Next Article in Journal
In Response to: ‘Impact of Glycosylation on Effector Functions of Therapeutic IgG’ (Pharmaceuticals 2010, 3, 146–157)
Next Article in Special Issue
Cannabinoid-Induced Hyperemesis: A Conundrum—From Clinical Recognition to Basic Science Mechanisms
Previous Article in Journal
Use of p38 MAPK Inhibitors for the Treatment of Werner Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cannabinoids and Viral Infections

by
Carol Shoshkes Reiss
Department of Biology, Center for Neural Science, NYU Cancer Institute and Department of Microbiology, New York University, 100 Washington Square East, New York, NY, 10003, USA
Pharmaceuticals 2010, 3(6), 1873-1886; https://doi.org/10.3390/ph3061873
Submission received: 21 May 2010 / Revised: 28 May 2010 / Accepted: 9 June 2010 / Published: 9 June 2010
(This article belongs to the Special Issue Cannabinoids)

Abstract

:
Exogenous cannabinoids or receptor antagonists may influence many cellular and systemic host responses. The anti-inflammatory activity of cannabinoids may compromise host inflammatory responses to acute viral infections, but may be beneficial in persistent infections. In neurons, where innate antiviral/pro-resolution responses include the activation of NOS-1, inhibition of Ca2+ activity by cannabinoids, increased viral replication and disease. This review examines the effect(s) of cannabinoids and their antagonists in viral infections.

Graphical Abstract

1. Introduction

Both endogenous and exogenous cannabinoids can influence the course of infections in vitro and in vivo. This review will focus on viral infections of mammals, but will also describe what is known about other infections. Readers are directed to the excellent accompanying reviews in this issue which expertly discuss the clinical trials, cell biology, mechanisms of action, impact on inflammation, clinical applications, and so forth.
Cannabinoids may act either through the CB1 or the CB2 receptor, which are found on distinct cell types. The CB1 receptor is found on neurons as well as some astrocytes and skeletal muscle cells; neurons are frequently the target of viral infection. Engagement of the CB1 receptor by its endogenous or exogenous agonists may inhibit the release of Ca2+ from intracellular or extracellular stores. Since many important intracellular proteins are Ca2+-dependent for activation, signal transduction through the CB1 receptor may impair these secondary pathways and have a profound influence on the ability of viruses to replicate in neurons.
In contrast, the response of cells expressing the CB2 receptor may influence not only the responses in that cell, but may alter the course of the host innate and adaptive immune response to the pathogen, suppressing inflammation and the development of virus-specific cellular and humoral responses. The outcome on the viral infection will depend on whether inflammation is beneficial or pathogenic in the specific case.

2. Discussion

When a host is infected with a virus, there is a dynamic competition between the ability of the host to first marshal innate (hours to days) and then adaptive immunity (>7 days post infection) vs. the replication and spread of the virus first within the host and then to additional susceptible individuals. When a virus is able to out-pace the containment efforts, the host may succumb. Pathology may result from damage to tissues by viral-induced cellular apoptosis or necrosis, or alternatively, host immune responses may result in immunopathology or the perceived symptoms of the infection. If, however, innate and adaptive immunity successfully suppress viral replication, specific life-long immunity may result.
In order to understand the influences on the host response which may be the result of cannabinoids, it is important to examine some of the cellular pathways which are dependent on Ca2+-dependent enzymes. Table 1 indicates some of the well characterized pathways involved and their potential impact on viral infections.
The common recurring impact of Ca2+-dependent enzymes is a role in inflammation. This ranges from regulation of many signal transduction pathways, production of pro-inflammatory and pro-resolving lipid mediators downstream of arachidonic acid, to activation of Nitric Oxide Synthase and the production of reactive nitrogen intermediates, to proteolytic enzymes which remodel the cytoskeleton or extracellular matrix, and apoptosis.
Inflammation is essential for recruitment of both innate and adaptive immune cells to the site of infection to control virus production and limit spread, and then to promote recovery. Inflammation is comprised not only of non-specific cells (sequentially these are polymorphonuclear leukocytes, natural killer cells, macrophages) and then pathogen-specific T lymphocytes recruited from circulation, and activation of antibody-secreting B lymphocytes, but also induction of production and secretion of cytokines, chemokines, interferons, complement components, acute phase reactants, reactive oxygen and nitrogen intermediates, and other mediators [24,25,26]. Readers are referred to the accompanying review by Bani, Mannaioni, Passani, and Masini [27]. Thus, many of these critical pathways may be impaired or compromised when endogenous or exogenous cannabinoids are present during an infection [28].
Cannabinoids have been used both recreationally by groups of people who have viral infections, and experimentally by scientists investigating their impact in vitro or in animal models. Table 2 presents what has been published about these populations in peer reviewed journals. In most of the infections studied (Table 2), it is apparent that cannabinoid treatment, whether in vitro or in vivo, had profound impact on the virus-host (cell) interactions. For HSV-2, HIV-1, KSHV, influenza and VSV viral replication, or surrogate measures of infection, were found to be substantially increased upon cannabinoid treatment [30,34,39,50,52,63]. In HIV-1 infection, syncytia formation was enhanced, and monocytes were stickier on endothelial cells [57,58]. In one study, KHSV was more likely to exit latency and enter lytic infection when transformed cells were treated with THC [39], however, another study found the opposite result in several herpesvirus infections [38].
Table 1. Some Ca2+-dependent enzymes which may be inhibited by Cannabinoids and speculated role in host responses relevant for viral infections.
Table 1. Some Ca2+-dependent enzymes which may be inhibited by Cannabinoids and speculated role in host responses relevant for viral infections.
Enzyme primary/secondaryPathwaysRef.Role(s) in viral infection-host responses
cPhospholipase A2Arachidonic acid metabolites (prostaglandins, leukotrienes, lipoxins, resolvins) and inflammation[1,2]Inflammation and its resolution
Phospholipase C
 - Receptor-mediated tyrosine kinase
Production of Inositol 1,4,5-triphosphate from phosophotidylinositol[3]Signal transduction
Phospholipase D1Exocytosis in neuroendocrine cells[4]Neurotransmission
CalcineurinActivation of NFAT—gene expression[5,6]Signal transduction
Ca2+-Calmodulin
 - Nitric oxide synthase-1
 - Nitric oxide synthase-3
Conversion of argenine to NO in neurons and endothelial cells; production of ONOO-, -SNO, -R-NO2
Inhibition of viral infection
[7,8,9,10,11,12]Anti-viral; NO2-decoration of viral proteins; capillary dilation; inflammation
Ca2+-Calmodulin dependent protein kinases
    - CREB
    - CaMKK activation of AMPK
Wnt-2-dependent dendrite growth & cardiomyogenesis

Energy, epithelial cell polarity

T cell activation
[13,14,15,16,17]Adaptive immune responses; inflammation
Calpains [Ca2+-dependent proteases]Neutral proteases [many tissues]
Cell membrane fusion, synaptic remodeling, activating PKC, remodeling cytoskeleton, transcription factors
[18,19,20]Cytoskeletal plasticity, cell migration, inflammation
Matrix metalloproteinasesExtracellular matrix remodeling, inflammation[21]Inflammation
CalpastatinCell fusion in fertilization[22]Formation of heterokaryons /giant cells
TransglutaminasesCross-linking/deamination of proteins –wound healing, tissue repair, apoptosis, cell cycle control, inflammation and fibrosis[23]Inflammation, fibrosis, cell cycle and programmed cell death
Table 2. Cannabinoids and Viral Infections.
Table 2. Cannabinoids and Viral Infections.
Viral pathogenIn vivo In vitroAgonist / AntagonistTiter changePathogenesisInflammation Immunoregu-lationCommentsRef.
HSV-2,
L. monocyto-genes
In vivoΔ9-THC decreased resistance to LD50 systemic infection[29]
HSV-2In vivoΔ9-THCincreased sheddingincreased severity of lesions & mortalitydelayed onset of DTH responsevaginal model B6C3H F1 mouse[30]
HSV-2In vivoΔ9-THC decreased Type I IFN responsei.v. infection[31]
HSV-2In vivoΔ9-THC decreased resistance to infection; increased severity of lesions vaginal guinea pig model[32]
HSV-1,-2In vitroΔ9-THCfailed to replicate antiviral effect in human & monkey cells[33]
HSV-2In vitroΔ9-THC100-fold increase in released virus Vero cells, increased CPE[34]
HSV-2bothΔ9-THC decreased T cell proliferationB6C3H F1 mice immunized then T cells cultured[35]
HSVIn vitroΔ9-THCdecreased infectivity in TC virus incubated with THC [36]
HSV-1bothΔ9-THC decreased CD8 CTL activityC3H mice immunized, L929 targets[37]
EBV, KSHV, HVS, HSV-1, MHV-68In vivoΔ9-THCImmediate early ORF promoter activity inhibitedreactivation from latency inhibited latently infected B cells in tissue culture[38]
KSHVIn vivoΔ9-THCincreased viral loadincreased efficiency of infection, activation of lytic switchincreased transformation of endothelial cellsprimary human dermal microvascular cells[39]
CowpoxIn vivoMarijuana cigarettes generalized infectionweak Ab production, no neutralizing AbsCase report[40]
TMEVIn vitroAnandamide decreased release of NO2- and TNF-αNO is antiviral for TMEV [41,42]
TMEVIn vitroAnandamide increased IL-6 productionastrocyte culture B6 and SJL mice[43]
TMEVIn vivoWIN-55,212 ameliorates progression of autoimmune disease TMEV-IDDdecreased DTH, decreased IL-1, IL-6, IFN-γ , TNF-α, TMEV-IDD a mouse model of MS[44]
TMEVIn vivoOMDM1, OMDM2 ameliorated motor symptomsdecreased MHC II, inhibited NOS-2, reduced proinflammatory cytokinesTMEV-IDD proposed MS therapy with cannabinoids[45]
TMEVIn vitroJWH-133 SR144558 role of CB2 receptors in anti-inflammatory actionsreduced IL-12p40, reduced ERK1/2 signaling [46]
TMEVIn vitroWIN-55,212 CB2-dependent COX-2 induction increased vs. TMEV-alonerole of PI3 kinase pathway in CB2 but MAPK for TMEV signalingproposed role on blood-flow and immune activity[47]
TMEVIn vivoPalmitoyl-ethanol-amine reduction in motor disability in TMEV-IDDanti-inflammatory effectTMEV-IDD[48]
TMEVbothWIN-55,212 inhibited ICAM & VCAM on endothelium; role for PPAR-γ receptors in mechanismreduced inflammationTMEV-IDD [49]
InfluenzaIn vivoΔ9-THCHA mRNA increasedinflammation, metaplasia of mucous celldecreased CD4, CD8, and macrophage recruitment [50]
InfluenzaIn vivoΔ9-THCHA mRNA decreased in CB1/CB2KO miceTHC-mediated airway pathology +/- CB1/CB2KO mice had increased CD4 and IFN-γ recruitmentCB1/CB2 KO mice[51]
VSVIn vitroWIN-55,212increased viral titersCB1-dependent; decreased NOS-1 activityantagonized IFN-γ-mediated antiviral pathwaysuggested disease progression likely in neurons/viral encephalitis[52]
BDVIn vivoWIN-55,212 protected BrdU-positive neural progenitor cells in striatumsuppressed microglial activationsuggested treatment of encephalitis with microglial inflammation and neuro-degeneration[53]
HCVIn vivoMarijuana cigarettes progression of liver fibrosis epidemiological study[54]
HCVIn vivoOral cannabinoids improved weight no viral markers or immune markers studied7 week clinical trial for anorexia and nausea[55]
HCVIn vivoMarijuana cigarettes progression of liver fibrosis; increased disease severity clinical pathological survey of 204 HCV patients[56]
HIV-1In vitroΔ9-THC, CP-55,940, WIN-55,212 increased syn-cytia formation MT-2 cells (CB1 & CB2+) speculate cannabinoids enhance HIV-1 infection[57]
HIV-1In vitroanandamide increased adherence for monocytesuncoupled NO release, inhibited NOhuman saphenous vein or internal thoracic artery; speculate higher titers in vivo[58]
HIV-1 TatIn vitroWIN-55,212 reduced tat-induced cytotoxicityinhibited NOS-2 activityC6 rat glioma cell line[59]
HIV-1In vivoMarijuana cigarettes increased appetite insufficient numbers of individuals3 week trial[60]
HIV-1In vivoMarijuana cigarettesmRNA unchanged CD4+ and CD8+ cells unchanged3 week trial, placebo-controlled[61]
HIV-1 WIN-55,212inhibited expression CD4 and microglial cultures[62]
HIV-1In vivoTHCincreased viral replica-tion 50-fold decreased CD4 IFN-γ-producing cells, increased co-receptor expressionscid-Hu mouse model[63]
HIV-1 Gp120In vitro2-AG, CP55940 inhibited Ca+2-flux-induced substance P, decreased permeability model of BBB, co-culture of Human brain microvascular endothelial cells and astrocytes[64]
HIV-1In vivoWIN-55,212 dose-related hypothermia in mouse pre-optic anterior hypothalamus infusionWIN-55,212 is antagonist for SDF-1a/ CXCL12/ CXCR4 [HIV-1 coReceptor] pathwaymouse model for HIV-thermoreg-ulation by direct injection of WIN-55,212 to brain POAH center[65]
HIV-1 TatIn vitroCP55940, Δ9-THC CB2-dependent inhibition of U937 migration to Tat possible anti-inflammatory mechanismU937 cells in culture[66]
Legend: BDV, Borna disease virus; EBV, Epstein-Barr virus; HCV, Hepatitis C virus; HIV, Human immunodeficiency virus; HSV, Herpes simplex virus; HVS, Herpes virus samirii; KO, knock-out mice; KSHV, Kaposi's sarcoma herpes virus; L. monocytogenes, Listeria monocytogenes; MHV-68, Murine herpes virus-68; TMEV, Theiler's murine encephalomyelitis virus; VSV, Vesicular stomatitis virus.
Disease was more severe in HSV-2-infected guinea pigs which were treated with THC [29,30,32]. In HCV infections, clinical studies have shown a profound co-morbidity of recreational cannabinoid use, for disease progression [54,56]. One case report of Cowpox infection, a very rare human pathogen, indicated that recreational use of cannabinoids was associated with generalized infection and very poor immune responses to the virus [40].
In contrast, in those infections where host inflammatory responses are often associated with pathology, and not with clearance and recovery, cannabinoid treatment of hosts was beneficial. These included one mouse model of multiple sclerosis, the Theiler's murine encephalomyelocarditis virus (TMEV)-induced demyelinating disease (IDD), where progression towards the paralysis and disability were ameliorated [44,45,48] and in Borna disease virus (BDV) where neural progenitors were protected from proinflammatory cytokine-mediated damage [53] infections. TMEV-IDD is characterized by microglial activation in the spinal cord of mice and a T cell-mediated autoimmune demyelinating disease, triggered by the viral infection [42,67,68,69]. Persistent BDV infection of the central nervous system is associated with immunopathology associate with inflammation and production of pro-inflammatory cytokines, induction of NOS-2 in microglia, and breakdown of the blood-brain barrier [70,71,72,73]. In both BVD and TMEV-IDD, the targets for the anti-inflammatory effects of the cannabinoid treatment are lymphocytes and mononuclear cells.
Two excellent reviews of the impact of cannabinoids on bacterial, yeast, and protozoan infections were published in the same issue of Journal of Neuroimmunology [26,74]. These infections included Treponema pallidum (Syphilis), Legonella pneumophila (Legionnaires' disease), Staphylococci aureus and S. albus, Listeria monocytogenes, Candida albicans (Thrush), and Naegleria fowleri. Both reviews concluded that THC significantly reduced host resistance to infection of experimental animals, and speculated that similar host compromise would be found in man. In the more than 12 years since those reviews were published, additional findings have extended the serious consequences of cannabinoids on host responses to pathogens and opportunistic infections. Marijuana use is a risk factor for Mycobacterium tuberculosis (TB) infections [75,76,77]; this author speculates the suppression of host innate immune responses by THC contributes to the increased severity of TB in users. Similarly, more serious exacerbations central nervous system infection by Acanthamoeba among HIV-infected patients has been attributed to marijuana consumption [78], possibly by inhibiting macrophage chemotaxis [79]. However, the antiinflammatory effects of cannabinoids have been found to be beneficial in attenuating fever induced by bacterial endotoxin [65,80], inhibiting cytokine responses to Corynebacterium parvum endotoxin [81]. These drugs may also offer therapeutic efficacy in meningitis caused by Streptococcus pneumoniae [82] and in irritable bowel syndrome [83,84].
Cannabinoids may relieve pain and may induce hyperphagia, which could be beneficial in cancer [85,86]. However, these physiological characteristics are not relevant to most viral, bacterial fungal or parasitic infections, where the regulation of inflammation is central to controlling pathogen replication and immunopathology. However, the same anti-inflammatory properties of cannabinoids just described are detrimental to the host in handling the other infections. In most cases, a rapid and robust inflammatory response, associated with production of proinflammatory cytokines and effect T lymphocytes capable of eliminating infected cells is essential to recovery and survival.

3. Conclusions

Cannabinoids are profoundly anti-inflammatory and impair many Ca2+-dependent enzyme systems which are central to inflammatory and cell-autonomous antiviral responses. When viral-induced host responses lead to immunopathology, as is seen in a rodent model of multiple sclerosis, TMEV-IDD, or in a persistent infection of the central nervous system caused by a non-lytic virus, BDV, cannabinoid treatment was beneficial.
In all other virus infections, both in vitro and in vivo, cannabinoid treatment led to disease progression, increased pathology, and sometimes to host death. Therefore, in many clinical settings, including latent infections caused by HIV-1 or HSV-1, and persistent infection of the liver caused by HCV, cannabinoids lead to worsened disease outcome.

Acknowledgements

The generous support of the U.S. National Institutes of Health, both grants DC039746 and NS039746, enabled my lab to perform these studies, and to summarize the work of many other labs. Two former students, Ramon Antonio Herrera and Joseph H. Oved, contributed published [52] and unpublished data, provided stimulating discussions and critically read this manuscript.

References

  1. Bannenberg, G.L.; Chiang, N.; Ariel, A.; Arita, M.; Tjonahen, E.; Gotlinger, K.H.; Hong, S.; Serhan, C.N. Molecular circuits of resolution: Formation and actions of resolvins and protectins. J. Immunol. 2005, 174, 4345–4355. [Google Scholar]
  2. Machado, F.S.; Johndrow, J.E.; Esper, L.; Dias, A.; Bafica, A.; Serhan, C.N.; Aliberti, J. Anti-inflammatory actions of lipoxin A4 and aspirin-triggered lipoxin are SOCS-2 dependent. Nat. Med. 2006, 12, 330–334. [Google Scholar]
  3. Morita, M.; Yoshiki, F.; Nakane, A.; Okubo, Y.; Kudo, Y. Receptor- and calcium-dependent induced inositol 1,4,5-trisphosphate increases in PC12h cells as shown by fluorescence resonance energy transfer imaging. FEBS J. 2007, 274, 5147–5157. [Google Scholar]
  4. Vitale, N. Synthesis of fusogenic lipids through activation of phospholipase D1 by GTPases and the kinase RSK2 is required for calcium-regulated exocytosis in neuroendocrine cells. Biochem. Soc. Trans. 2010, 38, 167–171. [Google Scholar]
  5. Oh-hora, M.; Rao, A. The calcium/NFAT pathway: Role in development and function of regulatory T cells. Microbes Infect. 2009, 11, 612–619. [Google Scholar]
  6. Rao, A. Signaling to gene expression: Calcium, calcineurin and NFAT. Nat. Immunol. 2009, 10, 3–5. [Google Scholar]
  7. Knowles, R.G.; Moncada, S. Nitric oxide synthases in mammals. Biochem. J. 1994, 298, 249–258. [Google Scholar]
  8. Lopez-Jaramillo, P.; Teran, E.; Moncada, S. Calcium supplementation prevents pregnancy-induced hypertension by increasing the production of vascular nitric oxide. Med. Hypotheses 1995, 45, 68–72. [Google Scholar]
  9. Edelstein, C.L.; Yaqoob, M.M.; Schrier, R.W. The role of the calcium-dependent enzymes nitric oxide synthase and calpain in hypoxia-induced proximal tubule injury. Ren Fail. 1996, 18, 501–511. [Google Scholar]
  10. Reiss, C.S.; Komatsu, T. Does nitric oxide play a critical role in viral infections? J. Virol. 1998, 72, 4547–4551. [Google Scholar] [PubMed]
  11. Akaike, T.; Maeda, H. Nitric oxide and virus infection. Immunology 2000, 101, 300–308. [Google Scholar]
  12. Akuta, T.; Zaki, M.H.; Yoshitake, J.; Okamoto, T.; Akaike, T. Nitrative stress through formation of 8-nitroguanosine: Insights into microbial pathogenesis. Nitric. Oxide. 2006, 14, 101–108. [Google Scholar]
  13. Alvania, R.S.; Chen, X.; Ginty, D.D. Calcium signals control Wnt-dependent dendrite growth. Neuron 2006, 50, 813–815. [Google Scholar]
  14. Wayman, G.A.; Impey, S.; Marks, D.; Saneyoshi, T.; Grant, W.F.; Derkach, V.; Soderling, T.R. Activity-dependent dendritic arborization mediated by CaM-kinase I activation and enhanced CREB-dependent transcription of Wnt-2. Neuron 2006, 50, 897–909. [Google Scholar]
  15. Flaherty, M.P.; Dawn, B. Noncanonical Wnt11 signaling and cardiomyogenic differentiation. Trends Cardiovasc. Med. 2008, 18, 260–268. [Google Scholar]
  16. Caplan, M.J.; Seo-Mayer, P.; Zhang, L. Epithelial junctions and polarity: Complexes and kinases. Curr. Opin. Nephrol. Hypertens. 2008, 17, 506–512. [Google Scholar]
  17. Liu, J.O. Calmodulin-dependent phosphatase, kinases, and transcriptional corepressors involved in T-cell activation. Immunol. Rev. 2009, 228, 184–198. [Google Scholar] [CrossRef] [PubMed]
  18. Pontremoli, S.; Melloni, E. Extralysosomal protein degradation. Annu. Rev Biochem. 1986, 55, 455–481. [Google Scholar]
  19. Mellgren, R.L. Calcium-dependent proteases: An enzyme system active at cellular membranes? FASEB J. 1987, 1, 110–115. [Google Scholar] [PubMed]
  20. Dargelos, E.; Poussard, S.; Brule, C.; Daury, L.; Cottin, P. Calcium-dependent proteolytic system and muscle dysfunctions: a possible role of calpains in sarcopenia. Biochimie 2008, 90, 359–368. [Google Scholar]
  21. Consolo, M.; Amoroso, A.; Spandidos, D.A.; Mazzarino, M.C. Matrix metalloproteinases and their inhibitors as markers of inflammation and fibrosis in chronic liver disease (Review). Int. J. Mol. Med. 2009, 24, 143–152. [Google Scholar]
  22. Rojas, F.J.; Brush, M.; Moretti-Rojas, I. Calpain-calpastatin: A novel, complete calcium-dependent protease system in human spermatozoa. Mol. Hum. Reprod. 1999, 5, 520–526. [Google Scholar]
  23. Elli, L.; Bergamini, C.M.; Bardella, M.T.; Schuppan, D. Transglutaminases in inflammation and fibrosis of the gastrointestinal tract and the liver. Dig. Liver Dis. 2009, 41, 541–550. [Google Scholar]
  24. Reiss, C.S. Innate immunity in viral encephalitis. In Neurotropic Virus Infections.; Cambridge University Press: Cambridge, UK, 2008; pp. 265–291. [Google Scholar]
  25. Reiss, C.S. VSV infection elicits distinct host responses in the periphery and the brain. In RNA Viruses: Host Gene Responses to Infection; Yang, D., Ed.; World Scientific Publishing: Hackensack, NJ, USA, 2009; pp. 229–246. [Google Scholar]
  26. Klein, T.W.; Friedman, H.; Specter, S. Marijuana, immunity and infection. J. Neuroimmunol. 1998, 83, 102–115. [Google Scholar]
  27. Bani, D.; Mannaioni, G.; Passani, M.B.; Masini, E. Role of cannaboboids in the modulation f inflammatory processes. Pharmaceuticals 2010. submitted. [Google Scholar]
  28. Klein, T.W.; Cabral, G.A. Cannabinoid-induced immune suppression and modulation of antigen-presenting cells. J. Neuroimmune. Pharmacol. 2006, 1, 50–64. [Google Scholar]
  29. Morahan, P.S.; Klykken, P.C.; Smith, S.H.; Harris, L.S.; Munson, A.E. Effects of cannabinoids on host resistance to Listeria monocytogenes and herpes simplex virus. Infect. Immun. 1979, 23, 670–674. [Google Scholar]
  30. Mishkin, E.M.; Cabral, G.A. Delta-9-Tetrahydrocannabinol decreases host resistance to herpes simplex virus type 2 vaginal infection in the B6C3F1 mouse. J. Gen. Virol. 1985, 66, 2539–2549. [Google Scholar]
  31. Cabral, G.A.; Lockmuller, J.C.; Mishkin, E.M. Delta 9-tetrahydrocannabinol decreases alpha/beta interferon response to herpes simplex virus type 2 in the B6C3F1 mouse. Proc. Soc. Exp. Biol. Med. 1986, 181, 305–311. [Google Scholar]
  32. Cabral, G.A.; Mishkin, E.M.; Marciano-Cabral, F.; Coleman, P.; Harris, L.; Munson, A.E. Effect of delta 9-tetrahydrocannabinol on herpes simplex virus type 2 vaginal infection in the guinea pig. Proc. Soc. Exp. Biol. Med. 1986, 182, 181–186. [Google Scholar]
  33. Blevins, R.D.; Dumic, M.P. The effect of delta-9-tetrahydrocannabinol on herpes simplex virus replication. J. Gen. Virol. 1980, 49, 427–431. [Google Scholar]
  34. Cabral, G.A.; McNerney, P.J.; Mishkin, E.M. Delta-9-tetrahydrocannabinol enhances release of herpes simplex virus type 2. J. Gen. Virol. 1986, 67, 2017–2022. [Google Scholar]
  35. Cabral, G.A.; McNerney, P.J.; Mishkin, E.M. Delta-9-tetrahydrocannabinol inhibits the splenocyte proliferative response to herpes simplex virus type 2. Immunopharmacol. Immunotoxicol. 1987, 9, 361–370. [Google Scholar]
  36. Lancz, G.; Specter, S.; Brown, H.K. Suppressive effect of delta-9-tetrahydrocannabinol on herpes simplex virus infectivity in vitro. Proc. Soc. Exp. Biol. Med. 1991, 196, 401–404. [Google Scholar] [PubMed]
  37. Fischer-Stenger, K.; Updegrove, A.W.; Cabral, G.A. Delta 9-tetrahydrocannabinol decreases cytotoxic T lymphocyte activity to herpes simplex virus type 1-infected cells. Proc. Soc. Exp. Biol. Med. 1992, 200, 422–430. [Google Scholar]
  38. Medveczky, M.M.; Sherwood, T.A.; Klein, T.W.; Friedman, H.; Medveczky, P.G. Delta-9 tetrahydrocannabinol (THC) inhibits lytic replication of gamma oncogenic herpesviruses in vitro. BMC. Med. 2004, 2, 34. [Google Scholar] [CrossRef] [PubMed]
  39. Zhang, X.; Wang, J.F.; Kunos, G.; Groopman, J.E. Cannabinoid modulation of Kaposi's sarcoma-associated herpesvirus infection and transformation. Cancer Res. 2007, 67, 7230–7237. [Google Scholar]
  40. Huemer, H.P.; Himmelreich, A.; Honlinger, B.; Pavlic, M.; Eisendle, K.; Hopfl, R.; Rabl, W.; Czerny, C.P. "Recreational" drug abuse associated with failure to mount a proper antibody response after a generalised orthopoxvirus infection. Infection 2007, 35, 469–473. [Google Scholar] [CrossRef] [PubMed]
  41. Molina-Holgado, F.; Lledo, A.; Guaza, C. Anandamide suppresses nitric oxide and TNF-alpha responses to Theiler's virus or endotoxin in astrocytes. Neuroreport 1997, 8, 1929–1933. [Google Scholar]
  42. Oleszak, E.L.; Katsetos, C.D.; Kuzmak, J.; Varadhachary, A. Inducible nitric oxide synthase in Theiler's murine encephalomyelitis virus infection. J. Virol. 1997, 71, 3228–3235. [Google Scholar]
  43. Molina-Holgado, F.; Molina-Holgado, E.; Guaza, C. The endogenous cannabinoid anandamide potentiates interleukin-6 production by astrocytes infected with Theiler's murine encephalomyelitis virus by a receptor-mediated pathway. FEBS Lett. 1998, 433, 139–142. [Google Scholar]
  44. Croxford, J.L.; Miller, S.D. Immunoregulation of a viral model of multiple sclerosis using the synthetic cannabinoid R(+)WIN55,212. J. Clin. Invest. 2003, 111, 1231–1240. [Google Scholar]
  45. Mestre, L.; Correa, F.; Arevalo-Martin, A.; Molina-Holgado, E.; Valenti, M.; Ortar, G.; Di Marzo, V.; Guaza, C. Pharmacological modulation of the endocannabinoid system in a viral model of multiple sclerosis. J. Neurochem. 2005, 92, 1327–1339. [Google Scholar]
  46. Correa, F.; Mestre, L.; Docagne, F.; Guaza, C. Activation of cannabinoid CB2 receptor negatively regulates IL-12p40 production in murine macrophages: Role of IL-10 and ERK1/2 kinase signaling. Br. J. Pharmacol. 2005, 145, 441–448. [Google Scholar]
  47. Mestre, L.; Correa, F.; Docagne, F.; Clemente, D.; Guaza, C. The synthetic cannabinoid WIN 55,212-2 increases COX-2 expression and PGE2 release in murine brain-derived endothelial cells following Theiler's virus infection. Biochem. Pharmacol. 2006, 72, 869–880. [Google Scholar]
  48. Loria, F.; Petrosino, S.; Mestre, L.; Spagnolo, A.; Correa, F.; Hernangomez, M.; Guaza, C.; Di Marzo, V.; Docagne, F. Study of the regulation of the endocannabinoid system in a virus model of multiple sclerosis reveals a therapeutic effect of palmitoylethanolamide. Eur. J. Neurosci 2008, 28, 633–641. [Google Scholar]
  49. Mestre, L.; Docagne, F.; Correa, F.; Loria, F.; Hernangomez, M.; Borrell, J.; Guaza, C. A cannabinoid agonist interferes with the progression of a chronic model of multiple sclerosis by downregulating adhesion molecules. Mol. Cell. Neurosci. 2009, 40, 258–266. [Google Scholar]
  50. Buchweitz, J.P.; Karmaus, P.W.; Harkema, J.R.; Williams, K.J.; Kaminski, N.E. Modulation of airway responses to influenza A/PR/8/34 by Delta9-tetrahydrocannabinol in C57BL/6 mice. J. Pharmacol. Exp. Ther. 2007, 323, 675–683. [Google Scholar]
  51. Buchweitz, J.P.; Karmaus, P.W.; Williams, K.J.; Harkema, J.R.; Kaminski, N.E. Targeted deletion of cannabinoid receptors CB1 and CB2 produced enhanced inflammatory responses to influenza A/PR/8/34 in the absence and presence of Delta9-tetrahydrocannabinol. J. Leukoc. Biol. 2008, 83, 785–796. [Google Scholar]
  52. Herrera, R.A.; Oved, J.H.; Reiss, C.S. Disruption of the IFN-g-mediated antiviral activity in neurons: The role of Cannabinoids. Viral Immunol. 2008, 21, 141–152. [Google Scholar]
  53. Solbrig, M.V.; Hermanowicz, N. Cannabinoid rescue of striatal progenitor cells in chronic Borna disease viral encephalitis in rats. J. Neurovirol. 2008, 14, 252–260. [Google Scholar]
  54. Hezode, C.; Roudot-Thoraval, F.; Nguyen, S.; Grenard, P.; Julien, B.; Zafrani, E.S.; Pawlotsky, J.M.; Dhumeaux, D.; Lotersztajn, S.; Mallat, A. Daily cannabis smoking as a risk factor for progression of fibrosis in chronic hepatitis C. Hepatology 2005, 42, 63–71. [Google Scholar]
  55. Costiniuk, C.T.; Mills, E.; Cooper, C.L. Evaluation of oral cannabinoid-containing medications for the management of interferon and ribavirin-induced anorexia, nausea and weight loss in patients treated for chronic hepatitis C virus. Can. J. Gastroenterol. 2008, 22, 376–380. [Google Scholar]
  56. Ishida, J.H.; Peters, M.G.; Jin, C.; Louie, K.; Tan, V.; Bacchetti, P.; Terrault, N.A. Influence of cannabis use on severity of hepatitis C disease. Clin Gastroenterol. Hepatol. 2008, 6, 69–75. [Google Scholar]
  57. Noe, S.N.; Nyland, S.B.; Ugen, K.; Friedman, H.; Klein, T.W. Cannabinoid receptor agonists enhance syncytia formation in MT-2 cells infected with cell free HIV-1MN. Adv. Exp. Med. Biol. 1998, 437, 223–229. [Google Scholar]
  58. Stefano, G.B.; Salzet, M.; Bilfinger, T.V. Long-term exposure of human blood vessels to HIV gp120, morphine, and anandamide increases endothelial adhesion of monocytes: Uncoupling of nitric oxide release. J. Cardiovasc. Pharmacol. 1998, 31, 862–868. [Google Scholar] [CrossRef]
  59. Esposito, G.; Ligresti, A.; Izzo, A.A.; Bisogno, T.; Ruvo, M.; Di Rosa, M.; Di Marzo, V.; Iuvone, T. The endocannabinoid system protects rat glioma cells against HIV-1 Tat protein-induced cytotoxicity: Mechanism and regulation. J. Biol. Chem. 2002, 277, 50348–50354. [Google Scholar]
  60. Bredt, B.M.; Higuera-Alhino, D.; Shade, S.B.; Hebert, S.J.; McCune, J.M.; Abrams, D.I. Short-term effects of cannabinoids on immune phenotype and function in HIV-1-infected patients. J. Clin. Pharmacol. 2002, 42, 82S–89S. [Google Scholar]
  61. Abrams, D.I.; Hilton, J.F.; Leiser, R.J.; Shade, S.B.; Elbeik, T.A.; Aweeka, F.T.; Benowitz, N.L.; Bredt, B.M.; Kosel, B.; Aberg, J.A.; et al. Short-term effects of cannabinoids in patients with HIV-1 infection: A randomized, placebo-controlled clinical trial. Ann. Intern. Med. 2003, 139, 258–266. [Google Scholar] [PubMed]
  62. Peterson, P.K.; Gekker, G.; Hu, S.; Cabral, G.; Lokensgard, J.R. Cannabinoids and morphine differentially affect HIV-1 expression in CD4(+) lymphocyte and microglial cell cultures. J. Neuroimmunol. 2004, 147, 123–126. [Google Scholar]
  63. Roth, M.D.; Tashkin, D.P.; Whittaker, K.M.; Choi, R.; Baldwin, G.C. Tetrahydrocannabinol suppresses immune function and enhances HIV replication in the huPBL-SCID mouse. Life Sci. 2005, 77, 1711–1722. [Google Scholar]
  64. Lu, T.S.; Avraham, H.K.; Seng, S.; Tachado, S.D.; Koziel, H.; Makriyannis, A.; Avraham, S. Cannabinoids inhibit HIV-1 Gp120-mediated insults in brain microvascular endothelial cells. J. Immunol. 2008, 181, 6406–6416. [Google Scholar]
  65. Benamar, K.; Yondorf, M.; Meissler, J.J.; Geller, E.B.; Tallarida, R.J.; Eisenstein, T.K.; Adler, M.W. A novel role of cannabinoids: Implication in the fever induced by bacterial lipopolysaccharide. J. Pharmacol. Exp. Ther. 2007, 320, 1127–1133. [Google Scholar]
  66. Raborn, E.S.; Cabral, G.A. Cannabinoid inhibition of macrophage migration to the TAT protein of HIV-1 is linked to the CB2 cannabinoid receptor. J. Pharmacol. Exp. Ther. 2010. [Google Scholar] [CrossRef]
  67. Jakob, J.; Roos, R.P. Molecular determinants of Theiler's murine encephalomyelitis-induced disease. J. Neurovirol. 1996, 2, 70–77. [Google Scholar]
  68. Olson, J.K.; Miller, S.D. The innate immune response affects the development of the autoimmune response in Theiler's virus-induced demyelinating disease. J. Immunol. 2009, 182, 5712–5722. [Google Scholar]
  69. Villarreal, D.; Young, C.R.; Storts, R.; Ting, J.W.; Welsh, C.J. A comparison of the neurotropism of Theiler's virus and poliovirus in CBA mice. Microb. Pathog. 2006, 41, 149–156. [Google Scholar]
  70. Dietzschold, B.; Morimoto, K. Signaling pathways in virus-induced CNS inflammation. J. Neurovirol. 1997, 3 (Suppl. 1), S58–S59. [Google Scholar] [PubMed]
  71. Gosztonyi, G.; Ludwig, H. Borna disease--neuropathology and pathogenesis. Curr. Top. Microbiol. Immunol. 1995, 190, 39–73. [Google Scholar]
  72. Hooper, D.C.; Kean, R.B.; Scott, G.S.; Spitsin, S.V.; Mikheeva, T.; Morimoto, K.; Bette, M.; Rohrenbeck, A.M.; Dietzschold, B.; Weihe, E. The central nervous system inflammatory response to neurotropic virus infection is peroxynitrite dependent. J. Immunol. 2001, 167, 3470–3477. [Google Scholar]
  73. Rohrenbeck, A.M.; Bette, M.; Hooper, D.C.; Nyberg, F.; Eiden, L.E.; Dietzschold, B.; Weihe, E. Upregulation of COX-2 and CGRP expression in resident cells of the Borna disease virus-infected brain is dependent upon inflammation. Neurobiol. Dis. 1999, 6, 15–34. [Google Scholar]
  74. Cabral, G.A.; Dove Pettit, D.A. Drugs and immunity: Cannabinoids and their role in decreased resistance to infectious disease. J. Neuroimmunol. 1998, 83, 116–123. [Google Scholar]
  75. Munckhof, W.J.; Konstantinos, A.; Wamsley, M.; Mortlock, M.; Gilpin, C. A cluster of tuberculosis associated with use of a marijuana water pipe. Int. J Tuberc. Lung Dis. 2003, 7, 860–865. [Google Scholar]
  76. Han, B.; Gfroerer, J.C.; Colliver, J.D. Associations between duration of illicit drug use and health conditions: Results from the 2005-2007 national surveys on drug use and health. Ann. Epidemiol. 2010, 20, 289–297. [Google Scholar]
  77. Holtz, T.H.; Lancaster, J.; Laserson, K.F.; Wells, C.D.; Thorpe, L.; Weyer, K. Risk factors associated with default from multidrug-resistant tuberculosis treatment, South Africa, 1999–2001. Int. J. Tuberc. Lung Dis. 2006, 10, 649–655. [Google Scholar] [PubMed]
  78. Cabral, G.A.; Marciano-Cabral, F. Cannabinoid-mediated exacerbation of brain infection by opportunistic amebae. J. Neuroimmunol. 2004, 147, 127–130. [Google Scholar]
  79. Marciano-Cabral, F.; Raborn, E.S.; Martin, B.R.; Cabral, G.A. Delta-9-tetrahydrocannabinol, the major psychoactive component in marijuana, inhibits macrophage chemotaxis to Acanthamoeba. J. Eukaryot. Microbiol. 2006, 53 (Suppl. 1), S15–S17. [Google Scholar] [CrossRef] [PubMed]
  80. Benamar, K.; Yondorf, M.; Geller, E.B.; Eisenstein, T.K.; Adler, M.W. Physiological evidence for interaction between the HIV-1 co-receptor CXCR4 and the cannabinoid system in the brain. Br. J. Pharmacol. 2009, 157, 1225–1231. [Google Scholar]
  81. Smith, S.R.; Terminelli, C.; Denhardt, G. Modulation of cytokine responses in Corynebacterium parvum-primed endotoxemic mice by centrally administered cannabinoid ligands. Eur. J. Pharmacol. 2001, 425, 73–83. [Google Scholar]
  82. Bass, R.; Engelhard, D.; Trembovler, V.; Shohami, E. A novel nonpsychotropic cannabinoid, HU-211, in the treatment of experimental pneumococcal meningitis. J. Infect. Dis. 1996, 173, 735–738. [Google Scholar] [CrossRef] [PubMed]
  83. Storr, M.A.; Yuce, B.; Andrews, C.N.; Sharkey, K.A. The role of the endocannabinoid system in the pathophysiology and treatment of irritable bowel syndrome. Neurogastroenterol. Motil. 2008, 20, 857–868. [Google Scholar]
  84. Storr, M.A.; Keenan, C.M.; Emmerdinger, D.; Zhang, H.; Yuce, B.; Sibaev, A.; Massa, F.; Buckley, N.E.; Lutz, B.; Goke, B.; et al. Targeting endocannabinoid degradation protects against experimental colitis in mice: Involvement of CB1 and CB2 receptors. J. Mol. Med. 2008, 86, 925–936. [Google Scholar] [CrossRef] [PubMed]
  85. Elikkottil, J.; Gupta, P.; Gupta, K. The analgesic potential of cannabinoids. J. Opioid. Manag. 2009, 5, 341–357. [Google Scholar]
  86. Aggarwal, S.K.; Carter, G.T.; Sullivan, M.D.; ZumBrunnen, C.; Morrill, R.; Mayer, J.D. Medicinal use of cannabis in the United States: Historical perspectives, current trends, and future directions. J. Opioid. Manag. 2009, 5, 153–168. [Google Scholar] [PubMed]

Share and Cite

MDPI and ACS Style

Reiss, C.S. Cannabinoids and Viral Infections. Pharmaceuticals 2010, 3, 1873-1886. https://doi.org/10.3390/ph3061873

AMA Style

Reiss CS. Cannabinoids and Viral Infections. Pharmaceuticals. 2010; 3(6):1873-1886. https://doi.org/10.3390/ph3061873

Chicago/Turabian Style

Reiss, Carol Shoshkes. 2010. "Cannabinoids and Viral Infections" Pharmaceuticals 3, no. 6: 1873-1886. https://doi.org/10.3390/ph3061873

Article Metrics

Back to TopTop