Canna~Fangled Abstracts

Acute inflammation and pathogenesis of SARS-CoV-2 infection: Cannabidiol as a potential anti-inflammatory treatment?

By May 20, 2020May 30th, 2020No Comments

Logo of pheelsevier

[Epub ahead of print]
PMCID: PMC7239000

1. Acute inflammation and pathogenesis of SARS-CoV-2 infection

SARS-CoV-2 is a new beta coronavirus first reported in China with a 14-day incubation period []. Infected persons may be either asymptomatic carriers, during which time they can transmit the virus to others [], or develop mild disease involving cough and rhinitis, with or without mild pneumonia. In severe disease, persons develop dyspnea and hypoxia, accompanied by patchy infiltrates on chest x-ray within a few days. Individuals with severe infection by SARS-CoV-2 can also develop acute respiratory distress syndrome (ARDS). Other symptoms include fatigue, anorexia, myalgias and diarrhea []. In a proportion of individuals, critical illness develops and is characterized by respiratory failure, shock and multi-organ dysfunction []. Mortality estimates vary based on the study population examined, with mortality among persons requiring mechanical ventilation as high as 88% []. Severe illness has been seen in otherwise healthy persons of any age, but is most frequent in persons of advanced age with comorbidities such as cardiovascular and respiratory diseases and diabetes [].

SARS-CoV-2 infects types I and II pneumocytes via its receptor angiotensin converting enzyme (ACE)2 which is also the main receptor for SARS-CoV [,,]. Under healthy circumstances, bronchoalveolar lavage fluid is made up of predominantly alveolar macrophages (<80%) and lymphocytes (∼10−20%) []. Alveolar macrophages police the lungs for pathogens, eliminate senescent cells, engage in reparation of damaged tissue and enhance T-cell specific responses []. In addition, macrophages also facilitate neutrophil recruitment which contributes to pathogen clearance and further attraction of inflammatory cells [,]. Some coronaviruses including MERS-CoV [], SARS-CoV [], HCoV-229E [] and HCoV−OC43 [] can infect human macrophages and induce pro-inflammatory cytokine secretion. Although ACE2 in pulmonary tissues is expressed mainly by type I and II pneumocytes [], not only alveolar macrophages, but also tissue-resident CD169+ macrophages in spleens and lymph nodes, can be infected by SARS-CoV-2 [, , ]. Importantly, SARS-CoV-2 results in hyper-activation of lung macrophages as well as massive infiltration of pro-inflammatory monocyte-derived macrophages (MDMs) into small airways [,]. In lethal SARS-CoV infection of mice, disease progression depends on infiltration to the lungs of monocytes which produce interleukin (IL)-6, IL-1β and Tumor necrosis factor (TNF)-α []. Acute macrophage activation initiates a massive pro-inflammatory response including IL-6 and IL-1β which mediate the recruitment of neutrophils and cytotoxic CD8 T-cells into the lungs mucosal tissues []. Transcriptomic analysis, in addition to cytokine quantification in plasma and bronchoalveolar lavage fluid from SARS-CoV-2 patients demonstrate tremendous amounts of various cytokines, chemokines and soluble inflammatory mediators including tumor TNF-α, IL-6, IL-1β, IL-2R, IL-8, inducible protein (IP)-10, C-reactive protein and D-dimer [,], which culminates in a cytokine storm. In contrast to SARS-CoV, which is believed to elicit suboptimal interferon (IFN) responses, SARS-CoV-2 activates expression of many IFN-inducible genes having increased capacity for pathogenesis. Moreover, genes implicated in inflammation are overrepresented []. Furthermore, increased expression of chemokines critical for recruiting neutrophils (CXCL17) and monocytes (CCL2, CCL7) into the lungs is observed [].

T-cells also play an important role in lung mucosal immunity. During SARS-CoV and MERS-CoV infections, airway memory CD4 T-cells induce protection via the production of IFN-γ []. In SARS-CoV and MERS-CoV a rapid, specific memory CD8 T-cell response is needed to guard against infection [,]. In addition, SARS-CoV infection induces a potent and long-lived T-cell response in humans []. However, severe SARS-CoV-2 infection results in CD4 and CD8 T-cell lymphopenia and decreased INF-γ-producing T-cells [,]. Meanwhile, hyper-activation of both CD4 and CD8 T-cells (HLA-DR+CD38+ co-expression), an increase in CD8 T-cell cytotoxic granules, and increased frequencies of pro-inflammatory CCR6+ Th17 cells have been recently reported in SARS-CoV-2 patients [].

2. Cannabidiol to decrease SARS-CoV-2 associated inflammation

Cannabidiol (CBD) is a phytocannabinoid with various clinical applications and has proven efficacy for certain medical conditions, along with a favorable safety and tolerability profile [,]. Furthermore, unlike Δ9-tetrahydrocannabinol (THC), CBD does not induce any psychotropic effects, also making it desirable for therapeutic applications []. Cannabinoids can suppress immune activation and inflammatory cytokine production [], suggesting their potential for tempering excessive inflammation. Endocannabinoid receptors include CB1 and CB2. CB1 has higher expression in the central nervous system and a lesser expression on peripheral tissues, including the lungs []. Airway epithelial cells respond to both CB2 receptor-dependent and independent effects of cannabinoids []. CB2 is expressed by varieties of immune cells including circulating lymphocytes, monocytes and tissue mast cells and in lymphoid tissues [,]. Activation of CB2 receptor can suppress release of inflammatory IL-1, IL-6, IL-12 and TNF-α []. Constitutive production of endocannabinoids occurs by human lung resident macrophages, which is protective in acute and chronic inflammation, mostly via CB2 receptors []. Importantly, human lung resident macrophages also express both CB1 and CB2 receptors []. Agonists of CB2 have been shown to inhibit TNF-α from CD14+ monocytes and M1 macrophages, and increase expression of anti-inflammatory cytokine IL-10 []. CB2 agonists also induce anti-inflammatory FoxP3+ regulatory T-cells (Tregs) which produce TGF-β and IL-10 []. In addition, CBD has been shown to induce the differentiation of functional immunosupressive Tregs [].

In murine models of lung injury, CBD reduced lipopolysaccharide (LPS)-induced acute pulmonary inflammation [,]. In rat models of experimental asthma, CBD treatment reduced airway inflammation, as well as levels of serum IL-4, IL-5, IL-13, IL-6 and TNF-α, which are implicated in airway inflammation and fibrosis in asthma [,]. Moreover, CBD was able to directly suppress T-cell secretion of IL-1 and IFNγ []. In piglets with hypoxic-ischemic lung damage, CBD reduced histologic damage, decreased leukocyte infiltration and modulated IL-1 concentration in bronchoalveolar lavage fluid [], while in a rat model of sepsis, CBD reversed oxidative stress and reduced mortality []. In humans, cannabinoid use prevented induction of pro-inflammatory CD16+monocytes and production of IP-10, suggesting anti-inflammatory effects in humans []. In another human study, in addition to reduction of pro-inflammatory monocytes, heavy cannabis use was also associated with decreased frequencies of HLA-DR+CD38+activated CD4 and CD8 T-cells and frequencies of IL-10, IL-12 and TNF-α -producing antigen presenting cells compared to non-cannabis users []. The anti-inflammatory effects of cannabinoids are now under investigation in clinical trials, as such, our team in now conducting a clinical trial in the context of HIV infection [].

Therefore, as SARS-CoV2 induces significant damage through pro-inflammatory cytokine storm mediated by macrophages and other immune cells and based on the fact that CBD has broad anti-inflammatory properties, CBD might represent as a potential anti-inflammatory therapeutic approach against SARS-CoV2-induced inflammation. In this regard, first a deeper understanding of the specific effects of SARS-CoV2 on human macrophages and T-cell physiology and immunological functions is needed. As CBD is already a therapeutic agent used in clinical medicine and has a favorable safety profile, the results of in vitro and animal model proof-of-concept studies would provide the necessary supporting evidence required before embarking on costly and labor-intensive clinical trials.

Declaration of Competing Interest

Tilray Inc. will provide study medication for use in a clinical trial the authors will be conducting on safety, tolerability and efficacy of cannabinoids in people living with HIV (CIHR Canadian HIV Trials Pilot Study 028). There are no conflict of interests to declare regarding the publication of this specific paper and no funding was received for its preparation.

Acknowledgement

CTC holds a Fonds de recherche du Québec-Santé (FRQ-S) Junior 1 research salary award. MAJ holds the CIHR Canada Research Chair tier 2 in Immuno-virology.

Biographies

An external file that holds a picture, illustration, etc. Object name is fx1_lrg.jpg

Cecilia Costiniuk completed her medical training at McMaster University followed by a residency in internal medicine and fellowship training in infectious diseases at the University of Ottawa. After completing her MSc in Microbiology and Immunology in Ottawa, she then pursued post-doctoral training at the KwaZulu-Natal Research Institute for TB and HIV in Durban, South Africa. She joined McGill University in 2014, and she is currently an Associate Professor of Medicine in the Division of Infectious Diseases and Chronic Viral Illness Service of the McGill University Health Centre. Dr Costiniuk also holds a Chercheur-boursier-clinicien Junior 1 salary award from the FRQ-S As a clinician investigator, she leads a research program focused on pulmonary immunity and inflammation in the context of HIV infection. Her interest lies also in exploring the therapeutic potential of cannabinoids for various conditions involving excessive inflammation, including people living with HIV, in the context of well-designed studies.

An external file that holds a picture, illustration, etc. Object name is fx2_lrg.jpg

Mohammad-Ali Jenabian is an Associate Professor and the holder of the Canada Research Chair in Immnuo-Virology at the University of Quebec in Montreal (UQAM). He earned his D.V.M. degree in Veterinary Medicine in Iran. He then began Ph.D. studies in Virology at the Pierre and Marie Curie University in Paris. During his first postdoctoral training he furthered his expertise in HIV fundamental immunology at INSERM U955 in Paris. He also performed a second postdoctoral fellow at Research Institute of the McGill University Health Centre, Montreal, in HIV clinical immunology. His laboratory is now focused on pulmonary and intestinal mucosal immunity during HIV infection as well as accelerated aging in people living with HIV. Furthermore, his lab is also studying the anti-inflammatory properties of cannabinoids.

References

1. Zhu N., Zhang D., Wang W., Li X., Yang B., Song J., Zhao X., Huang B., Shi W., Lu R., Niu P., Zhan F., Ma X., Wang D., Xu W., Wu G., Gao G.F., Tan W. I. China novel coronavirus, T. research, a novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020;382(8):727–733. [PMC free article] [PubMed] []
2. Arons M.M., Hatfield K.M., Reddy S.C., Kimball A., James A., Jacobs J.R., Taylor J., Spicer K., Bardossy A.C., Oakley L.P., Tanwar S., Dyal J.W., Harney J., Chisty Z., Bell J.M., Methner M., Paul P., Carlson C.M., McLaughlin H.P., Thornburg N., Tong S., Tamin A., Tao Y., Uehara A., Harcourt J., Clark S., Brostrom-Smith C., Page L.C., Kay M., Lewis J., Montgomery P., Stone N.D., Clark T.A., Honein M.A., Duchin J.S., Jernigan J.A. Presymptomatic SARS-CoV-2 infections and transmission in a skilled nursing facility. N. Engl. J. Med. 2020 []
3. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., Zhang L., Fan G., Xu J., Gu X., Cheng Z., Yu T., Xia J., Wei Y., Wu W., Xie X., Yin W., Li H., Liu M., Xiao Y., Gao H., Guo L., Xie J., Wang G., Jiang R., Gao Z., Jin Q., Wang J., Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. [PMC free article] [PubMed] []
4. Richardson S., Hirsch J.S., Narasimhan M., Crawford J.M., McGinn T., Davidson K.W., C.-R.C. and the Northwell, Barnaby D.P., Becker L.B., Chelico J.D., Cohen S.L., Cookingham J., Coppa K., Diefenbach M.A., Dominello A.J., Duer-Hefele J., Falzon L., Gitlin J., Hajizadeh N., Harvin T.G., Hirschwerk D.A., Kim E.J., Kozel Z.M., Marrast L.M., Mogavero J.N., Osorio G.A., Qiu M., Zanos T.P. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City Area. JAMA. 2020 []
5. Mason R.J. Pathogenesis of COVID-19 from a cell biologic perspective. Eur. Respir. J. 2020 []
6. Hoffmann M., Kleine-Weber H., Schroeder S., Kruger N., Herrler T., Erichsen S., Schiergens T.S., Herrler G., Wu N.H., Nitsche A., Muller M.A., Drosten C., Pohlmann S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271–280. e8. [PMC free article] [PubMed] []
7. Yu Y.R., Hotten D.F., Malakhau Y., Volker E., Ghio A.J., Noble P.W., Kraft M., Hollingsworth J.W., Gunn M.D., Tighe R.M. Flow cytometric analysis of myeloid cells in human blood, Bronchoalveolar Lavage, and lung tissues. Am. J. Respir. Cell Mol. Biol. 2016;54(1):13–24. [PMC free article] [PubMed] []
8. Hussell T., Bell T.J. Alveolar macrophages: plasticity in a tissue-specific context. Nat. Rev. Immunol. 2014;14(2):81–93. [PubMed] []
9. Vardhana S.A., Wolchok J.D. The many faces of the anti-COVID immune response. J. Exp. Med. 2020;217(6) []
10. Hasenberg M., Stegemann-Koniszewski S., Gunzer M. Cellular immune reactions in the lung. Immunol. Rev. 2013;251(1):189–214. [PubMed] []
11. Arabi Y.M., Balkhy H.H., Hayden F.G., Bouchama A., Luke T., Baillie J.K., Al-Omari A., Hajeer A.H., Senga M., Denison M.R., Nguyen-Van-Tam J.S., Shindo N., Bermingham A., Chappell J.D., Van Kerkhove M.D., Fowler R.A. Middle east respiratory syndrome. N. Engl. J. Med. 2017;376(6):584–594. [PMC free article][PubMed] []
12. Yilla M., Harcourt B.H., Hickman C.J., McGrew M., Tamin A., Goldsmith C.S., Bellini W.J., Anderson L.J. SARS-coronavirus replication in human peripheral monocytes/macrophages. Virus Res. 2005;107(1):93–101. [PMC free article] [PubMed] []
13. Funk C.J., Wang J., Ito Y., Travanty E.A., Voelker D.R., Holmes K.V., Mason R.J. Infection of human alveolar macrophages by human coronavirus strain 229E. J. Gen. Virol. 2012;93(Pt 3):494–503. [PMC free article] [PubMed] []
14. Collins A.R. Human macrophages are susceptible to coronavirus OC43. Adv. Exp. Med. Biol. 1998;440:635–639. [PubMed] []
15. Qi F., Qian S., Zhang S., Zhang Z. Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses. Biochem. Biophys. Res. Commun. 2020;526(1):135–140. [PMC free article] [PubMed] []
16. Chu H., Chan J.F., Wang Y., Yuen T.T., Chai Y., Hou Y., Shuai H., Yang D., Hu B., Huang X., Zhang X., Cai J.P., Zhou J., Yuan S., Kok K.H., To K.K., Chan I.H., Zhang A.J., Sit K.Y., Au W.K., Yuen K.Y. Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19. Clin. Infect. Dis. 2020 []
17. chen y., Feng Z., Diao B., Wang R., Wang G., Wang C., Tan Y., Liu L., Wang C., Liu Y., Liu Y., Yuan Z., Ren L., Wu Y. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes. medRxiv. 2020 2020.03.27.20045427. []
18. Park M.D. Macrophages: a trojan horse in COVID-19? Nat. Rev. Immunol. 2020[]
19. Zhang W., Zhao Y., Zhang F., Wang Q., Li T., Liu Z., Wang J., Qin Y., Zhang X., Yan X., Zeng X., Zhang S. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China. Clin. Immunol. 2020;214:108393. [PMC free article][PubMed] []
20. Cao X. COVID-19: immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020;20(5):269–270. [PMC free article] [PubMed] []
21. Channappanavar R., Fehr A.R., Vijay R., Mack M., Zhao J., Meyerholz D.K., Perlman S. Dysregulated Type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe. 2016;19(2):181–193. [PMC free article] [PubMed] []
22. Giamarellos-Bourboulis E.J., Netea M.G., Rovina N., Akinosoglou K., Antoniadou A., Antonakos N., Damoraki G., Gkavogianni T., Adami M.E., Katsaounou P., Ntaganou M., Kyriakopoulou M., Dimopoulos G., Koutsodimitropoulos I., Velissaris D., Koufargyris P., Karageorgos A., Katrini K., Lekakis V., Lupse M., Kotsaki A., Renieris G., Theodoulou D., Panou V., Koukaki E., Koulouris N., Gogos C., Koutsoukou A. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020 []
23. Xiong Y., Liu Y., Cao L., Wang D., Guo M., Jiang A., Guo D., Hu W., Yang J., Tang Z., Wu H., Lin Y., Zhang M., Zhang Q., Shi M., Liu Y., Zhou Y., Lan K., Chen Y. Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg. Microbes Infect. 2020;9(1):761–770.[PMC free article] [PubMed] []
24. Zhuo Zhou L.R., Li Zhang, Zhong Jiaxin, Xiao Yan, Jia Zhilong, Li Guo, Yang Jing, Wang Chun, Jiang Shuai, Yang Donghong, Zhang Guoliang, Li Hongru, Chen Fuhui, Yu Xu, Chen Mingwei, Gao Zhancheng, Yang Jian, Dong Jie, Bo Liu, Zhang Xiannian, Wang Weidong, He Kunlun, Qi Jin, Li Mingkun, Wang Jianwei. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020 doi: 10.1016/j.chom.2020.04.017. In Press, PMID: 32407669, PMCID: PMC7196896. [CrossRef] []
25. Zhao J., Zhao J., Mangalam A.K., Channappanavar R., Fett C., Meyerholz D.K., Agnihothram S., Baric R.S., David C.S., Perlman S. Airway memory CD4(+) t cells mediate protective immunity against emerging respiratory coronaviruses. Immunity. 2016;44(6):1379–1391. [PMC free article] [PubMed] []
26. Channappanavar R., Fett C., Zhao J., Meyerholz D.K., Perlman S. Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J. Virol. 2014;88(19):11034–11044. [PMC free article][PubMed] []
27. Yang L.T., Peng H., Zhu Z.L., Li G., Huang Z.T., Zhao Z.X., Koup R.A., Bailer R.T., Wu C.Y. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin. Immunol. 2006;120(2):171–178. [PMC free article] [PubMed] []
28. Chen G., Wu D., Guo W., Cao Y., Huang D., Wang H., Wang T., Zhang X., Chen H., Yu H., Zhang X., Zhang M., Wu S., Song J., Chen T., Han M., Li S., Luo X., Zhao J., Ning Q. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020 []
29. Xu Z., Shi L., Wang Y., Zhang J., Huang L., Zhang C., Liu S., Zhao P., Liu H., Zhu L., Tai Y., Bai C., Gao T., Song J., Xia P., Dong J., Zhao J., Wang F.S. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020;8(4):420–422. [PMC free article] [PubMed] []
30. Devinsky O., Patel A.D., Cross J.H., Villanueva V., Wirrell E.C., Privitera M., Greenwood S.M., Roberts C., Checketts D., VanLandingham K.E., Zuberi S.M., Group G.S. Effect of Cannabidiol on drop seizures in the lennox-gastaut syndrome. N. Engl. J. Med. 2018;378(20):1888–1897. [PubMed] []
31. Larsen C., Shahinas J. Dosage, efficacy and safety of cannabidiol administration in adults: a systematic review of human trials. J. Clin. Med. Res. 2020;12(3):129–141.[PMC free article] [PubMed] []
32. Costiniuk C.T., Jenabian M.A. Cannabinoids and inflammation: implications for people living with HIV. AIDS. 2019;33(15):2273–2288. [PubMed] []
33. Galiegue S., Mary S., Marchand J., Dussossoy D., Carriere D., Carayon P., Bouaboula M., Shire D., Le Fur G., Casellas P. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur. J. Biochem. 1995;232(1):54–61. [PubMed] []
34. Sarafian T., Montes C., Harui A., Beedanagari S.R., Kiertscher S., Stripecke R., Hossepian D., Kitchen C., Kern R., Belperio J., Roth M.D. Clarifying CB2 receptor-dependent and independent effects of THC on human lung epithelial cells. Toxicol. Appl. Pharmacol. 2008;231(3):282–290. [PMC free article] [PubMed] []
35. Martin-Fontecha M., Angelina A., Ruckert B., Rueda-Zubiaurre A., Martin-Cruz L., van de Veen W., Akdis M., Ortega-Gutierrez S., Lopez-Rodriguez M.L., Akdis C.A., Palomares O. A fluorescent probe to unravel functional features of cannabinoid receptor CB1 in human blood and tonsil immune system cells. Bioconjug. Chem. 2018;29(2):382–389. [PubMed] []
36. Nichols J.M., Kaplan B.L.F. Immune responses regulated by cannabidiol. Cannabis Cannabinoid Res. 2020;5(1):12–31. [PMC free article] [PubMed] []
37. Gertsch J. Editorial: Lung macrophages high on cannabinoids: jamming PAMs and taming TAMs? J. Leukoc. Biol. 2016;99(4):518–520. [PubMed] []
38. Staiano R.I., Loffredo S., Borriello F., Iannotti F.A., Piscitelli F., Orlando P., Secondo A., Granata F., Lepore M.T., Fiorelli A., Varricchi G., Santini M., Triggiani M., Di Marzo V., Marone G. Human lung-resident macrophages express CB1 and CB2 receptors whose activation inhibits the release of angiogenic and lymphangiogenic factors. J. Leukoc. Biol. 2016;99(4):531–540. [PMC free article] [PubMed] []
39. Gentili M., Ronchetti S., Ricci E., Di Paola R., Gugliandolo E., Cuzzocrea S., Bereshchenko O., Migliorati G., Riccardi C. Selective CB2 inverse agonist JTE907 drives T cell differentiation towards a Treg cell phenotype and ameliorates inflammation in a mouse model of inflammatory bowel disease. Pharmacol. Res. 2019;141:21–31.[PubMed] []
40. Dhital S., Stokes J.V., Park N., Seo K.S., Kaplan B.L. Cannabidiol (CBD) induces functional Tregs in response to low-level T cell activation. Cell. Immunol. 2017;312:25–34. [PMC free article] [PubMed] []
41. Ribeiro A., Ferraz-de-Paula V., Pinheiro M.L., Vitoretti L.B., Mariano-Souza D.P., Quinteiro-Filho W.M., Akamine A.T., Almeida V.I., Quevedo J., Dal-Pizzol F., Hallak J.E., Zuardi A.W., Crippa J.A., Palermo-Neto J. Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A(2A) receptor. Eur. J. Pharmacol. 2012;678(1-3):78–85.[PubMed] []
42. Ribeiro A., Almeida V.I., Costola-de-Souza C., Ferraz-de-Paula V., Pinheiro M.L., Vitoretti L.B., Gimenes-Junior J.A., Akamine A.T., Crippa J.A., Tavares-de-Lima W., Palermo-Neto J. Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol. Immunotoxicol. 2015;37(1):35–41. [PubMed] []
43. Vuolo F., Petronilho F., Sonai B., Ritter C., Hallak J.E., Zuardi A.W., Crippa J.A., Dal-Pizzol F. Evaluation of serum cytokines levels and the role of cannabidiol treatment in animal model of asthma. Mediators Inflamm. 2015;2015 []
44. Vuolo F., Abreu S.C., Michels M., Xisto D.G., Blanco N.G., Hallak J.E., Zuardi A.W., Crippa J.A., Reis C., Bahl M., Pizzichinni E., Maurici R., Pizzichinni M.M.M., Rocco P.R.M., Dal-Pizzol F. Cannabidiol reduces airway inflammation and fibrosis in experimental allergic asthma. Eur. J. Pharmacol. 2019;843:251–259. [PubMed] []
45. Kaplan B.L., Springs A.E., Kaminski N.E. The profile of immune modulation by cannabidiol (CBD) involves deregulation of nuclear factor of activated T cells (NFAT) Biochem. Pharmacol. 2008;76(6):726–737. [PMC free article] [PubMed] []
46. Arruza L., Pazos M.R., Mohammed N., Escribano N., Lafuente H., Santos M., Alvarez-Diaz F.J., Hind W., Martinez-Orgado J. Cannabidiol reduces lung injury induced by hypoxic-ischemic brain damage in newborn piglets. Pediatr. Res. 2017;82(1):79–86.[PubMed] []
47. Cassol O.J., Jr., Comim C.M., Silva B.R., Hermani F.V., Constantino L.S., Felisberto F., Petronilho F., Hallak J.E., De Martinis B.S., Zuardi A.W., Crippa J.A., Quevedo J., Dal-Pizzol F. Treatment with cannabidiol reverses oxidative stress parameters, cognitive impairment and mortality in rats submitted to sepsis by cecal ligation and puncture. Brain Res. 2010;1348:128–138. [PubMed] []
48. Rizzo M.D., Crawford R.B., Henriquez J.E., Aldhamen Y.A., Gulick P., Amalfitano A., Kaminski N.E. HIV-infected cannabis users have lower circulating CD16+ monocytes and IFN-gamma-inducible protein 10 levels compared with nonusing HIV patients. AIDS. 2018;32(4):419–429. [PMC free article] [PubMed] []
49. Manuzak J.A., Gott T.M., Kirkwood J.S., Coronado E., Hensley-McBain T., Miller C., Cheu R.K., Collier A.C., Funderburg N.T., Martin J.N., Wu M.C., Isoherranen N., Hunt P.W., Klatt N.R. Heavy Cannabis use associated with reduction in activated and inflammatory immune cell frequencies in antiretroviral therapy-treated human immunodeficiency virus-infected individuals. Clin. Infect. Dis. 2018;66(12):1872–1882.[PMC free article] [PubMed] []
50. Costiniuk C.T., Saneei Z., Routy J.P., Margolese S., Mandarino E., Singer J., Lebouche B., Cox J., Szabo J., Brouillette M.J., Klein M.B., Chomont N., Jenabian M.A. Oral cannabinoids in people living with HIV on effective antiretroviral therapy: CTN PT028-study protocol for a pilot randomised trial to assess safety, tolerability and effect on immune activation. BMJ Open. 2019;9(1) []

doi: 10.1016/j.cytogfr.2020.05.008. Online ahead of print.

Affiliations

Conflict of interest statement

Declaration of Competing Interest Tilray Inc. will provide study medication for use in a clinical trial the authors will be conducting on safety, tolerability and efficacy of cannabinoids in people living with HIV (CIHR Canadian HIV Trials Pilot Study 028). There are no conflict of interests to declare regarding the publication of this specific paper and no funding was received for its preparation.

Leave a Reply