAL instilled controls in both 85 1500- :1 Wild type - cefivcey' *+ 1 200 - 900- e * 600- "‘°°ll]l nl ll 0 . Corn oil 119-THC Corn oil A9-THC Protein (pg/ml) Saline lnfluenza Figure 22. Effects of Ag-THC treatment on total protein in BALF in CBI'I'ICBz'I' and wild type mice. The effects of Ag-THC on total protein detected in BALF supematants from CB."l‘/CB2'/' and wild type mice challenged with PR8. Mice were treated with A9- THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Total protein was assessed for supematants from BALF that had been centrifuged to remove cellular debris. Data is expressed as mean i SEM: * = significantly different from respective control instilled with SAL. + = significantly different from respective group in wild type mice. 86 CB1’/'/CBz'/' and wild type mice. However, the difference in total leukocytes retrieved in the Ag—THC treated CB1'/'/CB2'/' mice infected with PR8 was significantly less than CBt'l' /CB2'/' mice challenged with PR8 alone. To further assess differences in individual leukocyte populations retrieved by lavage, differential cell counts were performed. Neutrophils (Figure 23 B) were markedly increased in PR8-infected CBt'l'lCB2'/' and wild type mice. Specifically, Ag-THC treated wild type mice infected with PR8 exhibited 2- fold increases in neutrophils when compared to wild type mice challenged with PR8 alone. Conversely, there were marked decreases in the number of BALF-associated neutrophils for PR8-infected CB1'/'/CBz'/' mice treated with Ag-THC when compared to CBt'/'/CB2'/' mice challenged with PR8 alone. BALF-associated lymphocytes were markedly increased (Figure 23C) with PR8 infection in both CBf/‘lCBz'l' and wild type mice. Ag-THC treated cert/C133“ and wild type mice infected with PR8 exhibited attenuation in the number of lymphocyte retrieved. There were no apparent changes in the number of macrophages retrieved in BALF from PR8 challenged wild type mice. A 2-fold increase was observed, however, in the number of BALF-associated macrophages from PR8-infected CB1'/’/CBz'/' mice that was attenuated in Ag-THC treated CB1'/'/CB2'/' mice infected with PR8 (Figure 23D). Lastly, PR8-infection of CB1" ”/CB2’/ ' and wild type mice resulted in an increase in the number of eosinophils retrieved in BALF (Figure 23E). A9-THC treated CBl'/'/CB2'/' and wild type mice infected with PR8 exhibited no effect in the number of BALF-associated eosinophils counted. 87 Figure 23. Effects of A9-THC treatment on total and differential leukocyte counts in BALF in CBf”/CB2"' and wild type mice. The effects of Ag-THC on leukocyte recruitment to the pulmonary airways of CBt'/'/CB2'/' and wild type mice challenged with PR8 were enumerated. Mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Total leukocyte (A) counts were enumerated by hemacytometer. Differential cell counts of neutrophils (B), lymphocytes (C), macrophages (D), and eosinophils (E) were assessed by counting 200 cells from cytospins of the BALF stained with Diff-quick. Data is expressed as mean :1: SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 88 v" v" o o . 3? 3: a D Wild type I a Wild type v - CB,"‘/CB;"‘ - cart/0132‘" 33 - 3 < _ m E E is 7, g. r: 8 E x s .2. _ _ _ a _ _ _ _ E Corn oil A9«THC Corn oil 119-THC 2 Com oil A9-THC Com oil Ag-THC '9 Saline lnfluenza Saline Influenza =7" 6" O O . 3 25 :1 Wild type 3 2° 2 £838; .,_ u. - cart/ca,"- u_ ‘ 2 _l 20 _l 15 < < at 15 m E E 1° 7, 1 B i“ 3 t a _ ._ _ _ 8 i Comoil AB-THC Comoil Ag-THC 2 Comoil 119-THC Comoil Ag-THC J Saline lnfluenza Saline Influenza «6‘ o . v; 20 D Wild type v - cart/c324- 5 < 15 a: E 1 . \ i g e -+ 5 § Com oil Ag-THC Com oil A9-THC Saline lnfluenza Figure 23. Effects of A9-THC treatment on total and differential leukocyte counts in BALF in CB1'/'/CB2"' and wild type mice. 89 E) BALF-associated CD4+ and CD8+ T cell levels following PR8 challenge in CBf" /CB;"' and wild type mice. To evaluate the contribution of CD4+ and CD8+ T cells within the pool of BALF- associated lymphocytes responding to PR8 challenge, the number of CD4+ T cells (Figure 24A) and CD8+ T-cells (Figure 24B) were enumerated by flow cytometric analysis. Wild type mice treated with either corn oil or Ag-THC exhibited modest levels of BALF-associated CD44r T cells. Interestingly, background levels of CD4+ T cells were greater in corn oil and A9-THC treated CB1'/'/CBZ'/' mice than in wild type mice with the same treatments. As a result, there was no observed difference in the number of CD4+ T cells retrieved in BALF between SAL instilled controls and CBt'/'/CB2'/' mice infected with PR8 alone. Alternatively, Ag-THC treated CBt'/'/CB2’/' mice infected with PR8 exhibited a significant increase in CD4+ T cells above background. The increase in BALF-associated CD4+ T cells in CB|'/'/CB2'/' mice treated with Ag-THC and infected with PR8 was comparatively greater than the respective treatment group in wild type mice. CD8+ T cells were not detected in BALF from SAL instilled controls in both CBf" /CB2‘/' and wild type mice. However, there were marked increases in the number of BALF-associated CD8+ T cells for both CB1'/'/CB2'/' and wild type mice challenged with PR8. In addition, there was a trend toward decreased numbers of BALF-associated CD8+ T cells in A9-THC treated wild type mice infected with PR8. 90 Figure 24. Effects of A9-THC treatment on CD4+ and CD8+ T cells retrieved in BALF in cert/cm" and wild type mice. The effects of Ag-THC on CD4+ T cell (A) and CD8+ T cell counts (B) recruited to the airways of CBt'/'/CB2'/' and wild type mice challenged with influenza. Mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. CD4+ and CD8+ T cells in BALF were quantified by flow cytometric analysis. Data is expressed as mean + SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. N.D.= not detected. 91 D Wild type mm H - car/7032+ 250- g *#+ 8 20» l— 150- + + 'k a 100- + * C) [Ill] [1 0 EH Corn oil Ag-THC Corn oil A9-THC Safine lnfluenza 200 I: Wild type B * - cert/032'“ :3. 150+ o 0 F' um- ... 8 0 501 0- Corn oil 119-THC Corn oil A9-THC Saline lnfluenza Figure 24. Effects of A9-THC treatment on CD4+ and CD8+ T cells retrieved in BALF in CB."‘/CB;"' and wild type mice. 92 F) Cannabinoid receptor deficient mice exhibit unique differences in epithelial and leukocytic chemokine and cytokine secretion in the pulmonary airways. One mechanism of cellular communication within a mixed population of leukocytes and between infected epithelium and leukocytes is through secretion of cytokines and chemokines. PR8-infected CB1'/'/CB2'/' mice exhibited marked increases in the chemokine MCP-l (Figure 25A), and pro-inflammatory cytokines TNF-a (Figure 25B), IL-6 (Figure 25C), and IFN-y (Figure 25D) measured in BALF. Likewise, wild type mice challenged with PR8 exhibited increased levels of BALF-associated MCP-l, TNF-a, IL-6, and IFN-y. In addition, there was a modest enhancement of IL-10 concentrations (Figure 25E) following PR8 challenge in CB1'/'/CB2’/' and wild type mice. There were no changes in BALF-associated IL-l2p70 in the presence or absence of A9- THC treatment or PR8 challenge (Figure 25F) in either CB1’/'/CB2‘/' or wild type mice. Ag-THC treated CBt'/'/CB2"' mice infected with PR8 exhibited an attenuation of BALF- associated IFN-y and IL-10 concentrations. Conversely, the concentrations of chemokines and cytokines detected in BALF in PR8-infected wild type mice were unaffected by A9- THC treatment. There were, however, comparative differences between detectable levels of BALF-associated TNF-a, and IFN-y (p = 0.061) in PR8-infected wild type and CB1'/’ /CB2'/' mice. The BALF-associated TH2 cytokines IL-2 (Figure 26A), IL-4 (Figure 26B), and IL-5 (Figure 26C) were also quantified by cytometric bead array. Of particular interest, PR-8 infected CB1'/'/CB2'/' mice exhibited marked increases in concentrations of IL-5 that were comparatively 2-fold less than concentrations observed in the cognate PR8-infected 93 Figure 25. Effects of A9-THC treatment on inflammatory chemokines and cytokines in secreted into the pulmonary airways in CBI'I'ICBz'l' and wild type mice. The effects of A9-THC on the release of soluble chemical mediators from leukocytes and epithelium in response to PR8 challenge of the pulmonary airways of CB.'/'/CB2'/' and wild type mice. Mice were treated with A9-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Concentrations of the inflammatory chemokine MCP-l (A) and cytokines TNF-a (B), IL-6 (C), IFN-g (D), IL-lO (E) and IL- 12p70 (F) were determined by cytometric bead array analysis. Data is expressed as mean :t SEM; * — significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 94 lL-6 (pg/ml) IL-1O (pg/ml) 1: Wild type - CBH‘ICBz'” Em E 8 3 :11» 1: a u'. 0 5 E :4- D - Wild type CB,"‘ICBZ‘" Corn oil A9-THC Corn oil Ag-THC Com oil 119-THC Corn oil 119-THC Saline Influenza Saline lnfluenza 1 D Wild type 400 D Wild type - CB{"/CB;"‘ - CBt"'/CBa"‘ Corn oil 119-THC Corn oil 119-THC Saline lnfluenza :1 Wild type - cart/032'“ Com oil Ag-THC Corn oil Ail-THC Saline lnfluenza IFN-y (pg/ml) lL-12p70 (pg/ml) Corn oil Ag-THC Com oil Ag-THC Saline lnfluenza :1 Wild type - CB1‘"/C82"' Corn oil Ag-THC Corn oil A9-THC Saline Influenza Figure 25. Effects of A9-THC treatment on inflammatory chemokines and cytokines in secreted into the pulmonary airways in CBt'l'lCBz'l' and wild type mice. 95 Figure 26. Effects of A9-THC treatment on TH2 cytokines secreted into the pulmonary airways in CBl'I'lCBz'I' and wild type mice. The effects of A9-THC on the concentrations of the TH2 cytokines IL-2 (A), IL-4 (B), and IL-5 (C) in BALF from CB1'/ ' /CB2'/’ and wild type mice challenged with PR8. Mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. BALF was collected by flushing the lungs with 2 ml of sterile SAL. Cytometric bead array was performed on BALF to enumerate the TH2 cytokines. Data is expressed as mean :1: SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 96 lL-4 (pg/ml) lL-2 (pg/ml) lL-5 (pg/ml) a Wild type - cafivcezt Corn oil A9-THC Corn oil A9-THC Saline lnfluenza 20 D Wild type - 03147035“ 16 12 + 8 4 o _ _ _ _ Corn oil AQ-THC Corn oil Ag-THC Saline lnfluenza 150 D Wild type - cert/032"- 125 1 00 75 50 25 Com oil A9-THC Corn oil Ag-THC Saline Influenza Figure 26. Effects of A9-THC treatment on TH2 cytokines secreted into the pulmonary airways in CB1'/'/CB2'/' and wild type mice. 97 wild type mice. The concentrations of lL-2 and IL-4 in BALF were unaffected by PR8 challenge in CB.'/'/CBz'/' mice. Ag—THC treated CB1'/'/CBz'/’ mice infected with PR8 had modestly attenuated concentrations of BALF-associated IL-2, but exhibited no effect on the concentrations of lL-4 or IL-5. PR8-infected wild type mice exhibited marked increases in BALF IL-5 concentrations, subtle increases in the concentrations of IL-2, and no detectable differences for IL-4 when compared to SAL instilled mice. A9-THC treated wild type mice infected with PR8 had a modest attenuation of BALF IL-2 concentrations with no observed effect on either IL-4 or IL-5 concentrations. G) Cytokines and chemokines detected in blood serum. In CB 1'/'/CB2’/' mice, PR8 challenge alone resulted in marked increases in serum associated MCP-l (Figure 27A), TNF-a (Figure 27B), lL-6 (Figure 27C), and IFN- y (Figure 271)) with respect to SAL control. In wild type mice, PR8 challenge increased circulating levels of the chemokine, MCP-l and the cytokine IFN-y with respect to SAL instilled mice. In CB1'/'/CB2'/' there was no difference observed between SAL controls and PR8 challenge for serum concentrations of IL-12p70 (Figure 27E). In wild type mice there were modest, yet significant, increases observed in the concentrations of IL-12p70 with PR8 instillation as compared to SAL instillation alone in the presence or absence of Ag-THC. CB1'/'/CB2'/' mice treated with Ag-THC had attenuated concentrations of MCP-l and IFN-y with respect to PR8 challenge alone, whereas wild type mice treated with A9- THC exhibited markedly enhanced circulating levels of IL-6 with respect to mice challenged with PR8 alone. In addition, there were comparative differences observed between CB1'/'/CB2'/' and wild type mice with PR8 challenge for the concentrations of 98 Figure 27. Effects of A9-THC treatment on inflammatory chemokines and cytokines released into blood serum in CBI'I'ICBz'l' and wild type mice. The effects of A9-THC on the dispersion of soluble chemical mediators from leukocytes and epithelium into the circulation in response to PR8 challenge of the pulmonary airways of CB1'/'/CB2'/' and wild type mice. Mice were treated with A9-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. Serum was collected from the abdominal aorta. Concentrations of the inflammatory chemokine MCP-l (A) and cytokines TNF-a (B), IL-6 (C), IFN-g (D), and IL-12p70 (E) were determined by cytometric bead array analysis. Data is expressed as mean i SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. N.D.= not detected. 99 a Wild type :1 Wild type -. cert/cm" - cert/ca."- g ... a 3 a ‘_ 100 ‘j o'. it o 2 5 Com oil A9-THC Comoil Ag-THC Corn oil Ag-THC Comoil Ag—THC Saline lnfluenza Saline lnfluenza 1: Wild type :1 Wild e - CB,"'/CBZ‘" - CB."‘/CBZ"‘ so A 2 ‘° E E s 3 3° 3 e — 10 Comoil Ag-THC Cornell AS-THC Comoil Aime Comoil ,9ch Saline lnfluenza Saline lnfluenza 10 D Wild type - cert/cs,"- lL-12p70 (pg/ml) Comoil Ag-THC Corn oil 119-THC Saline Influenza Figure 27. Effects of Ag-THC treatment on inflammatory chemokines and cytokines released into blood serum in CBi'/'/CB{" and wild type mice. 100 MC P-l detected. H) The absence of cannabinoid receptors CB; and CB; enhances the observed pulmonary histopathology. As previously reported by our laboratory, infection of mice with PR8 induces significant pulmonary inflammation 7 dpi in wild-type mice (Figure 28A-D) (113). Infection of the CBi'/'/CB2'/' mice with PR8 resulted in a similar inflammatory reaction (Figure 28E). As we have reported, the inflammation consists of primarily lymphocytes and neutrophils, with fewer macrophages and plasma cells centered upon the bronchiolo- alveolar junction, and extending out into the surrounding parenchyma. Treatment of PR8 infected wild-type mice with A9-THC resulted in a significant decrease in inflammation 7 dpi compared to corn oil treated mice infected with PR8 (Figure 28D). These mice often had no to few inflammatory cells present; the remaining inflammation, was comprised of primarily lymphocytes and alveolar macrophages. In contrast, treatment of PR8 infected CBt'/'/CB2'/' mice with A9—THC resulted in a vigorous inflammatory and cellular reaction in the mice (Figure 28F). In these mice there were large numbers of mature lymphocytes around the conducting airways and extending into the surrounding alveolar parenchyma where they were admixed with fewer macrophages and neutrophils. The bronchiolar epithelium was moderately hypertrophied. Treatment of wild type and CBi'/'/CB2'/' mice with corn oil vehicle or Ag-THC alone did not result in significant histologic changes in any of the lung sections examined. 101 Figure 28. Inflammatory response to PR8 in the proximal section of the left lung lobe in CBl'l’lCBz'l' and wild type mice. Light photomicrographs of the hilar region of the left lung lobe of wild type (WT) and CB1'/'/CB2"' (KO) mice sectioned at generation 5 along the main axial airway (maa). Panel (A): corn oil treated wild type mouse instilled with saline (SAL) with no evidence of an inflammatory cell infiltrate (arrow) in the peribronchiolar (maa)/perivascular (a) submucosa (sm) and no evidence of inflammation in the alveolar parenchyma (p). Panel (B): A9-THC-treated wild type mouse instilled with saline with marked inflammatory cell infiltrate of the submucosa and diffuse inflammation within the alveolar airspace. Panel (C): corn oil treated wild type mouse instilled with PR8 with marked inflammatory cell infiltrate in the peribronchiolar /perivascular submucosa and alveolar airspace. Panel (D): Ag-THC-treated wild type mouse instilled with PR8 with modest to no inflammatory cell infiltrate of the submucosa and alveolar airspace. Panel (E): corn oil treated CBt’/'/CBz'" mouse instilled with PR8 with marked inflammatory cell infiltrate in the peribronchiolar /perivascular submucosa and alveolar airspace. Panel (F): A9-THC-treated CB1'/'/CB2"' mouse instilled with PR8 with severe inflammation within the submucosa and alveolar airspace. Bar = 50 microns. Images provided in this dissertation are in color. 102 9 Corn oil treated A -THC treated A 1&5": ’9"! '3' 1"!” be"? B ~ .' 153%}: r'i‘l‘i-K, . ' , _ \-v‘:"‘ ‘1‘ ' ' ’1': . I. {k Figure 28. Inflammatory response to PR8 in the proximal section of the left lung lobe from wild type mice. 103 I) A9-THC affects the magnitude of the inflammatory response to PR8 in wild type and CB] and CB; deficient mice. The magnitude and severity of inflammation observed in tissue sections isolated from the left lung lobe were independently scored (0 to 3, with 0 = no inflammation and 3 = severe inflammation) and compared between treatment groups and wild type and CBi'/’/CB2'/' mice at 7 dpi (Figures 29). There was no inflammation observed in the lungs of wild type mice intranasally instilled with SAL. However, 2 out of 8 CBi'I'lCBz'I' mice instilled with SAL exhibited modest evidence of ongoing inflammation. Indeed, the CB{ "/CBZ'I' mice were subjected to an extensive battery of serological screens prior to their use and were found to be negative for all pathogens tested. Interestingly, there was a marked and identical increase in the inflammation score noted for lungs in PR8-infected wild type and CBi'/'/CB2'/' mice. Similar to previous findings by our laboratory (113), A9- THC treated wild type mice challenged with PR8 exhibited a suppression of the inflammatory response in the lung airways. In marked contrast, A9-THC treated CBf” /CB2'/' mice infected with PR8 had a trend toward increased (p = 0.079) inflammation of the pulmonary airways. J) Effects of CB1 and CB; deficiency on the numeric cell densities of apoptotic cells and metaplastic goblet cells. Bronchiolar epithelial cell apoptosis in influenza-infected mice is directed by cell- mediated immune responses to virally infected cells (114). The numeric cell density for the apoptotic cell marker, CAS-3 (Figure 30) was quantified at 7 dpi in the epithelium lining generation 5 of the main axial airway. CAS-3 numeric cell densities were not 104 3.0- 1: Wild type 25 - cefivcefi 2.0- * + 1.5- * * 1.0-l # 0.5- lll 0.0- ND. i N.D. i Corn oil A9-THC Corn oil All-THC Inflammation score Saline lnfluenza Figure 29. Effects of A9-THC treatment on histopathology-based inflammation scores in CBi'/'/CB2‘/' and wild type mice. Effects of Ag-THC on the inflammatory response gathered within the subepithelial interstitium and alveolar parenchyma following influenza challenge of the pulmonary airways in CB1'/'/CB2'/' and wild type mice. Mice were treated with A9-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza Virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 post infection. Lungs were fixed with 10% neutral- buffered formalin for 24 hours, then sectioned and stained with hematoxylin and eosin (H&E). Inflammation scores were recorded from lung sections taken on 7 dpi. Scores were tabulated and averaged for the two sections taken from generations 5 and 11 of the left lung lobe. Scores: 0 = no inflammation, l= mild, inflammatory cell infiltrate of the perivascular/peribronchiolar compartment, 2 = moderate, inflammatory cell infiltrate of the perivascular/peribronchiolar space with modest extension into the alveolar parenchyma, and 3 = severe, inflammatory cell infiltrate of the perivascular/peribronchiolar space with a greater magnitude of inflammatory foci found in the alveolar parenchyma. Data is expressed as mean :h SEM; N.D. = not detectable. * = significantly different from respective control instilled with SAL. + = significantly different from respective group in wild type mice. 105 2.5- D Wild type * - ce1"'/c324' 2.0- 1.5- 1.0- 0.5- 0,. N.D.N.D. N.D.N.D. N.D. Corn oil Ag-THC Corn oil A°.THC Caspase-3 positive cells I mm basal lamina Saline lnfluenza Figure 30. Effects of A9-THC treatment on the numeric cell density of caspase-3 positive epithelial cells in CBl'I'lCBz'I' and wild type mice. The effects of Ag-THC on apoptotic cell death in response to influenza challenge. CB1'/'/CB2"/' and wild type mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on day 7 and 10 post infection. Lungs were immersed in TRI reagent for the isolation of total RNA or fixed with 10% neutral-buffered formalin for 24 hours, then sectioned and stained by immunohistochemistry for Caspase-3. Numeric cell densities were determined by enumerating caspase-3 positive nuclei per length of basal lamina. Data is expressed as mean i SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 106 distinguishably different between treatment groups in wild type mice. In PR8-infected CB1'/'/CB2'/' mice, CAS-3 levels were modestly increased above SAL instilled controls, but were no different than cell densities enumerated for CB1°/'/CB2’/' mice infected with PR8 and treated with Aq-THC. During the recovery from viral infection a metaplastic change occurs in the epithelium lining the bronchi that includes increased numbers of mucus producing goblet cells (115). Consistent with our previous finding (113), there was no difference in the numeric cell density of mucus production (Figure 31) quantified at 7 dpi in wild type mice with any treatment. Rather interestingly, Ag-THC treated CBi‘/' /CBz'/' mice in the presence or absence of PR8 infection, exhibited significant increases in the numeric cell density for mucin-positive epithelial cells as compared to wild type mice. 107 Mucin positive cells I mm basal lamina 35- * + 1: Wild type 30- - cep’vcey' 25- 20- + 15- 10- 5 fli ii 0 7 — Com oil Ail-THC Corn oil A9-THC Saline Influenza Figure 31. Effects of Ag-THC treatment on the numeric cell density of mucosubstances in airway epithelial cells in CB.'/'/CBz'/' and wild type mice. The effects of Ag-THC on the development of mucous cell metaplasia in the epithelium lining the main axial airway following influenza challenge and subsequent inflammatory cell responses. CBfL/CBZ‘X' and wild type mice were treated with Ag-THC (75 mg/kg) or its corn oil vehicle for 5 consecutive days surrounding the intranasal instillation of influenza virus A/PR/8 or its vehicle SAL. Mice were euthanized on days 7 and 10 post infection. Lungs were fixed with 10% neutral-buffered formalin for 24 hours, then sectioned and stained with alcian blue/periodic acid Schiff (neutral and acidic mucosubstances). Numeric cell densities were determined by enumerating alcian blue positive secretory vesicles in epithelial cells per length of basal lamina from stained sections. Data is expressed as mean + SEM; * = significantly different from respective control instilled with SAL. # = significantly different from respective control gavaged with corn oil. + = significantly different from respective group in wild type mice. 108 DISCUSSION The studies outlined in this dissertation were a part of an effort to evaluate the effects of A9-THC and the role of the cannabinoid receptors CB1 and. CB2 on immune and pulmonary epithelial cell responses in a murine model of host-resistance to influenza virus A/PR/8/34 (Table 5). 1. Animals utilized in these studies In the studies outlined in this dissertation, C57BL/6 mice were utilized. C57BL/6 mice do not exhibit a genetically determined preference to either a THl or TH2 predominant immune response to pathogens like other mouse strains (e.g. Al or BALB-C mice). In addition, CB1/C82 receptor knockout mice were developed on a C57BL/6 background. 11. The method of delivery and the dose of PR utilized In the studies outlined in this dissertation, a PR8 exposure paradigm was chosen such that the mice were instilled with a low concentration (50 pfu) of PR8 to ensure the survival of the mice for the duration of the kinetic study (21 days). In addition, PR8 was intranasally instilled in a large volume of SAL (50 ill) to facilitate the delivery of PR8 to the lower airways. This paradigm is unlike most studies, wherein immune responses to influenza are examined within the first 6 — 10 days following a high concentration virus exposure. III. The method of delivery and dose of A9-THC utilized in these studies There are two ma'or routes of ex osure to Ag-THC in humans, inhalation and oral .l P 109 I1 Effect of corn oil on: Wild CB Saline Saline E MCM H&E Inflammato cells BALF Leuk BALF C kines Histoc Effect of A -THC on: Wild T CB] /CBz Saline Saline PR8 MCM -H-+ + H&E --- .H. Inflamma cells BALF Leuk + BALF C + Histochemi +1—1- Table 5. Summary of the effects of corn oil and A9-THC on BALF and histochemistry measurements taken for epithelial and inflammatory cells in wild type and CBt'l'lCBz'l' mice. Dashed line denotes no effect. A single plus denotes a mild effect, a double plus indicates a modest effect, and a triple plus signifies a marked response. 110 consumption. In the studies outlined in this dissertation, Ag-THC was administered orally in corn oil for five consecutive days. The oral route of administration was selected due to the fact that Ag-THC is a highly lipophilic molecule requiring a nonaqueous diluent for drug delivery, which in itself has the likely potential for inducing irritation and damage to the airways. In contrast, oral administration of Ag-THC in corn oil is well tolerated but poorly absorbed from the gastrointestinal tract resulting in modest blood concentrations of Ag-THC and its metabolites. Oral administration of 75 mg/kg A9-THC for 5 consecutive days resulted in a serum concentration of 66.2 ng/ml of the parent compounds, 446.5 ng/ml of the 9-COOH metabolite and 10.5 ng/ml of the ll-OH metabolite, four hours after the last Ag-THC dose. The levels of Ag-THC observed systemically correlate well with a previous report by Azorlosa and coworkers (1992) (116) in which peak human plasma levels ranged from 57 to 268 ng/ml. PR8 was administered by intranasal instillation in SAL on Day 3, with Ag-THC co-administration surrounding the day of infection. The rationale for this dosing paradigm was to investigate the putative affects of Ag-THC treatment on the immune response to PR8 during the early stages of the primary infection. IV. Effect of A9-THC on viral H1 mRNA levels in wild type and CB."' /CB¢"' mice. Viral H1 mRNA levels were quantified by real-time PCR, which allows for the analysis of large numbers of samples in a rapid manner while retaining the specificity and sensitivity of conventional methods such as the hemagglutination assays (56, 117). A9- THC treated mice exhibited a higher pulmonary viral H1 mRNA content, that was dose- dependent and without an effect on mortality, when compared to PR8 infected mice 111 treated with corn oil. These results suggest that Ag-THC administration impairs immune effectors involved in the clearance of PR8. By 10 dpi H1 mRNA levels approached the level of detection in all groups, hence suggesting that the clearance of the virus had occurred in all PR8 treatment groups. In comparison, influenza viral titres have been shown to return to baseline within 8 days of an uncomplicated influenza infection in humans (118). In the comparison of wild type and CBi’/'/CB2'/' mice, we observed greater H] mRNA levels in the lungs of Ag-THC treated mice challenged with PR8 when compared to mice challenged with PR8 alone in both wild type and CBi'/’/CB2'/' mice (113). Similar to our previous observation, we observed H1 mRNA levels in the lungs of A9-THC treated CB1"'/CB2'" and wild type mice challenged with PR8 that were greater than H1 mRNA levels in CB1'/'/CB2'/' and wild type mice challenged with PR8 alone. Interestingly, H1 mRNA levels were reduced in the lungs of CBi"'/CB2'/' mice by two orders of magnitude when compared to H1 mRNA levels observed for wild type mice. The profound reduction in H1 mRNA levels suggests that the cellular environment that supports viral entry or growth in the airway epithelium has been impaired in CB1'/'/CBz'/' mice when compared to wild type mice, or that the kinetics of the cell-mediated immune response are markedly enhanced in CBi'/'/CB2'/' mice. Our findings of increased background levels of CD4+ T cells in CB1‘/'/CB2'/' mice, in the current study, support the latter explanation by suggesting that a hyper-responsive immunity toward PR8 infection brought about by the absence of functional cannabinoid receptors CB] and CB2 exists. Moreover, since cannabinoid-mediated effects occurring through CB] and CB2 receptors on the immune system are most often associated with the suppression of immune 112 responses, the results presented herein would be consistent with the idea that CB1 and CB2 receptors play a role in dampening immune responses to maintain immune homeostasis. V. Effect of PR8 on inflammatory cell recruitment to the pulmonary airways in the presence or absence of A9-THC in wild type and CBi'" /CB2"' mice. Inflammatory cells entering the airways of PR8 infected mice followed a classic pattern of inflammation, in which an early pro-inflammatory response, represented by neutrophilic influx followed by monocytes and macrophages, was observed early at 3 and 7 dpi, respectively, and a delayed host-immune response, characterized predominantly by an influx of lymphocytes and to a lesser extent eosinophils peaked by 10 dpi. A9-THC treatment did not influence the total number of leukocytes retrieved in BALF between mice challenged with PR8 in the absence and presence of A9-THC treatment. In spite of trends toward decreased total leukocytes in BALF with Ag-THC treatment, further analysis of differential cell counts indicated that Ag-THC treatment decreased, in a dose-related manner, the number of macrophages and lymphocytes in the airways. Berdyshev and coworkers (119) have previously reported similar findings with the dose-dependent modulation of immune cell recruitment to the lungs by Ag-THC following endotoxin exposure. In light of the observation that Ag-THC modulated the differential leukocyte counts present in BALF, we further quantified the absolute number of CD4+ and CD8+ lymphocytes. Interestingly, Ag-THC administration attenuated both CD4+ and CD8+ cell counts by 2-fold. It is noteworthy that measurements of BALF primarily provided 113 information on the cellular and soluble mediators present in the alveolar air space and not from cells within the alveolar tissue. Given the marked lymphocytic and monocytic infiltration of the airway submucosa, it was important to consider these data in conjunction with histopathology, which suggest that Ag-THC modulated the immune response to PR8 through an influence on leukocyte migration. These findings are consistent with the effects of cannabinoids on lymphocyte and macrophage chemotaxis reported by others ( 120-123). In the comparison between wild type and CBi‘/'/CB2‘/‘ mice, the magnitude of total leukocyte recruitment to the pulmonary airways and the cellular subsets present were similar between CB1'/’/CB2’/' and wild type mice challenged with PR8, in the absence of Ag—THC treatment, suggesting that mice lacking functional CBi/CBz receptors were capable of mounting an aggressive immune response to PR8 infection. Differential cell counts provided evidence that the immune response to PR8 by CBi'/'/CB2'/' mice was more robust or efficient than the immune response mounted against PR8 in wild type mice. More specifically, the BALF-associated leukocytic subsets of macrophages and neutrophils were notably decreased in Ag-THC treated CB1'/'/CB2'/' mice infected with PR8. The decreased recruitment of macrophages and neutrophils to the airways might point to a cannabinoid receptor dependent effect on the chemotaxis of these leukocytes. Contrary to findings with macrophages and neutrophils, there were remarkable similarities in the total number of BALF-associated lymphocytes among all treatment groups for CBi'/'/CBz'/' and wild type mice. Upon further examination of the T cell subsets retrieved in BALF, there was evidence that the T cell response to PR8 challenge with A9-THC treatment was predominantly CD4+ T cells in CBi'/'/CBz'/' mice. As 114 mentioned previously, increased numbers of CD4+ T cells might suggest hyperresponsive immunity in CB1'/'/CB2'/' mice. However, the balance of the T-helper subset population, and the level of activity of these cells in the pulmonary airways remain unresolved. VI. Effect of PR8 on the release of soluble mediators into the pulmonary airways in the presence or absence of A9-THC in wild type and CBi'" ICBz'l‘ mice. In addition to replicating in the epithelial cells of the respiratory tract, influenza also infects monocytes/macrophages and other leukocytes by binding to sialic acid receptor moieties on these cell types (28). Virus infection activates several transcription factors within these cells that are involved in the induction of chemokine and cytokine gene expression. These include nuclear factor kappa B (NF -ch), interferon regulatory factors (IRFS), activating protein (AP)-1, signal transducers and activators of transcription (STATS) and nuclear factor-interleukin 6 (NF-IL-6 or C/EBPB) (36). Therefore, influenza plays a direct role in influencing the chemokine and cytokine makeup of the inflammatory response (reviewed by Julkunen (2001) (36) and Julkunen (2000) (29)). In these studies, the BALF-associated concentrations of MCP-l, TNF-a, IL-6, and IFN-y were primarily affected by PR8 infection of mice as compared to mice instilled with SAL. MCP-l is a chemokine that can be derived from either influenza- infected epithelia or infected monocytes/macrophages. lL-6 and TNF-a are pro- inflammatory cytokines produced predominantly by monocytes/macrophages. Instead of participating in an anti-viral capacity, TNF-a has been suggested to be a driving force for MCP-l expression. IL-6 serves as a differentiation factor for lymphocytes and stimulates immunoglobulin production by B cells. Interferon-y, produced by NK cells and/or TH] 115 cells, enhances the overall development of cell-mediated immunity, macrophage activation, antigen presentation, and chemokine gene expression. In these studies, each of these cytokines was Significantly elevated at 7 dpi. As Shown in the kinetic study (115), the immune response to PR8 treatment resulted in increased levels of lL-6, TNF-a, IFN-y, MCP-l and IL-10 by 7 dpi in BALF in the study that examined the dose-related effects of Ag-THC. Collectively, no clear or obvious profile of activity emerged concerning the effects of Ag-THC on PR8 cytokine or chemokine induction. This should not be altogether surprising as each cytokine and chemokine is regulated in its own unique manner with its own distinct kinetics. In addition, the molecular mechanism by which certain cytokines and chemokines are modulated by cannabinoids is only partially understood. In spite of this, several of the soluble inflammatory mediators evaluated in this study did exhibit dose-dependent modulation in response to Ag-THC treatment. Specifically, treatment of PR8 infected mice with Ag-THC at 50 mg/kg and 75 mg/kg led to increased levels of MCP-l and IL-6 in the BALF, whereas IL-10 concentrations were decreased at these dosage levels of A9- THC. The modulation of these cytokines in response to cannabinoids is supported by previous findings in other experimental models (121, 124). In the comparison of wild type and CB1'/'/CB2'/' mice, we observed increased total BALF associated protein in wild type mice challenged with PR8 that was mildly attenuated in Ag-THC treated wild type mice infected with PR8. Interestingly, total protein levels in BALF from CBi"'/CB2'/' mice challenged with PR8 alone were Similar to wild type mice challenged with PR8 alone, yet were enhanced in Ag-THC treated CBIJ’ /CB2"' mice challenged with PR8. Comparatively, a marked difference existed between 116 Ag-THC treated CB1'/'/CB2'/' and wild type mice infected with PR8, suggesting an increased severity in either vascular leakiness or inflammatory cell secretions in CBi'I' /CB2'/' mice. Consistent with increased amounts of total protein in BALF, inflammation scores accurately reflected changes observed in vascular leakiness. In addition to total protein, we also measure the BALF-associated chemokine/cytokine levels to provide information regarding the activity of leukocytes transient to the pulmonary airways. BALF-associated concentrations of MCP-l, TNF-a, and IFN-y in CBi'/'/CB2'/' mice infected with PR8, in the absence of A9-THC treatment were modestly elevated with respect to PR8 infected wild type mice. There was a trend toward decreased concentrations of these same cytokines in Ag-THC-treated CB1'/'/CB2'/' mice infected with PR8; however, the collective findings did not provide evidence for CB1 and/or CB2 receptor-dependent or -independent regulation of the secretion of these cytokines. Since CB1‘/'/CB2'/‘ mice had a lymphocytic response consisting of greater numbers of CD4+ T cells, and Ag-THC treatment of CBi'/'/CB2"' mice challenged with PR8 resulted in decreased concentrations of IFN-y, we also measured the BALF-associated concentrations of the TH2 cytokines IL-2, 1L-4, and IL-5. It has been suggested that A9- THC treatment modulates cytokine production in a way that results in decreased THl cell- mediated immunity and increased TH2 humoral immunity (125, 126). Hence, we explored the potential for decreased THl- type with concomitantly increased TH2-type cytokines in BALF. Only the cytokine IL-5 was markedly influenced by PR8 infection in wild type mice in the presence or absence of Ag-THC. IL-S concentrations were reduced in PR8- infected CB1'/'/CBz'/' mice in the presence or absence of Ag-THC. Ag-THC treatment of CBl'/'/CB2”' or wild type mice infected with PR8 exhibited no effect on the levels of 1L5 117 when compared to CBt'/'/CB2’/' or wild type mice infected with PR8 alone. The data suggest that the increased numbers of CD4+ T cells observed in CB1'/'/CBz'/' mice were not actively secreting more TH2-type cytokines than wild type mice and that there was not an imbalance between THl- type and TH2-type cytokines secreted due to A9-THC treatment in response to PR8. We further evaluated the magnitude of the inflammatory response to PR8 by measuring serum chemokine/cytokine levels. The serum profile of the pro-inflammatory chemokine MCP-l is markedly reduced for CBi'/'/CB2'/' mice challenged with PR8 in the presence or absence of Ag-THC as compared to wild type mice. The reduction in serum MCP-l is in stark contrast to elevated MCP-l concentrations for the same treatment groups observed locally in BALF in CBi'/’/CB2'/' mice. The cytokines TNF-a, IL-6 and IF N-y were elevated with PR8 challenge in the presence or absence of A9-THC. However, these cytokines were elevated modestly with respect to the detection limits of the assay. To glean a better understanding of the possible kinetic contribution of these chemokines/cytokines to the status of immune effectors responding to PR8, their combined serum and BALF concentrations were considered. The combined local and serum concentrations for MCP-l following PR8 challenge in the presence or absence of A9-THC suggest that macrophage recruitment Signals are localized to the pulmonary airways in CBi'/‘/CBz'/' mice and are more peripherally abundant in wild type mice. Furthermore, macrophage activity, as measured by TNF-a secretion, is enhanced both locally and peripherally with PR8 challenge in the presence and absence of Ag-THC in CB1'/'/CB2’/' mice. More importantly, IFN-y,a cytokine derived from TH] cells, is markedly elevated in CBi'/'/CB2'/' mice challenged with PR8 alone and suppressed by A9- 118 THC co-treatment. The combined peripheral and local chemokine/cytokine profiles in CB1'/'/CBp_'/' mice suggest a more vigorous recruitment of leukocytes to the pulmonary airways following PR8 challenge. VII. Effect of a single PR8 instillation on pulmonary histopathology in the presence or absence of A9-THC in wild type and CB." ' /CBz'/ ' mice. To clarify the relationship between cellular and biochemical findings in BALF with the dynamic process of airway inflammation, we examined differences in immune cell recruitment and associated epithelial cell morphology by histopathology. Challenging mice with PR8 yielded an observed airway epithelial degeneration and necrosis by 3 dpi with more extensive epithelial regeneration by 7 and 10 dpi, evidenced by the basophilic nature of the epithelium observed with H&E staining and the positive staining of cellular nuclei with antibodies directed against PCNA. Epithelial regeneration peaked at 10 dpi, and resolved by 15 dpi. Coincident with the regenerative process, was an increase in the total number of epithelial cells enumerated in the PR8 infected group. While epithelial regeneration was still active at 10 dpi, we observed a metaplastic change of the epithelium to include more mucus-producing goblet cells that did not resolve by 21 dpi. Given the marked inflammatory response evident at 7 dpi and the unique finding of MCM at 10 dpi, we examined the effects of Ag-THC on leukocyte infiltration and morphologic changes of the airway epithelium associated with influenza infection. Inflammation scores for histopathology were assigned to assess the influence of A9-THC on the host immune cell responses to influenza infection. Ag-THC, at all dose levels, attenuated the magnitude of inflammation at 10 dpi. Most impressively, the 119 histopathology clearly demonstrates that the inflammatory response in the lungs of mice challenged with influenza alone extends well beyond the perivascular/peribronchiolar submucosal compartment and into the alveolar parenchyma. With Ag-THC treatment, the inflammatory response was still centered around the perivascular/peribronchiolar submucosal compartment; however, the magnitude and severity of inflammation extending into the alveolar parenchyma was greatly reduced. Therefore, there was less tissue injury observed with A9-THC treatment. In the comparison of wild type and CB i'/'/CB2'/' mice, lung sections taken from the levels of the fifth (proximal) and eleventh (distal) airway generations to the main axial airway of the left lung lobe were scored for the magnitude and severity of inflammation present. Within saline instilled wild type mice there was no inflammation observed. On the other hand, in CBi"'/CB2'/' mice there were 2 out of 8 mice instilled with saline, in the presence or absence of Ag-THC treatment, which had modest evidence of inflammatory foci (Figures 8A and 8C). It remains unclear whether the isolated incidence of inflammation was the result of a subclinical infection in CB i'/'/CB2'/' mice that went undetected by serological screens or was a spontaneous occurrence of an underlying pathology in a mouse for which the absence of the cannabinoid receptors CB1 and CB2 might have rendered them immunologically hyper-responsive. Indeed, the CBi'l' /CB2'/' mice were subjected to an extensive battery of serological screens prior to their use and were found to be negative for all pathogens tested. Regardless, inflammation scores were similar for lung sections from CB1'/'/CB2'/' and wild type mice challenged with PR8, in the absence of Ag-THC treatment. In fact, the histopathology for both PR8- infected CB1'/'/CB2'/' and wild type mice demonstrated a similar pattern of inflammatory 120 cells infiltrating the perivascular/peribronchiolar submucosa and exhibiting a diffuse infiltration of the alveolar airspace (Figures 7B and SB). In A9-THC treated wild type mice challenged with PR8 there was a marked reduction in the inflammation score that was consistent with our previous finding (113). Also similar to our previous finding was the localization of inflammation to the submucosa with occasional inflammatory foci appearing in the alveolar parenchyma (Figure 8D). In contrast, inflammation scores for lung sections from Ag-THC-treated CB1'/'/CB2'/' mice challenged with PR8 were modestly enhanced with respect to scores assessed for lung sections from CB1°/'/CB2'/' mice challenged with PR8 alone. CBi'/'/CB2'/' mice treated with Ag-THC and infected with PR8 represented some of the most severely affected mice in the study and exhibited more vigorous inflammatory responses than CBi'/'/CB2'/‘ mice challenged with PR8 alone. Moreover the inflammatory response in Ag-THC treated CB1'/'/CB2'/' mice infected with PR8 was more pronounced than the respective A9-THC treated wild type mice challenged with PR8. Also of interest, was the Similarity between inflammation scores and concentrations of total BALF-associated protein and CD4+ T cells, suggesting that immune responses to PR8 in the CB i"'/CB2"' mouse are hypersensitive and include increased vascular permeability and a disproportionate number of CD4+ T cells. VIII. Effect of a single PR8 instillation on epithelial cell apoptosis and MCM in the presence or absence of A9-THC in wild type and CBt'l' lCBz"' mice. Lastly, we examined epithelial cell changes in airways (e.g., apoptosis and MCM) that occurred during the immune response to PR8. CAS-3 is a well-known marker for committed activation of apoptosis (127). Apoptosis can be initiated either directly by the 121 virus in infected host cells (128, 129), or by effector cell functions of cytotoxic T cells or NK cells. PR8 treatment alone enhanced mRNA levels of CAS-3 and tissue staining as expected. There was a dose-related decrease in CAS-3 tissue staining, but not mRNA levels with Ag-THC treatment. In the comparison of wild type and CB1'/'/CB2'/' mice, PR8 challenge led to increased CAS-3 staining as expected in both genotypes; however, there were no effects observed with Ag-THC administration. MCM is an adaptive response of the epithelium brought about by soluble mediators of inflammation. An early indicator of increased mucin production, and possibly MCM, is the expression of MUC5AC messenger RNA that encodes for the goblet cell-derived mucin MUC5AC. Mice challenged with PR8 in the absence of A9- THC exhibited an increase in both the levels of MUC5AC mRNA and alcian blue staining of tissue, as shown previously (115). Interestingly, A9-THC treatment at a dose of 25 mg/kg enhanced the levels of MUC5AC mRNA at 7 dpi, whereas the observed staining of mucosubstances was decreased with 75 mg/kg A9-THC at 10 dpi. Moreover, the dose-dependent effects of Ag-THC treatment on MUC5AC mRNA at 7 dpi and mucosubstance staining at 10 dpi were Similar in profile. It should be re-emphasized, that mRNA levels are reflective of whole lung homogenates, whereas quantification of morphologic changes in tissue sections (3 days later) is limited in scope to a single section of the lung. Therefore, the magnitude of the effect elicited by A9-THC is relative to the measurements made. It is presently unclear whether Ag-THC treatment can directly interfere with upregulation of MUC5AC gene expression or whether the effect is mediated indirectly through the suppression of the inflammatory response, more specifically soluble mediators derived from macrophages and lymphocytes (CD4+ and 122 CD8+ T cells). In the comparison of wild type and CBi'/'/CB2'/' mice, we surprisingly found that CBi'/'/CB2'/' mice had significantly greater numbers of goblet cells lining the airways at G5 when treated with Ag-THC alone at 7 dpi, suggesting that Ag-THC is potentially interacting directly with a signaling pathway tied with increased mucin production or mucous cell metaplasia in CB1" '/CB2'/' mice. VIII. Significance and relevance The body of work presented in this dissertation is a significant contribution to our understanding of Ag-THC and host-immunity to influenza infection. The present project characterized time—dependent epithelial and leukocyte responses to a low titre PR8 infection of the pulmonary airways of mice. A rather unique finding in our characterization was time-dependent PR8-induced MCM of the bronchiolar epithelium. Although, MCM has been previously reported for adenovirus (23), respiratory syncytial virus (RSV) (24) and influenza virus (25) infection, the study conducted as a part of this project was the first to characterize the adaptive epithelial response using morphometric analysis of the epithelium and cytometric bead array technology to examine a panel of cytokines that have been implicated in its etiology. A second unique contribution of these studies was the use of orally administered Ag-THC. Typically, A9-THC has been administered either intraperitoneally or intravenously for host immunity Studies. These routes of administration result in larger circulating concentrations of Ag-THC, initially. The circulating levels of A9-THC measured in the present studies were in the ng/ml range for Ag-THC and its metabolites. The concentrations of Ag-THC measured were a reflection of the poor absorption of A9- 123 THC from the gastrointestinal tract following oral administration. Nevertheless, it was interesting that these low circulating levels of Ag-THC were still capable of altering host- immunity, in particular leukocyte recruitment, to result in increased levels of viral H1 mRNA. Another unique aspect of these studies was the analysis of viral burden by quantitative real-time RT-PCR of the H1 gene. Traditionally, viral burden is measured by the Madin-Darby Canine Kidney (MDCK) cell assay. The difficulty with the MDCK assay is that it is not practical for a large number of samples and is not sensitive enough to distinguish between small differences in burden brought about by the introduction of another variable, like A9-THC treatment. Quantitative PCR-based methods have been Shown to have greater sensitivity and Specificity as compared to conventional methods for quantifying influenza virus, such as hemagglutination assays (56, 117). Moreover, RT-PCR allows for the analysis of large numbers of samples in a rapid and sensitive manner. Accordingly, the detection of viral H1 message in these studies demonstrated that A9-THC administration influenced viral H1 mRNA levels in a dose-dependent manner. Finally, these studies employed the use of CBi'/'/CB2'/' mice to elucidate the role of the cannabinoid receptors CB1 and CB; in the modulation of immune and epithelial cell responses to PR8. Prior to the generation of CB1‘/'/CB2'/' mice, the identification of CB1 and CB2 receptor-mediated events brought about by Ag-THC treatment were assessed by utilizing cannabinoid receptor antagonists for CB1 and CB2 (e.g., SR141716 and SR144528, respectively). However, these cannabinoid receptor antagonists have also been Shown to have partial and inverse agonist properties (130-132). Since the 124 generation of CB 1" '/CB2'/ ' mice, studies from our laboratory have begun to address the CB1 and CB2 receptor-independent actions for the suppression of the expression of cytokines IL-2 and IFN-y by the endocannabinoid 2-arachidonyl glycerol (83, 133). In the current studies, wild type and CB1'/'/CB2"' mice infected with PR8 in the presence or absence of Ag-THC demonstrate similarities in detectable levels of viral H1 mRNA and leukocyte recruitment, suggesting a CB1/CB2 receptor-independent mechanism of increased viral H1 mRNA content. However, the magnitude of expression of viral H1 mRNA was reduced by several orders of magnitude in CB1'/'/CB2'/' mice and there were markedly greater numbers of CD4+ T cells in the receptor-null genotype, suggesting that the kinetics for immune-mediated viral clearance were altered due to the lack of CB1/CB2 receptors. Therefore, the latter finding emphasized a CBt/CBz receptor-dependence for normal leukocyte recruitment and activation. In addition, another finding unique to the CB1'/'/CB2’/' mice was the increased numeric cell density of mucin-positive epithelial cells with Ag-THC treatment alone. The induction of MCM by A9-THC in the absence of PR8 challenge and, more importantly, in the absence of CBi/CBz receptors indicates that there are, indeed, other CB1/CB2 receptor-independent activities commenced by treatment with Ag-THC. This body of work is a relevant contribution to our understanding of the potential human health impacts of exposure to chemical agents that render individuals more susceptible to infection. In these studies we demonstrated that the use of an oral dose of Ag-THC, that produced serum levels of the parent compound in mice comparable to those found in humans, decreased host-immunity (e.g.; decreased leukocyte recruitment) to influenza virus while leading to increased levels of H1 mRNA in the lungs. This finding 125 is important in light of recent concerns regarding the potential for pandemic influenza infections and the susceptibility to infection of individuals who lack competent immune systems. In the last four years attention has been drawn to the newly emergent H5N1 influenza strain that has killed millions of birds in Asia. This virulent strain has jumped Species by infecting and killing a small number of people in Asia. Currently, the H5N1 strain has not mutated enough to allow for the efficient human-to-human transmission. However, it is noteworthy that the influenza pandemic of 1918 similarly started as a bird flu. Unlike previous pandemics, the threat of spreading influenza virus globally with greater speed has been enhance by our advances in travel. Therefore early detection has become a priority amongst health organizations like the Centers for Disease Control and the World Health Organization. In addition, people regarded as belonging to a group of individuals that are more “susceptible” to influenza have been encouraged to seek early vaccination. However the term “susceptible” is nebulous, and based on current guidelines, includes individuals with chemical-induced immune suppression. Within the last two decades we have witnessed, or in some states voted on, legislation dealing with the use of marijuana for medicinal purposes. This issue has been brought to our attention by advocates of medical marijuana for persons suffering from the adverse wasting effects of acquired immune deficiency syndrome (AIDS) or from nausea resulting from AIDS or chemotherapy (for treatment of AIDS or cancer). In 1985, the Food and Drug Administration approved the drug Marinol, a synthetic Ag-THC, for the treatment of these symptoms; however, patients have contested that the effects of this pure psychoactive drug rendered them incapacitated. Although patients suffering from AIDS or cancer may claim a therapeutic benefit from marijuana or its components, little is known regarding increased susceptibility of AIDS or cancer patients consuming Marinol or marijuana to infection by respirable pathogens, in light of an already compromised immunity. This is an important point Since one of the two known cannabinoid receptors (CB2) is found on immune cells, and cannabinoids have been shown in human and animal-derived immune cells to be immune suppressive. Therefore, even within the general population, recreational users of marijuana may be placing themselves at an added risk for a respiratory infection. Given the increased viral burden resulting from treating PR8-infected mice with Ag-THC, we explored the role of CB1 and CB; receptors in regulating Ag-THC-mediated immune suppression. The observations in these studies clearly point to the importance these receptors have in maintaining homeostatic immune responsiveness. These studies also provide evidence that there are other potential receptor targets for A9-THC. For example, the observation of increased MCM in CB1'/°/CB2'/' mice treated with Ag-THC may provide a tool with which to evaluate Signaling pathways linked with this metaplastic event in bronchiolar epithelium. 127 10. ll. 12. LITERATURE CITED Reznik, G. K. 1990. Comparative anatomy, physiology, and function of the upper respiratory tract. Environ Health Perspect 85 :1 71 . Ward, H. E., and T. E. Nicholas. 1984. Alveolar type I and type II cells. Aust N Z JMed14:731. Castranova, V., J. Rabovsky, J. H. Tucker, and P. R. Miles. 1988. The alveolar type II epithelial cell: a multifunctional pneumocyte. Toxicol Appl Pharmacol 93:472. Houtmeyers, E., R. Gosselink, G. Gayan-Ramirez, and M. Decramer. 1999. Regulation of mucociliary clearance in health and disease. Eur Respir J 13:11 77. Clarke, S. W. 1989. Rationale of airway clearance. Eur Respir J Suppl 7.'599S. Swain, S. L., J. N. Agrewala, D. M. Brown, D. M. Jelley-Gibbs, S. Golech, G. Huston, S. C. Jones, C. Kamperschroer, W. H. Lee, K. K. McKinstry, E. Roman, T. Strutt, and N. P. Weng. 2006. CD4+ T-cell memory: generation and multi- faceted roles for CD4+ T cells in protective immunity to influenza. Immunol Rev 21 1:8. Brown, D. M., A. M. Dilzer, D. L. Meents, and S. L. Swain. 2006. CD4 T cell- mediated protection from lethal influenza: perforin and antibody-mediated mechanisms give a one-two punch. J Immunol 177:2888. Brown, D. M., E. Roman, and S. L. Swain. 2004. CD4 T cell responses to influenza infection. Semin Immunol 16:] 71. Yagita, H., S. Hanabuchi, Y. Asano, T. Tamura, H. Nariuchi, and K. Okumura. 1995. F as-mediated cytotoxicity--a new immunoregulatory and pathogenic function of Th1 CD4+ T cells. Immunol Rev [46:223. Hahn, S., R. Gehri, and P. Erb. 1995. Mechanism and biological Significance of CD4-mediated cytotoxicity. Immunol Rev 146:5 7. Kumar, R. K., C. Herbert, and M. Kasper. 2004. Reversibility of airway inflammation and remodelling following cessation of antigenic challenge in a model of chronic asthma. Clin Exp Allergy 34:] 796. Reader, J. R., J. S. Tepper, E. S. Schelegle, M. C. Aldrich, L. F. Putney, J. W. Pfeiffer, and D. M. Hyde. 2003. Pathogenesis of mucous cell metaplasia in a murine asthma model. Am J Pathol 162.2069. 128 13. 14. 15. l6. 17. 18. 19. 20. 21. 22. 23. 24. Tesfaigzi, Y. 2002. The role of apoptotic regulators in metaplastic mucous cells. Novartis Found Symp 248:221. Fanucchi, M. V., J. A. Hotchkiss, and J. R. Harkema. 1998. Endotoxin potentiates ozone-induced mucous cell metaplasia in rat nasal epithelium. T oxicol App] Pharmacol 152:1. Harkema, J. R., and J. A. Hotchkiss. 1993. Ozone- and endotoxin-induced mucous cell metaplasias in rat airway epithelium: novel animal models to study toxicant-induced epithelial transformation in airways. T oxico] Lett 68:25] . Jamil, S., R. Breuer, and T. G. Christensen. 1997. Abnormal mucous cell phenotype induced by neutrophil elastase in hamster bronchi. Exp Lung Res 23:285. Kawano, H., A. Haruta, Y. Tsuboi, Y. Kim, P. A. Schachem, M. M. Paparella, and J. Lin. 2002. Induction of mucous cell metaplasia by tumor necrosis factor alpha in rat middle ear: the pathological basis for mucin hyperproduction in mucoid otitis media. Ann 0tol Rhino] Laryngol 111:415. Dabbagh, K., K. Takeyama, H. M. Lee, I. F. Ueki, J. A. Lausier, and J. A. Nadel. 1999. IL-4 induces mucin gene expression and goblet cell metaplasia in vitro and in vivo. J Immunol 162:6233. Justice, J. P., J. Crosby, M. T. Borchers, A. Tomkinson, J. J. Lee, and N. A. Lee. 2002. CD4(+) T cell-dependent airway mucus production occurs in response to IL-5 expression in lung. Am J Physio] Lung Cell Mol Physiol 28211066. Reader, J. R., D. M. Hyde, E. S. Schelegle, M. C. Aldrich, A. M. Stoddard, M. P. McLane, R. C. Levitt, and J. S. Tepper. 2003. Interleukin-9 induces mucous cell metaplasia independent of inflammation. Am J Respir Cell Mol Biol 28: 664. Shim, J. J., K. Dabbagh, I. F. Ueki, T. Dao-Pick, P. R. Burgel, K. Takeyama, D. C. Tam, and J. A. Nadel. 2001. IL-13 induces mucin production by stimulating epidermal growth factor receptors and by activating neutrophils. Am J Physiol Lung Cell Mol Physio] 280.1134. Holtzman, M. J., J. T. Battaile, and A. C. Patel. 2006. Immunogenetic programs for viral induction of mucous cell metaplasia. Am J Respir Cell Mo] Biol 35:29. Kajon, A. E., A. P. Gigliotti, and K. S. Harrod. 2003. Acute inflammatory response and remodeling of airway epithelium after subspecies Bl human adenovirus infection of the mouse lower respiratory tract. J Med Virol 71:233. Harrod, K. S., R. J. Jaramillo, C. L. Rosenberger, S. Z. Wang, J. A. Berger, J. D. McDonald, and M. D. Reed. 2003. Increased susceptibility to RSV infection by 129 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. exposure to inhaled diesel engine emissions. Am J Respir Cell Mol Bio] 28:451. Wohlleben, G., J. Muller, U. Tatsch, C. Hambrecht, U. Herz, H. Renz, E. Schmitt, H. M011, and K. J. Erb. 2003. Influenza A virus infection inhibits the efficient recruitment of Th2 cells into the airways and the development of airway eosinophilia. J Immunol 1 70.4601. Uhal, B. D., K. M. Flowers, and D. E. Rannels. 1991. Type II pneumocyte proliferation in vitro: problems and future directions. Am J Physiol 261:110. Palese, P., and J. L. Schulman. 1976. Mapping of the influenza virus genome: identification of the hemagglutinin and the neuraminidase genes. Proc Nat] Acad Sci USA 732142. Ronni, T., T. Sareneva, J. Pirhonen, and I. Julkunen. 1995. Activation of IFN- alpha, IFN-gamma, MxA, and IFN regulatory factor 1 genes in influenza A virus- infected human peripheral blood mononuclear cells. J Immunol 15 4:2 764. Julkunen, I., K. Melen, M. Nyqvist, J. Pirhonen, T. Sareneva, and S. Matikainen. 2000. Inflammatory responses in influenza A virus infection. Vaccine 19 Suppl 1:532. Chu, V. C., and G. R. Whittaker. 2004. Influenza virus entry and infection require host cell N-linked glycoprotein. Proc Natl Acad Sci U S A 101:18153. Tsuru, S., H. Fujisawa, M. Taniguchi, Y. Zinnaka, and K. Nomoto. 1987. Mechanism of protection during the early phase of a generalized viral infection. 11. Contribution of polymorphonuclear leukocytes to protection against intravenous infection with influenza virus. J Gen Virol 68 (Pt 2) :41 9. Allan, W., Z. Tabi, A. Cleary, and P. C. Doherty. 1990. Cellular events in the lymph node and lung of mice with influenza. Consequences of depleting CD4+ T cells. J Immunol [44:3980. Graham, M. B., V. L. Braciale, and T. J. Braciale. 1994. Influenza virus-specific CD4+ T helper type 2 T lymphocytes do not promote recovery from experimental virus infection. J Exp Med 180:12 73. Moran, T. M., H. Park, A. Femandez-Sesma, and J. L. Schulman. 1999. Th2 responses to inactivated influenza virus can Be converted to Th1 responses and facilitate recovery from heterosubtypic virus infection. J Infect Dis 180:5 79. Baumgarth, N., O. C. Herman, G. C. Jager, L. E. Brown, L. A. Herzenberg, and J. Chen. 2000. B-1 and B-2 cell-derived immunoglobulin M antibodies are nonredundant components of the protective response to influenza virus infection. J Exp Med 192:2 71. 130 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. Julkunen, I., T. Sareneva, J. Pirhonen, T. Ronni, K. Melen, and S. Matikainen. 2001. Molecular pathogenesis of influenza A virus infection and virus-induced regulation of cytokine gene expression. Cytokine Growth Factor Rev 12:1 71. Zlotnik, A., and O. Yoshie. 2000. Chemokines: a new classification system and their role in immunity. Immunity 12:121. Sprenger, H., R. G. Meyer, A. Kaufmann, D. Bussfeld, E. Rischkowsky, and D. Gemsa. 1996. Selective induction of monocyte and not neutrophil-attracting chemokines after influenza A virus infection. J Exp Med 184:1191. Bussfeld, D., A. Kaufmann, R. G. Meyer, D. Gemsa, and H. Sprenger. 1998. Differential mononuclear leukocyte attracting chemokine production after stimulation with active and inactivated influenza A virus. Cell Immunol 186:1. Matikainen, S., J. Pirhonen, M. Miettinen, A. Lehtonen, C. Govenius-Vintola, T. Sareneva, and I. Julkunen. 2000. Influenza A and sendai viruses induce differential chemokine gene expression and transcription factor activation in human macrophages. Virology 276:138. Matsukura, S., F. Kokubu, H. Noda, H. Tokunaga, and M. Adachi. 1996. Expression of IL-6, IL-8, and RANTES on human bronchial epithelial cells, NCI- H292, induced by influenza virus A. J Allergy Clin Immunol 98:1080. Adachi, M., S. Matsukura, H. Tokunaga, and F. Kokubu. 1997. Expression of cytokines on human bronchial epithelial cells induced by influenza virus A. Int Arch Allergy Immunol 1 13:30 7. Sareneva, T., S. Matikainen, M. Kurimoto, and I. Julkunen. 1998. Influenza A virus—induced IFN-alphafbeta and IL-18 synergistically enhance IF N-gamma gene expression in human T cells. J Immunol 160:6032. Nain, M., F. Hinder, J. H. Gong, A. Schmidt, A. Bender, H. Sprenger, and D. Gemsa. 1990. Tumor necrosis factor-alpha production of influenza A virus- infected macrophages and potentiating effect of lipopolysaccharides. J Immunol 145:1921. Gong, J. H., H. Sprenger, F. Hinder, A. Bender, A. Schmidt, S. Horch, M. Nain, and D. Gemsa. 1991. Influenza A virus infection of macrophages. Enhanced tumor necrosis factor-alpha (TNF-alpha) gene expression and lipopolysaccharide- triggered TNF-alpha release. J Immunol 1 4 7:350 7. Ronni, T., S. Matikainen, T. Sareneva, K. Melen, J. Pirhonen, P. Keskinen, and I. Julkunen. 1997. Regulation of IFN-alpha/beta, MxA, 2',5'-oligoadenylate synthetase, and HLA gene expression in influenza A-infected human lung 131 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. epithelial cells. J Immunol 1 58:23 63. Cella, M., M. Salio, Y. Sakakibara, H. Langen, I. Julkunen, and A. Lanzavecchia. 1999. Maturation, activation, and protection of dendritic cells induced by double- stranded RNA. J Exp Med 189:821. Baggiolini, M. 1998. Chemokines and leukocyte traffic. Nature 392:5 65. Stark, G. R. 1997. Genetic analysis of interferon and other mammalian signaling pathways. Harvey Lect 93:1. Marrack, P., J. Kappler, and T. Mitchell. 1999. Type I interferons keep activated T cells alive. J Exp Med 189:521 . Choi, A. M., K. Knobil, S. L. Otterbein, D. A. Eastman, and D. B. Jacoby. 1996. Oxidant stress responses in influenza virus pneumonia: gene expression and transcription factor activation. Am J Physiol 271 :L383. Hofmann, P., H. Sprenger, A. Kaufmann, A. Bender, C. Hasse, M. Nain, and D. Gemsa. 1997. Susceptibility of mononuclear phagocytes to influenza A virus infection and possible role in the antiviral response. J Leukoc Biol 61:408. Pahl, H. L., and P. A. Baeuerle. 1995. Expression of influenza virus hemagglutinin activates transcription factor NF -kappa B. J Virol 69:1480. Flory, E., M. Kunz, C. Scheller, C. Jassoy, R. Stauber, U. R Rapp, and S. Ludwig. 2000. Influenza virus-induced NF-kappaB-dependent gene expression is mediated by overexpression of viral proteins and involves oxidative radicals and activation of IkappaB kinase. J Biol Chem 275.8307. Hahon, N., J. A. Booth, F. Green, and T. R. Lewis. 1985. Influenza virus infection in mice after exposure to coal dust and diesel engine emissions. Environ Res 37:44. Jaspers, 1., J. M. Ciencewicki, W. Zhang, L. E. Brighton, J. L. Carson, M. A. Beck, and M. C. Madden. 2005. Diesel exhaust enhances influenza virus infections in respiratory epithelial cells. T oxico] Sci 85 .' 990. Cai, J., Y. Chen, S. Seth, S. Furukawa, R. W. Compans, and D. P. Jones. 2003. Inhibition of influenza infection by glutathione. Free Radic Biol Med 34:928. Lippmann, M., and R. B. Schlesinger. 2000. Toxicological bases for the setting of health-related air pollution standards. Annu Rev Public Health 21:309. Graham, D. E., and H. S. Koren. 1990. Biomarkers of inflammation in ozone- exposed humans. Comparison of the nasal and bronchoalveolar lavage. Am Rev 132 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. Respir Dis 142:152. Wolcott, J. A., Y. C. Zee, and J. W. Osebold. 1982. Exposure to ozone reduces influenza disease severity and alters distribution of influenza viral antigens in murine lungs. Appl Environ Microbiol 44: 723. Boatman, E. S., and R. Frank. 1974. Morphologic and ultrastructural changes in the lungs of animals during acute exposure to ozone. Chest 65 :Suppl:9S. Dungworth, D. L., W. L. Castleman, C. K. Chow, P. W. Mellick, M. G. Mustafa, B. Tarkington, and W. S. Tyler. 1975. Effect of ambient levels of ozone on monkeys. Fed Proc 34: 1 6 70. Freeman, G., R. J. Stephens, D. L. Coffin, and J. F. Stara. 1973. Changes in dogs' lung after long-terrn exposure to ozone: light and electron microscopy. Arch Environ Health 26:209. Dewey, W. L. 1986. Cannabinoid pharmacology. Pharmacol Rev 38:15] . Mechoulam, R., and Y. Gaoni. 1965. Hashish. IV. The isolation and structure of cannabinolic cannabidiolic and cannabigerolic acids. Tetrahedron 21:1223. Turk, R F., J. E. Manno, N. C. Jain, and R. B. Fomey. 1971. The identification, isolation, and preservation of delta-9-tetrahydrocannabinol (delta-9-THC). J Pharm Pharmacol 23:190. Coper, H. 1982. Chapter 8. Pharmacology and Toxicology of Cannabis. In Handbook of Experimental Pharmacology. Vol. 55/III. F. Hoffmeister, Stille, G., ed. Springer-Verlag , Berlin Heidelberg New York, p. 135. Coper, H., F ernandes,M., Honecker, H., Kluwe, S. 1971. The influence of solvent agents on the effects of cannabis. Acta. Pharmacol. Toxicol. 29:89. Perez-Reyes, M., M. A. Lipton, M. C. Timmons, M. E. Wall, D. R. Brine, and K. H. Davis. 1973. Pharmacology of orally administered 9 -tetrahydrocannabinol. Clin Pharmacol T her 14:48. Klausner, H. A., and J. V. Dingell. 1971. The metabolism and excretion of delta 9-tetrahydrocannabinol in the rat. Life Sci I 10:49. Ho, B. T., Fritchie, G.E., Kralik, P.M., Englert, L.F., McIsaac, W.M., Idanpaan- Heikkila, J. 1970. Distribution of tritiated-l-A9-tetrahydrocannabinol in rat tissue after inhalation. Journal of Pharmacy and Pharmacology 22:538. Matsuda, L. A., S. J. Lolait, M. J. Brownstein, A. C. Young, and T. I. Bonner. 1990. Structure of a cannabinoid receptor and functional expression of the cloned 133 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. cDNA. Nature 346:561. Munro, 8., K. L. Thomas, and M. Abu-Shaar. 1993. Molecular characterization of a peripheral receptor for cannabinoids. Nature 365:6]. Pettit, D. A., D. L. Anders, M. P. Harrison, and G. A. Cabral. 1996. Cannabinoid receptor expression in immune cells. Adv Exp Med Biol 402:119. Schatz, A. R., M. Lee, R. B. Condie, J. T. Pulaski, and N. E. Kaminski. 1997. Cannabinoid receptors C81 and CB2: a characterization of expression and adenylate cyclase modulation within the immune system. T oxico] Appl Pharmacol 1 42:2 78. McCoy, K. L., M. Matveyeva, S. J. Carlisle, and G. A. Cabral. 1999. Cannabinoid inhibition of the processing of intact lysozyme by macrophages: evidence for CB2 receptor participation. J Pharmacol Exp T her 289:1620. Buckley, N. E., K. L. McCoy, E. Mezey, T. Bonner, A. Zimmer, C. C. Felder, M. Glass, and A. Zimmer. 2000. Immunomodulation by cannabinoids is absent in mice deficient for the cannabinoid CB(2) receptor. Eur J Pharmacol 396:141. Zhu, L. X., S. Sharma, M. Stolina, B. Gardner, M. D. Roth, D. P. Tashkin, and S. M. Dubinett. 2000. Delta-9-tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-mediated, cytokine-dependent pathway. J Immunol 165:3 73. Ruiz, L., A. Miguel, and I. Diaz-Laviada. 1999. Delta9-tetrahydrocannabinol induces apoptosis in human prostate PC-3 cells via a receptor-independent mechanism. F EBS Lett 458:400. Chen, Y., and J. Buck. 2000. Cannabinoids protect cells from oxidative cell death: a receptor-independent mechanism. J Pharmacol Exp Ther 293 :80 7. Zygmunt, P. M., D. A. Andersson, and E. D. Hogestatt. 2002. Delta 9- tetrahydrocannabinol and cannabinol activate capsaicin-sensitive sensory nerves via a C81 and CB2 cannabinoid receptor-independent mechanism. J Neurosci 22:4720. Kaplan, B. L., C. E. Rockwell, and N. E. Kaminski. 2003. Evidence for cannabinoid receptor-dependent and -independent mechanisms of action in leukocytes. J Pharmacol Exp T her 306: 1077. Kaplan, B. L., Y. Ouyang, C. E. Rockwell, G. K. Rao, and N. E. Kaminski. 2005. 2-Arachidonoyl-glycerol suppresses interferon-gamma production in phorbol ester/ionomycin-activated mouse splenocytes independent of CB1 or CB2. J Leukoc Biol 77:966. 134 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. Costa, B., M. Colleoni, S. Conti, D. Parolaro, C. Franke, A. E. Trovato, and G. Giagnoni. 2004. Oral anti—inflammatory activity of cannabidiol, a non- psychoactive constituent of cannabis, in acute carrageenan-induced inflammation in the rat paw. Naunyn Schmiedebergs Arch Pharmacol 3 69:294. Watzl, B., P. Scuderi, and R. R. Watson. 1991. Influence of marijuana components (THC and CBD) on human mononuclear cell cytokine secretion in vitro. Adv Exp Med Biol 288:63. Srivastava, M. D., B. I. Srivastava, and B. Brouhard. 1998. Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 40:1 79. Zimmer, A., A. M. Zimmer, A. G. Hohmann, M. Herkenham, and T. I. Bonner. 1999. Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proc Nat] Acad Sci U S A 96:5 780. Galiegue, S., S. Mary, J. Marchand, D. Dussossoy, D. Carriere, P. Carayon, M. Bouaboula, D. Shire, G. Le Fur, and P. Casellas. 1995. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem 232:54. Specter, S., G. Lancz, G. Westrich, and H. Friedman. 1991. Delta-9- tetrahydrocannabinol augments murine retroviral induced immunosuppression and infection. Int J Immunopharmacol 13:41 1. Zheng, Z. M., S. Specter, and H. Friedman. 1992. Inhibition by delta-9- tetrahydrocannabinol of tumor necrosis factor alpha production by mouse and human macrophages. Int J Immunopharmaco] 14:1445. Coffey, R. G., Y. Yamamoto, E. Snella, and S. Pross. 1996. Tetrahydrocannabinol inhibition of macrophage nitric oxide production. Biochem Pharmacol 52: 743. Zhu, W., C. Newton, Y. Daaka, H. Friedman, and T. W. Klein. 1994. delta 9- Tetrahydrocannabinol enhances the secretion of interleukin 1 from endotoxin- stimulated macrophages. J Pharmacol Exp Ther 270:133 4 . McCoy, K. L., D. Gainey, and G. A. Cabral. 1995. delta 9-Tetrahydrocannabinol modulates antigen processing by macrophages. J Pharmacol Exp T her 273:1216. Smith, S. R, G. Denhardt, and C. Terminelli. 2001. The anti-inflammatory activities of cannabinoid receptor ligands in mouse peritonitis models. Eur J Pharmacol 432:1 07. Kawakarni, Y., T. W. Klein, C. Newton, J. Y. Djeu, G. Dennert, S. Specter, and H. Friedman. 1988. Suppression by cannabinoids of a cloned cell line with natural 135 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. killer cell activity. Proc Soc Exp Biol Med 1 8 7:355. Klein, T. W., C. A. Newton, R. Widen, and H. Friedman. 1985. The effect of delta-9-tetrahydrocannabinol and 11-hydroxy-delta-9-tetrahydrocannabinol on T- lymphocyte and B-lymphocyte mitogen responses. J Immunopharmacol 7:451. Kawakami, Y., T. W. Klein, C. Newton, J. Y. Djeu, S. Specter, and H. Friedman. 1988. Suppression by delta-9-tetrahydrocannabinol of interleukin 2-induced lymphocyte proliferation and lymphokine-activated killer cell activity. Int J Immunopharmacol 10:485. Zhu, W., T. Igarashi, Z. T. Qi, C. Newton, R. E. Widen, H. Friedman, and T. W. Klein. 1993. delta-9-Tetrahydrocannabinol (THC) decreases the number of high and intermediate affinity IL-2 receptors of the IL-2 dependent cell line NKB61A2. Int J Immunopharmacol 15:401. Klein, T. W., C. A. Newton, N. Nakachi, and H. Friedman. 2000. Delta 9- tetrahydrocannabinol treatment suppresses immunity and early IFN-gamma, IL- 12, and IL-12 receptor beta 2 responses to Legionella pneumophila infection. J Immunol 164:6461. Fischer-Stenger, K., A. W. Updegrove, and G. A. Cabral. 1992. Delta 9- tetrahydrocannabinol decreases cytotoxic T lymphocyte activity to herpes simplex virus type 1-infected cells. Proc Soc Exp Biol Med 200:422. Berdyshev, E. V., E. Boichot, N. Germain, N. Allain, J. P. Anger, and V. Lagente. 1997. Influence of fatty acid ethanolamides and delta9-tetrahydrocannabinol on cytokine and arachidonate release by mononuclear cells. Eur J Pharmacol 330:231. Condie, R., A. Herring, W. S. Koh, M. Lee, and N. E. Kaminski. 1996. Cannabinoid inhibition of adenylate cyclase-mediated signal transduction and interleukin 2 (IL-2) expression in the murine T—cell line, EL4.IL-2. J Biol Chem 271:13175. Jeon, Y. J., K. H. Yang, J. T. Pulaski, and N. E. Kaminski. 1996. Attenuation of inducible nitric oxide synthase gene expression by delta 9-tetrahydrocannabinol is mediated through the inhibition of nuclear factor- kappa B/Rel activation. Mol Pharmacol 50:334. Zheng, Z. M., and S. Specter. 1996. Delta-9-tetrahydrocannabinol: an inhibitor of STATl alpha protein tyrosine phosphorylation. Biochem Pharmacol 51:967. Klein, T. W., C. Newton, R Widen, and H. Friedman. 1993. Delta 9- tetrahydrocannabinol injection induces cytokine-mediated mortality of mice infected with Legionella pneumophila. J Pharmacol Exp T her 26 7: 635. 136 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. Morahan, P. S., P. C. Klykken, S. H. Smith, L. S. Harris, and A. E. Munson. 1979. Effects of cannabinoids on host resistance to Listeria monocytogenes and herpes simplex virus. Infect Immun 23:670. Mishkin, E. M., and G. A. Cabral. 1985. delta-9-Tetrahydrocannabinol decreases host resistance to herpes simplex virus type 2 vaginal infection in the B6C3Fl mouse. J Gen Virol 66 (Pt 12):2539. Lu, T., C. Newton, I. Perkins, H. Friedman, and T. W. Klein. 2006. Role of cannabinoid receptors in Delta-9-tetrahydrocannabinol suppression of IL-12p40 in mouse bone marrow-derived dendritic cells infected with Legionella pneumophila. Eur J Pharmacol 53 2:1 70. Lu, T., C. Newton, I. Perkins, H. Friedman, and T. W. Klein. 2006. Cannabinoid treatment suppresses the T-helper cell-polarizing function of mouse dendritic cells stimulated with Legionella pneumophila infection. J Pharmacol Exp Ther 319:269. Plopper, C. G., A. T. Mariassy, and L. O. Lollini. 1983. Structure as revealed by airway dissection. A comparison of mammalian lungs. Am Rev Respir Dis 128:S4. Harkema, J. R., and J. A. Hotchkiss. 1992. In vivo effects of endotoxin on intraepithelial mucosubstances in rat pulmonary airways. Quantitative histochemistry. Am J Pathol 141:307. Harkema, J. R., C. G. Plopper, D. M. Hyde, J. A. St George, and D. L. Dungworth. 1987. Effects of an ambient level of ozone on primate nasal epithelial mucosubstances. Quantitative histochemistry. Am J Pathol 127: 90. Buchweitz, J. P., P. W. F. Karmaus, K. J. Williams, J. R. Harkema, and N. E. Kaminski. 2007. Modulation of airway responses to influenza A/PR/8/34 by delta-9-tetrahydrocannabinol in C57BL/6 mice. Submitted for publication. Topham, D. J., R. A. Tripp, and P. C. Doherty. 1997. CD8+ T cells clear influenza virus by perforin or Fas-dependent processes. J Immunol 159:5197. Buchweitz, J. P., J. R. Harkema, and N. E. Kaminski. 2007. Time-dependent airway epithelial and inflammatory cell responses induced by influenza virus A/PR/8/34 in C57BL/6 mice. Tox Path. Azorlosa, J. L., S. J. Heishman, M. L. Stitzer, and J. M. Mahaffey. 1992. Marijuana smoking: effect of varying delta 9-tetrahydrocannabinol content and number of puffs. J Pharmacol Exp Ther 261 :1 1 4. Spackman, E., D. A. Senne, T. J. Myers, L. L. Bulaga, L. P. Garber, M. L. Perdue, 137 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. K. Lohman, L. T. Daum, and D. L. Suarez. 2002. Development of a real-time reverse transcriptase PCR assay for type A influenza virus and the avian H5 and H7 hemagglutinin subtypes. J Clin Microbiol 40:3256. Hayden, F. G., R. Fritz, M. C. Lobo, W. Alvord, W. Strober, and S. E. Straus. 1998. Local and systemic cytokine responses during experimental human influenza A virus infection. Relation to symptom formation and host defense. J Clin Invest 101:643. Berdyshev, E., E. Boichot, M. Corbel, N. Germain, and V. Lagente. 1998. Effects of cannabinoid receptor ligands on LPS-induced pulmonary inflammation in mice. Life Sci 63 :PL125 . Joseph, J., B. Niggemann, K. S. Zaenker, and F. Entschladen. 2004. Anandamide is an endogenous inhibitor for the migration of tumor cells and T lymphocytes. Cancer Immunol Immunother 53:723. Sacerdote, P., C. Martucci, A. Vaccani, F. Bariselli, A. E. Panerai, A. Colombo, D. Parolaro, and P. Massi. 2005. The nonpsychoactive component of marijuana cannabidiol modulates chemotaxis and IL-10 and 1L-12 production of murine macrophages both in vivo and in vitro. J Neuroimmuno] 1 5 9:97. Sacerdote, P., P. Massi, A. E. Panerai, and D. Parolaro. 2000. In vivo and in vitro treatment with the synthetic cannabinoid CP55, 940 decreases the in vitro migration of macrophages in the rat: involvement of both C31 and CB2 receptors. J Neuroimmuno] 109:155. Stefano, G. B., M. Salzet, C. M. Rialas, D. Mattocks, C. Fimiani, and T. V. Bilfinger. 1998. Macrophage behavior associated with acute and chronic exposure to HIV GP120, morphine and anandamide: endothelial implications. Int J Cardiol 64 Suppl 1:S3. Molina-Holgado, F ., E. Molina-Holgado, and C. Guaza. 1998. The endogenous cannabinoid anandamide potentiates interleukin-6 production by astrocytes infected with Theiler's murine encephalomyelitis virus by a receptor-mediated pathway. FEBS Lett 433:139. Newton, C. A., T. W. Klein, and H. Friedman. 1994. Secondary immunity to Legionella pneumophila and Th1 activity are suppressed by delta—9- tetrahydrocannabinol injection. Infect Immun 62: 4015. Yuan, M., S. M. Kiertscher, Q. Cheng, R. Zoumalan, D. P. Tashkin, and M. D. Roth. 2002. Delta 9-Tetrahydrocannabinol regulates Th1/Th2 cytokine balance in activated human T cells. J Neuroimmunol 133:124. Fischer, U., R. U. Janicke, and K. Schulze-Osthoff. 2003. Many cuts to ruin: a 138 128. 129. 130. 131. 132. 133. comprehensive update of caspase substrates. Cell Death Differ 10:76. Wurzer, W. J., O. Planz, C. Ehrhardt, M. Giner, T. Silberzahn, S. Pleschka, and S. Ludwig. 2003. Caspase 3 activation is essential for efficient influenza virus propagation. Embo J 22:27] 7. Takizawa, T., C. Tatematsu, K. Ohashi, and Y. Nakanishi. 1999. Recruitment of apoptotic cysteine proteases (caspases) in influenza virus-induced cell death. Microbiol Immunol 43:245. Krylatov, A. V., L. N. Maslov, O. V. Lasukova, and R. G. Pertwee. 2005. Cannabinoid receptor antagonists SR141716 and SR144528 exhibit properties of partial agonists in experiments on isolated perfused rat heart. Bull Exp Biol Med [39:558. Landsman, R. S., T. H. Burkey, P. Consroe, W. R Roeske, and H. I. Yamamura. 1997. SR141716A is an inverse agonist at the human cannabinoid CB1 receptor. Eur J Pharmacol 33 4:R I . Rhee, M. H., and S. K. Kim. 2002. SR144528 as inverse agonist of CB2 cannabinoid receptor. J Vet Sci 3:] 79. Rockwell, C. E., N. T. Snider, J. T. Thompson, J. P. Vanden Heuvel, and N. E. Kaminski. 2006. Interleukin-2 suppression by 2-arachidonyl glycerol is mediated through peroxisome proliferator-activated receptor gamma independently of cannabinoid receptors 1 and 2. Mol Pharmacol 70:101. 139 APPENDIX 1. Experimental design for examining the magnitude and severity of inflammatory response in mouse left lung lobes. Concentration Number of PR8 of mice 50 pfu 3 300 pfu 3 500 pfu 3 All animals were intranasally instilled with 50 ul of PR8 at concentrations of 50, 300, or 500 pfu. Animals were sacrificed 13 dpi. Lungs were inflation fixed with formalin and microdissected according to the material and methods. Sections of G5 and G11 were stained with H&E for analysis. 2. Experimental Design for examining the blood concentrations of Ag-THC and its metabolites ll-hydroxy-A9-THC and 9-carboxy—A9-THC. Dose Number of THC of mice 0 mg/kg 4 5 mg/kg 4 75 mg/kg 4 All animals were orally gavaged with either corn oil or Ag-THC for 5 consecutive days. On the fifth day, 4 h after the last dose of Ail-THC, the animals were anesthetized with 4% isoflurane and blood was removed from the animals by cardiac puncture. Dr. Jim Klaunig’s laboratory at IUPUI analyzed the blood samples for Ag-THC and its metabolites. 140 3. Experimental Design for examining the time-dependent changes in pulmonary responses to PR8. Intranasa] Sacrificed Number Treatment (dpi) of mice Saline 3 6 Influenza 3 6 Saline 7 6 Influenza 7 6 Saline 1 O 6 Influenza 1 0 6 Saline 1 5 6 Influenza l 5 6 Saline 2 1 6 Influenza 2 1 6 All animals were intranasally instilled with 50 ul of saline or PR8 at a concentration of 50 pfu. Animals were sacrificed at 3, 7, 10, 15, and 21 dpi. Lungs were lavaged with 2 ml of sterile saline and then inflation fixed with formalin and microdissected according to the material and methods. Sections of G5 and G11 were stained with H&E, AB/PAS, and PCNA for analysis. Lavage was quantified for total and differential cell counts, protein, elastase, and cytokines by cytometric bead array. 141 4. Experimental design for examining the concentration-dependent effects of A9- THC on pulmonary responses to PR8 (3 studies). Oral Intranasa] Sacrificed Number Treatment Treatment (dpi) of mice Corn oil Saline 7 5 Corn oil Influenza 7 5 THC 75 mg/kg Saline 7 5 THC 25 mg/kg Influenza 7 5 THC 50 mg/kg Influenza 7 5 THC 75 mg/kg Influenza 7 5 Corn oil Saline 10 5 Corn oil Influenza 10 5 THC 75 mg/kg Saline 10 5 THC 25 mg/kg Influenza 10 5 THC 50 mg/kg lnfluenza 10 5 THC 75 mg/kg Influenza 10 5 Three separate experiments with this design were conducted. All animals were orally gavaged with corn oil or 119-THC (25, 50, or 75 mg/kg) and intranasally instilled with 50 pl of saline or PR8 at a concentration of 50 pfu on the third day of gavage 4h prior to gavage. Animals were sacrificed at 7 and 10 dpi. In one experiment, lungs were lavaged with 2 m1 of sterile saline. Lavage was quantified for total and differential cell counts, protein, elastase, and cytokines by cytometric bead array. In the second experiment, the lungs were inflation fixed with formalin and microdissected according to the material and methods. Sections of R1-4, G5 and G11 were stained with H&E, AB/H for analysis. In the third experiment, the entire lung was immersed in TRI reagent and total RNA was isolated for H1, MUC5AC, and CAS-3 analysis. 142 5. Experimental design for examining the role of cannabinoid receptors in mediating the effects of A9-THC on immune cells and mediators following PR8 challenge (1" study). Ora] Intranasa] C5 7Bl/6 Sacrificed Number Treatment Treatment genotype (dpi) of mice Corn oil Saline wild-type 7 8 Corn oil Influenza wild-type 7 8 THC 75 mg/kg Saline wild-type 7 8 THC 75 mg/kg Influenza wild-type 7 8 Corn oil Saline CBl(—/-)/CB2(-/-) 7 8 Corn oil Influenza CBl(-/—)/CB2(-/-) 7 8 THC 75 mg/kg Saline CB1(-/-)/CBZ(-/-) 7 8 THC 75 mg/kg Influenza CBl(-/-)/CBZ(-/-) 7 8 Experimental design for examining the role of cannabinoid receptors in mediating the effects of A9-THC on immune cells and mediators following PR8 challenge (2"d study). Ora] Intranasa] C5 781/6 Sacrificed Number Treatment Treatment genotype (dpi) of mice Corn oil Saline wild-type 7 5 Corn oil Influenza wild-type 7 10 THC 75 mg/kg Saline wild-type 7 5 THC 75 mg/kg Influenza wild-type 7 10 Corn oil Saline CBl(-/-)/CBZ(-/-) 7 5 Corn oil Influenza CB1(-/-)/CB2(-/-) 7 10 THC 75 mg/kg Saline CB1(-/-)/CBZ(-/-) 7 5 THC 75 mg/kg Influenza CB1(-/-)/CBZ(-/—) 7 14 Using the first experimental design, all animals were orally gavaged with corn oil or A9- THC (75 mg/kg) and intranasally instilled with 50 pl of saline or PR8 at a concentration of 50 pfu on the third day of gavage 4h prior to gavage. Animals were sacrificed at 7 dpi. In the first experiment, right lung lobes were tied off and immersed in TRI reagent for the isolation of total RNA and the measurement of H1 mRNA. The left lung lobe was 143 lavaged with 1 ml of sterile saline and then inflation fixed with formalin and microdissected according to the material and methods. Lavage was quantified for total and differential cell counts, protein, elastase, and cytokines by cytometric bead array. The left lung lobe was sectioned at G5 and G11 and stained with H&E, AB/PAS for analysis. Using the second experimental design, the entire lung was immersed in TRI reagent and total RNA was isolated for H1 analysis. 144 u11111111111111“11113111