Skip to main content
Top

11-04-2024 | Human Immunodeficiency Virus | Review

The Impact of Cannabis Use on Cognition in People with HIV: Evidence of Function-Dependent Effects and Mechanisms from Clinical and Preclinical Studies

Authors: Samantha M. Ayoub, Breanna M. Holloway, Alannah H. Miranda, Benjamin Z. Roberts, Jared W. Young, Arpi Minassian, Ronald J. Ellis

Published in: Current HIV/AIDS Reports

Login to get access

Abstract

Purpose of Review

Cannabis may have beneficial anti-inflammatory effects in people with HIV (PWH); however, given this population’s high burden of persisting neurocognitive impairment (NCI), clinicians are concerned they may be particularly vulnerable to the deleterious effects of cannabis on cognition. Here, we present a systematic scoping review of clinical and preclinical studies evaluating the effects of cannabinoid exposure on cognition in HIV.

Recent Findings

Results revealed little evidence to support a harmful impact of cannabis use on cognition in HIV, with few eligible preclinical data existing. Furthermore, the beneficial/harmful effects of cannabis use observed on cognition were function-dependent and confounded by several factors (e.g., age, frequency of use).

Summary

Results are discussed alongside potential mechanisms of cannabis effects on cognition in HIV (e.g., anti-inflammatory), and considerations are outlined for screening PWH that may benefit from cannabis interventions. We further highlight the value of accelerating research discoveries in this area by utilizing translatable cross-species tasks to facilitate comparisons across human and animal work.
Appendix
Available only for authorised users
Literature
1.
go back to reference Thames AD, Becker BW, Marcotte TD, Hines LJ, Foley JM, Ramezani A, et al. Depression, cognition, and self-appraisal of functional abilities in HIV: an examination of subjective appraisal versus objective performance. Clin Neuropsychol. 2011;25(2):224–43.PubMedPubMedCentralCrossRef Thames AD, Becker BW, Marcotte TD, Hines LJ, Foley JM, Ramezani A, et al. Depression, cognition, and self-appraisal of functional abilities in HIV: an examination of subjective appraisal versus objective performance. Clin Neuropsychol. 2011;25(2):224–43.PubMedPubMedCentralCrossRef
2.
go back to reference Knippels HM, Goodkin K, Weiss JJ, Wilkie FL, Antoni MH. The importance of cognitive self-report in early HIV-1 infection: validation of a cognitive functional status subscale. AIDS. 2002;16(2):259–67.PubMedCrossRef Knippels HM, Goodkin K, Weiss JJ, Wilkie FL, Antoni MH. The importance of cognitive self-report in early HIV-1 infection: validation of a cognitive functional status subscale. AIDS. 2002;16(2):259–67.PubMedCrossRef
4.
go back to reference Nakao A, Yamanouchi J, Takenaka K, Takada K. The Iowa Gambling Task on HIV-infected subjects. J Infect Chemother. 2020;26(3):240–4.PubMedCrossRef Nakao A, Yamanouchi J, Takenaka K, Takada K. The Iowa Gambling Task on HIV-infected subjects. J Infect Chemother. 2020;26(3):240–4.PubMedCrossRef
5.
go back to reference Hinkin CH, Castellon SA, Hardy DJ, Granholm E, Siegle G. Computerized and traditional stroop task dysfunction in HIV-1 infection. Neuropsychology. 1999;13(2):306–16.PubMedCrossRef Hinkin CH, Castellon SA, Hardy DJ, Granholm E, Siegle G. Computerized and traditional stroop task dysfunction in HIV-1 infection. Neuropsychology. 1999;13(2):306–16.PubMedCrossRef
6.
go back to reference Hinkin CH, Hardy DJ, Mason KI, Castellon SA, Lam MN, Stefaniak M, et al. Verbal and spatial working memory performance among HIV-infected adults. J Int Neuropsychol Soc. 2002;8(4):532–8.PubMedCrossRef Hinkin CH, Hardy DJ, Mason KI, Castellon SA, Lam MN, Stefaniak M, et al. Verbal and spatial working memory performance among HIV-infected adults. J Int Neuropsychol Soc. 2002;8(4):532–8.PubMedCrossRef
7.
go back to reference Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, Leblanc S, et al. HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J Neurovirol. 2011;17(1):3–16.PubMedCrossRef Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, Leblanc S, et al. HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J Neurovirol. 2011;17(1):3–16.PubMedCrossRef
8.
go back to reference Wang Y, Liu M, Lu Q, Farrell M, Lappin JM, Shi J, et al. Global prevalence and burden of HIV-associated neurocognitive disorder: a meta-analysis. Neurology. 2020;95(19):e2610–21.PubMedCrossRef Wang Y, Liu M, Lu Q, Farrell M, Lappin JM, Shi J, et al. Global prevalence and burden of HIV-associated neurocognitive disorder: a meta-analysis. Neurology. 2020;95(19):e2610–21.PubMedCrossRef
9.
go back to reference Wei J, Hou J, Su B, Jiang T, Guo C, Wang W, et al. The prevalence of Frascati-criteria-based HIV-associated neurocognitive disorder (HAND) in HIV-infected adults: a systematic review and meta-analysis. Front Neurol. 2020;11:581346.PubMedPubMedCentralCrossRef Wei J, Hou J, Su B, Jiang T, Guo C, Wang W, et al. The prevalence of Frascati-criteria-based HIV-associated neurocognitive disorder (HAND) in HIV-infected adults: a systematic review and meta-analysis. Front Neurol. 2020;11:581346.PubMedPubMedCentralCrossRef
10.
go back to reference Korten V, Ay U, Hari E, Tigen Tukenmez E, Gencer S, Akca Kalem S, et al. Prevalence of HIV-associated neurocognitive disorder (HAND) in Turkey and assessment of Addenbrooke’s Cognitive Examination Revised (ACE-R) test as a screening tool. HIV Med. 2021;22(1):60–6.PubMedCrossRef Korten V, Ay U, Hari E, Tigen Tukenmez E, Gencer S, Akca Kalem S, et al. Prevalence of HIV-associated neurocognitive disorder (HAND) in Turkey and assessment of Addenbrooke’s Cognitive Examination Revised (ACE-R) test as a screening tool. HIV Med. 2021;22(1):60–6.PubMedCrossRef
11.
go back to reference Flatt A, Gentry T, Kellett-Wright J, Eaton P, Joseph M, Urasa S, et al. Prevalence and 1-year incidence of HIV-associated neurocognitive disorder (HAND) in adults aged >/=50 years attending standard HIV clinical care in Kilimanjaro Tanzania. Int Psychogeriatr. 2023;35(7):339–50.PubMedCrossRef Flatt A, Gentry T, Kellett-Wright J, Eaton P, Joseph M, Urasa S, et al. Prevalence and 1-year incidence of HIV-associated neurocognitive disorder (HAND) in adults aged >/=50 years attending standard HIV clinical care in Kilimanjaro Tanzania. Int Psychogeriatr. 2023;35(7):339–50.PubMedCrossRef
12.
go back to reference Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology. 2007;69(18):1789–99.PubMedCrossRef Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology. 2007;69(18):1789–99.PubMedCrossRef
13.
go back to reference • Dastgheyb RM, Sacktor N, Franklin D, Letendre S, Marcotte T, Heaton R, et al. Cognitive trajectory phenotypes in human immunodeficiency virus-infected patients. J Acquir Immune Defic Syndr. 2019;82(1):61–70 (This work showed that a specific cognitive impairment profile (deficits in executive function, learning, and processing-speed) was less likely in cannabis-using versus cannabis-abstinent PWH. However, other impairment profiles incorporating these domains (e.g., the learning and memory profile) were not associated with CU. These data highlight the necessity of testing discrete cognitive domain functions in PWH, and the importance of interpreting global cognition scores with caution). • Dastgheyb RM, Sacktor N, Franklin D, Letendre S, Marcotte T, Heaton R, et al. Cognitive trajectory phenotypes in human immunodeficiency virus-infected patients. J Acquir Immune Defic Syndr. 2019;82(1):61–70 (This work showed that a specific cognitive impairment profile (deficits in executive function, learning, and processing-speed) was less likely in cannabis-using versus cannabis-abstinent PWH. However, other impairment profiles incorporating these domains (e.g., the learning and memory profile) were not associated with CU. These data highlight the necessity of testing discrete cognitive domain functions in PWH, and the importance of interpreting global cognition scores with caution).
14.
go back to reference Rubin LH, Saylor D, Nakigozi G, Nakasujja N, Robertson K, Kisakye A, et al. Heterogeneity in neurocognitive change trajectories among people with HIV starting antiretroviral therapy in Rakai Uganda. J Neurovirol. 2019;25(6):800–13.PubMedPubMedCentralCrossRef Rubin LH, Saylor D, Nakigozi G, Nakasujja N, Robertson K, Kisakye A, et al. Heterogeneity in neurocognitive change trajectories among people with HIV starting antiretroviral therapy in Rakai Uganda. J Neurovirol. 2019;25(6):800–13.PubMedPubMedCentralCrossRef
15.
go back to reference Dastgheyb RM, Buchholz AS, Fitzgerald KC, Xu Y, Williams DW, Springer G, et al. Patterns and predictors of cognitive function among virally suppressed women with HIV. Front Neurol. 2021;12:604984.PubMedPubMedCentralCrossRef Dastgheyb RM, Buchholz AS, Fitzgerald KC, Xu Y, Williams DW, Springer G, et al. Patterns and predictors of cognitive function among virally suppressed women with HIV. Front Neurol. 2021;12:604984.PubMedPubMedCentralCrossRef
16.
go back to reference Rippeth JD, Heaton RK, Carey CL, Marcotte TD, Moore DJ, Gonzalez R, et al. Methamphetamine dependence increases risk of neuropsychological impairment in HIV infected persons. J Int Neuropsychol Soc. 2004;10(1):1–14.PubMedCrossRef Rippeth JD, Heaton RK, Carey CL, Marcotte TD, Moore DJ, Gonzalez R, et al. Methamphetamine dependence increases risk of neuropsychological impairment in HIV infected persons. J Int Neuropsychol Soc. 2004;10(1):1–14.PubMedCrossRef
17.
go back to reference Rothlind JC, Greenfield TM, Bruce AV, Meyerhoff DJ, Flenniken DL, Lindgren JA, et al. Heavy alcohol consumption in individuals with HIV infection: effects on neuropsychological performance. J Int Neuropsychol Soc. 2005;11(1):70–83.PubMedPubMedCentralCrossRef Rothlind JC, Greenfield TM, Bruce AV, Meyerhoff DJ, Flenniken DL, Lindgren JA, et al. Heavy alcohol consumption in individuals with HIV infection: effects on neuropsychological performance. J Int Neuropsychol Soc. 2005;11(1):70–83.PubMedPubMedCentralCrossRef
18.
go back to reference Sassoon SA, Fama R, Rosenbloom MJ, O’Reilly A, Pfefferbaum A, Sullivan EV. Component cognitive and motor processes of the digit symbol test: differential deficits in alcoholism, HIV infection, and their comorbidity. Alcohol Clin Exp Res. 2007;31(8):1315–24.PubMedCrossRef Sassoon SA, Fama R, Rosenbloom MJ, O’Reilly A, Pfefferbaum A, Sullivan EV. Component cognitive and motor processes of the digit symbol test: differential deficits in alcoholism, HIV infection, and their comorbidity. Alcohol Clin Exp Res. 2007;31(8):1315–24.PubMedCrossRef
19.
go back to reference Schulte T, Mueller-Oehring EM, Rosenbloom MJ, Pfefferbaum A, Sullivan EV. Differential effect of HIV infection and alcoholism on conflict processing, attentional allocation, and perceptual load: evidence from a Stroop Match-to-Sample task. Biol Psychiatry. 2005;57(1):67–75.PubMedCrossRef Schulte T, Mueller-Oehring EM, Rosenbloom MJ, Pfefferbaum A, Sullivan EV. Differential effect of HIV infection and alcoholism on conflict processing, attentional allocation, and perceptual load: evidence from a Stroop Match-to-Sample task. Biol Psychiatry. 2005;57(1):67–75.PubMedCrossRef
20.
go back to reference Rodriguez Salgado D, Rodriguez Alvarez M, Seoane PG. Neuropsychological impairment among asymptomatic HIV-positive former intravenous drug users. Cogn Behav Neurol. 2006;19(2):95–104.PubMedCrossRef Rodriguez Salgado D, Rodriguez Alvarez M, Seoane PG. Neuropsychological impairment among asymptomatic HIV-positive former intravenous drug users. Cogn Behav Neurol. 2006;19(2):95–104.PubMedCrossRef
21.
go back to reference Wakim KM, Freedman EG, Molloy CJ, Vieyto N, Cao Z, Foxe JJ. Assessing combinatorial effects of HIV infection and former cocaine dependence on cognitive control processes: a high-density electrical mapping study of response inhibition. Neuropharmacology. 2021;195:108636.PubMedPubMedCentralCrossRef Wakim KM, Freedman EG, Molloy CJ, Vieyto N, Cao Z, Foxe JJ. Assessing combinatorial effects of HIV infection and former cocaine dependence on cognitive control processes: a high-density electrical mapping study of response inhibition. Neuropharmacology. 2021;195:108636.PubMedPubMedCentralCrossRef
22.
go back to reference Wakim KM, Freedman EG, Tivarus ME, Heinecke A, Foxe JJ. Assessing combinatorial effects of HIV infection and former cocaine dependence on cognitive control processes: a functional neuroimaging study of response inhibition. Neuropharmacology. 2022;203:108815.PubMedCrossRef Wakim KM, Freedman EG, Tivarus ME, Heinecke A, Foxe JJ. Assessing combinatorial effects of HIV infection and former cocaine dependence on cognitive control processes: a functional neuroimaging study of response inhibition. Neuropharmacology. 2022;203:108815.PubMedCrossRef
23.
go back to reference Kumar P, Mahato DK, Kamle M, Borah R, Sharma B, Pandhi S, et al. Pharmacological properties, therapeutic potential, and legal status of Cannabis sativa L.: an overview. Phytother Res. 2021;35(11):6010–29.PubMedCrossRef Kumar P, Mahato DK, Kamle M, Borah R, Sharma B, Pandhi S, et al. Pharmacological properties, therapeutic potential, and legal status of Cannabis sativa L.: an overview. Phytother Res. 2021;35(11):6010–29.PubMedCrossRef
24.
go back to reference Shiau S, Arpadi SM, Yin MT, Martins SS. Patterns of drug use and HIV infection among adults in a nationally representative sample. Addict Behav. 2017;68:39–44.PubMedPubMedCentralCrossRef Shiau S, Arpadi SM, Yin MT, Martins SS. Patterns of drug use and HIV infection among adults in a nationally representative sample. Addict Behav. 2017;68:39–44.PubMedPubMedCentralCrossRef
25.
go back to reference Costiniuk CT, Saneei Z, Salahuddin S, Cox J, Routy JP, Rueda S, et al. Cannabis consumption in people living with HIV: reasons for use, secondary effects, and opportunities for health education. Cannabis Cannabinoid Res. 2019;4(3):204–13.PubMedPubMedCentralCrossRef Costiniuk CT, Saneei Z, Salahuddin S, Cox J, Routy JP, Rueda S, et al. Cannabis consumption in people living with HIV: reasons for use, secondary effects, and opportunities for health education. Cannabis Cannabinoid Res. 2019;4(3):204–13.PubMedPubMedCentralCrossRef
26.
go back to reference Harris GE, Dupuis L, Mugford GJ, Johnston L, Haase D, Page G, et al. Patterns and correlates of cannabis use among individuals with HIV/AIDS in Maritime Canada. Can J Infect Dis Med Microbiol. 2014;25(1):e1-7.PubMedPubMedCentralCrossRef Harris GE, Dupuis L, Mugford GJ, Johnston L, Haase D, Page G, et al. Patterns and correlates of cannabis use among individuals with HIV/AIDS in Maritime Canada. Can J Infect Dis Med Microbiol. 2014;25(1):e1-7.PubMedPubMedCentralCrossRef
27.
go back to reference Mechoulam R, Parker LA. The endocannabinoid system and the brain. Annu Rev Psychol. 2013;64:21–47.PubMedCrossRef Mechoulam R, Parker LA. The endocannabinoid system and the brain. Annu Rev Psychol. 2013;64:21–47.PubMedCrossRef
28.
go back to reference Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, Watanabe M. Endocannabinoid-mediated control of synaptic transmission. Physiol Rev. 2009;89(1):309–80.PubMedCrossRef Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, Watanabe M. Endocannabinoid-mediated control of synaptic transmission. Physiol Rev. 2009;89(1):309–80.PubMedCrossRef
29.
go back to reference Mackie K. Cannabinoid receptors: where they are and what they do. J Neuroendocrinol. 2008;20(Suppl 1):10–4.PubMedCrossRef Mackie K. Cannabinoid receptors: where they are and what they do. J Neuroendocrinol. 2008;20(Suppl 1):10–4.PubMedCrossRef
31.
go back to reference Zanettini C, Panlilio LV, Alicki M, Goldberg SR, Haller J, Yasar S. Effects of endocannabinoid system modulation on cognitive and emotional behavior. Front Behav Neurosci. 2011;5:57.PubMedPubMedCentralCrossRef Zanettini C, Panlilio LV, Alicki M, Goldberg SR, Haller J, Yasar S. Effects of endocannabinoid system modulation on cognitive and emotional behavior. Front Behav Neurosci. 2011;5:57.PubMedPubMedCentralCrossRef
32.
go back to reference Xu C, Hermes DJ, Nwanguma B, Jacobs IR, Mackie K, Mukhopadhyay S, et al. Endocannabinoids exert CB(1) receptor-mediated neuroprotective effects in models of neuronal damage induced by HIV-1 Tat protein. Mol Cell Neurosci. 2017;83:92–102.PubMedPubMedCentralCrossRef Xu C, Hermes DJ, Nwanguma B, Jacobs IR, Mackie K, Mukhopadhyay S, et al. Endocannabinoids exert CB(1) receptor-mediated neuroprotective effects in models of neuronal damage induced by HIV-1 Tat protein. Mol Cell Neurosci. 2017;83:92–102.PubMedPubMedCentralCrossRef
33.
go back to reference Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, et al. CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science. 2003;302(5642):84–8.PubMedCrossRef Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, et al. CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science. 2003;302(5642):84–8.PubMedCrossRef
34.
go back to reference Ehrhart J, Obregon D, Mori T, Hou H, Sun N, Bai Y, et al. Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation. J Neuroinflammation. 2005;2:29.PubMedPubMedCentralCrossRef Ehrhart J, Obregon D, Mori T, Hou H, Sun N, Bai Y, et al. Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation. J Neuroinflammation. 2005;2:29.PubMedPubMedCentralCrossRef
35.
go back to reference Murray CH, Javanbakht M, Cho GD, Gorbach PM, Fulcher JA, Cooper ZD. Changes in immune-related biomarkers and endocannabinoids as a function of frequency of cannabis use in people living with and without HIV. Cannabis and cannabinoid research. Advance online publication. 2023. https://doi.org/10.1089/can.2022.0287. Murray CH, Javanbakht M, Cho GD, Gorbach PM, Fulcher JA, Cooper ZD. Changes in immune-related biomarkers and endocannabinoids as a function of frequency of cannabis use in people living with and without HIV. Cannabis and cannabinoid research. Advance online publication. 2023. https://​doi.​org/​10.​1089/​can.​2022.​0287.
36.
go back to reference Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.PubMedCrossRef Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.PubMedCrossRef
37.
go back to reference Wise LE, Thorpe AJ, Lichtman AH. Hippocampal CB(1) receptors mediate the memory impairing effects of Delta(9)-tetrahydrocannabinol. Neuropsychopharmacology. 2009;34(9):2072–80.PubMedCrossRef Wise LE, Thorpe AJ, Lichtman AH. Hippocampal CB(1) receptors mediate the memory impairing effects of Delta(9)-tetrahydrocannabinol. Neuropsychopharmacology. 2009;34(9):2072–80.PubMedCrossRef
38.
go back to reference Nidadavolu P, Bilkei-Gorzo A, Kramer M, Schurmann B, Palmisano M, Beins EC, et al. Efficacy of Delta(9) -Tetrahydrocannabinol (THC) alone or in combination with a 1:1 ratio of cannabidiol (CBD) in reversing the spatial learning deficits in old mice. Front Aging Neurosci. 2021;13:718850.PubMedPubMedCentralCrossRef Nidadavolu P, Bilkei-Gorzo A, Kramer M, Schurmann B, Palmisano M, Beins EC, et al. Efficacy of Delta(9) -Tetrahydrocannabinol (THC) alone or in combination with a 1:1 ratio of cannabidiol (CBD) in reversing the spatial learning deficits in old mice. Front Aging Neurosci. 2021;13:718850.PubMedPubMedCentralCrossRef
39.
go back to reference Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–805.PubMedPubMedCentralCrossRef Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–805.PubMedPubMedCentralCrossRef
40.
go back to reference Martinez-Aguirre C, Carmona-Cruz F, Velasco AL, Velasco F, Aguado-Carrillo G, Cuellar-Herrera M, et al. Cannabidiol acts at 5-HT(1A) receptors in the human brain: relevance for treating temporal lobe epilepsy. Front Behav Neurosci. 2020;14:611278.PubMedPubMedCentralCrossRef Martinez-Aguirre C, Carmona-Cruz F, Velasco AL, Velasco F, Aguado-Carrillo G, Cuellar-Herrera M, et al. Cannabidiol acts at 5-HT(1A) receptors in the human brain: relevance for treating temporal lobe epilepsy. Front Behav Neurosci. 2020;14:611278.PubMedPubMedCentralCrossRef
41.
go back to reference Etemad L, Karimi G, Alavi MS, Roohbakhsh A. Pharmacological effects of cannabidiol by transient receptor potential channels. Life Sci. 2022;300:120582.PubMedCrossRef Etemad L, Karimi G, Alavi MS, Roohbakhsh A. Pharmacological effects of cannabidiol by transient receptor potential channels. Life Sci. 2022;300:120582.PubMedCrossRef
42.
43.
go back to reference Fusar-Poli P, Allen P, Bhattacharyya S, Crippa JA, Mechelli A, Borgwardt S, et al. Modulation of effective connectivity during emotional processing by Delta 9-tetrahydrocannabinol and cannabidiol. Int J Neuropsychopharmacol. 2010;13(4):421–32.PubMedCrossRef Fusar-Poli P, Allen P, Bhattacharyya S, Crippa JA, Mechelli A, Borgwardt S, et al. Modulation of effective connectivity during emotional processing by Delta 9-tetrahydrocannabinol and cannabidiol. Int J Neuropsychopharmacol. 2010;13(4):421–32.PubMedCrossRef
44.
go back to reference Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74.PubMedCrossRef Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74.PubMedCrossRef
45.
go back to reference Fusar-Poli P, Crippa JA, Bhattacharyya S, Borgwardt SJ, Allen P, Martin-Santos R, et al. Distinct effects of delta9-tetrahydrocannabinol and cannabidiol on neural activation during emotional processing. Arch Gen Psychiatry. 2009;66(1):95–105.PubMedCrossRef Fusar-Poli P, Crippa JA, Bhattacharyya S, Borgwardt SJ, Allen P, Martin-Santos R, et al. Distinct effects of delta9-tetrahydrocannabinol and cannabidiol on neural activation during emotional processing. Arch Gen Psychiatry. 2009;66(1):95–105.PubMedCrossRef
46.
go back to reference Curran T, Devillez H, YorkWilliams SL, Bidwell LC. Acute effects of naturalistic THC vs. CBD use on recognition memory: a preliminary study. J Cannabis Res. 2020;2(1):28.PubMedPubMedCentralCrossRef Curran T, Devillez H, YorkWilliams SL, Bidwell LC. Acute effects of naturalistic THC vs. CBD use on recognition memory: a preliminary study. J Cannabis Res. 2020;2(1):28.PubMedPubMedCentralCrossRef
47.
go back to reference Woelfl T, Rohleder C, Mueller JK, Lange B, Reuter A, Schmidt AM, et al. Effects of cannabidiol and delta-9-tetrahydrocannabinol on emotion, cognition, and attention: a double-blind, placebo-controlled, randomized experimental trial in healthy volunteers. Front Psychiatry. 2020;11:576877.PubMedPubMedCentralCrossRef Woelfl T, Rohleder C, Mueller JK, Lange B, Reuter A, Schmidt AM, et al. Effects of cannabidiol and delta-9-tetrahydrocannabinol on emotion, cognition, and attention: a double-blind, placebo-controlled, randomized experimental trial in healthy volunteers. Front Psychiatry. 2020;11:576877.PubMedPubMedCentralCrossRef
48.
go back to reference Toggas SM, Masliah E, Rockenstein EM, Rall GF, Abraham CR, Mucke L. Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice. Nature. 1994;367(6459):188–93.PubMedCrossRef Toggas SM, Masliah E, Rockenstein EM, Rall GF, Abraham CR, Mucke L. Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice. Nature. 1994;367(6459):188–93.PubMedCrossRef
49.
go back to reference Kim BO, Liu Y, Ruan Y, Xu ZC, Schantz L, He JJ. Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline. Am J Pathol. 2003;162(5):1693–707.PubMedPubMedCentralCrossRef Kim BO, Liu Y, Ruan Y, Xu ZC, Schantz L, He JJ. Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline. Am J Pathol. 2003;162(5):1693–707.PubMedPubMedCentralCrossRef
50.
go back to reference Reid W, Sadowska M, Denaro F, Rao S, Foulke J Jr, Hayes N, et al. An HIV-1 transgenic rat that develops HIV-related pathology and immunologic dysfunction. Proc Natl Acad Sci U S A. 2001;98(16):9271–6.PubMedPubMedCentralCrossRef Reid W, Sadowska M, Denaro F, Rao S, Foulke J Jr, Hayes N, et al. An HIV-1 transgenic rat that develops HIV-related pathology and immunologic dysfunction. Proc Natl Acad Sci U S A. 2001;98(16):9271–6.PubMedPubMedCentralCrossRef
52.
go back to reference Potash MJ, Chao W, Bentsman G, Paris N, Saini M, Nitkiewicz J, et al. A mouse model for study of systemic HIV-1 infection, antiviral immune responses, and neuroinvasiveness. Proc Natl Acad Sci U S A. 2005;102(10):3760–5.PubMedPubMedCentralCrossRef Potash MJ, Chao W, Bentsman G, Paris N, Saini M, Nitkiewicz J, et al. A mouse model for study of systemic HIV-1 infection, antiviral immune responses, and neuroinvasiveness. Proc Natl Acad Sci U S A. 2005;102(10):3760–5.PubMedPubMedCentralCrossRef
54.
go back to reference Mallard J, Williams K. An SIV macaque model of SIV and HAND: the need for adjunctive therapies in HIV that target activated monocytes and macrophages. J Neurovirol. 2018;24(2):213–9.PubMedCrossRef Mallard J, Williams K. An SIV macaque model of SIV and HAND: the need for adjunctive therapies in HIV that target activated monocytes and macrophages. J Neurovirol. 2018;24(2):213–9.PubMedCrossRef
55.
go back to reference Miller C, Abdo Z, Ericsson A, Elder J, VandeWoude S. Applications of the FIV model to study HIV pathogenesis. Viruses. 2018;10(4). Miller C, Abdo Z, Ericsson A, Elder J, VandeWoude S. Applications of the FIV model to study HIV pathogenesis. Viruses. 2018;10(4).
56.
go back to reference Del Prete GQ, Eilers B, Moldt B, Keele BF, Estes JD, Rodriguez A, Sampias M, Oswald K, Fast R, Trubey CM, Chertova E, Smedley J, LaBranche CC, Montefiori DC, Burton DR, Shaw GM, Markowitz M, Piatak M, Jr KewalRamani V N, Bieniasz PD, Hatziioannou T. Selection of unadapted, pathogenic SHIVs encoding newly transmitted HIV-1 envelope proteins. Cell host & microbe, 2014;16(3):412–418. https://doi.org/10.1016/j.chom.2014.08.003. Del Prete GQ, Eilers B, Moldt B, Keele BF, Estes JD, Rodriguez A, Sampias M, Oswald K, Fast R, Trubey CM, Chertova E, Smedley J, LaBranche CC, Montefiori DC, Burton DR, Shaw GM, Markowitz M, Piatak M, Jr KewalRamani V N, Bieniasz PD, Hatziioannou T. Selection of unadapted, pathogenic SHIVs encoding newly transmitted HIV-1 envelope proteins. Cell host & microbe, 2014;16(3):412–418. https://​doi.​org/​10.​1016/​j.​chom.​2014.​08.​003.
57.
go back to reference Heaton RK, Ellis RJ, Tang B, Marra CM, Rubin LH, Clifford DB, et al. Twelve-year neurocognitive decline in HIV is associated with comorbidities, not age: a CHARTER study. Brain. 2023;146(3):1121–31.PubMedCrossRef Heaton RK, Ellis RJ, Tang B, Marra CM, Rubin LH, Clifford DB, et al. Twelve-year neurocognitive decline in HIV is associated with comorbidities, not age: a CHARTER study. Brain. 2023;146(3):1121–31.PubMedCrossRef
58.
go back to reference Watson CW, Paolillo EW, Morgan EE, Umlauf A, Sundermann EE, Ellis RJ, et al. Cannabis exposure is associated with a lower likelihood of neurocognitive impairment in people living with HIV. J Acquir Immune Defic Syndr. 2020;83(1):56–64.PubMedPubMedCentralCrossRef Watson CW, Paolillo EW, Morgan EE, Umlauf A, Sundermann EE, Ellis RJ, et al. Cannabis exposure is associated with a lower likelihood of neurocognitive impairment in people living with HIV. J Acquir Immune Defic Syndr. 2020;83(1):56–64.PubMedPubMedCentralCrossRef
59.
go back to reference Naveed Z, Fox HS, Wichman CS, May P, Arcari CM, Meza J, et al. An assessment of factors associated with neurocognitive decline in people living with HIV. Int J STD AIDS. 2022;33(1):38–47.PubMedCrossRef Naveed Z, Fox HS, Wichman CS, May P, Arcari CM, Meza J, et al. An assessment of factors associated with neurocognitive decline in people living with HIV. Int J STD AIDS. 2022;33(1):38–47.PubMedCrossRef
60.
go back to reference Cristiani SA, Pukay-Martin ND, Bornstein RA. Marijuana use and cognitive function in HIV-infected people. J Neuropsychiatry Clin Neurosci. 2004;16(3):330–5.PubMedCrossRef Cristiani SA, Pukay-Martin ND, Bornstein RA. Marijuana use and cognitive function in HIV-infected people. J Neuropsychiatry Clin Neurosci. 2004;16(3):330–5.PubMedCrossRef
61.
go back to reference Schouten J, Su T, Wit FW, Kootstra NA, Caan MW, Geurtsen GJ, et al. Determinants of reduced cognitive performance in HIV-1-infected middle-aged men on combination antiretroviral therapy. AIDS. 2016;30(7):1027–38.PubMedCrossRef Schouten J, Su T, Wit FW, Kootstra NA, Caan MW, Geurtsen GJ, et al. Determinants of reduced cognitive performance in HIV-1-infected middle-aged men on combination antiretroviral therapy. AIDS. 2016;30(7):1027–38.PubMedCrossRef
62.
go back to reference Thames AD, Mahmood Z, Burggren AC, Karimian A, Kuhn TP. Combined effects of HIV and marijuana use on neurocognitive functioning and immune status. AIDS Care. 2016;28(5):628–32.PubMedCrossRef Thames AD, Mahmood Z, Burggren AC, Karimian A, Kuhn TP. Combined effects of HIV and marijuana use on neurocognitive functioning and immune status. AIDS Care. 2016;28(5):628–32.PubMedCrossRef
63.
go back to reference Saloner R, Campbell LM, Serrano V, Montoya JL, Pasipanodya E, Paolillo EW, et al. Neurocognitive superaging in older adults living with HIV: demographic, neuromedical and everyday functioning correlates. J Int Neuropsychol Soc. 2019;25(5):507–19.PubMedPubMedCentralCrossRef Saloner R, Campbell LM, Serrano V, Montoya JL, Pasipanodya E, Paolillo EW, et al. Neurocognitive superaging in older adults living with HIV: demographic, neuromedical and everyday functioning correlates. J Int Neuropsychol Soc. 2019;25(5):507–19.PubMedPubMedCentralCrossRef
64.
go back to reference Watson CW, Campbell LM, Sun-Suslow N, Hong S, Umlauf A, Ellis RJ, et al. Daily cannabis use is associated with lower CNS inflammation in people with HIV. J Int Neuropsychol Soc. 2021;27(6):661–72.PubMedPubMedCentralCrossRef Watson CW, Campbell LM, Sun-Suslow N, Hong S, Umlauf A, Ellis RJ, et al. Daily cannabis use is associated with lower CNS inflammation in people with HIV. J Int Neuropsychol Soc. 2021;27(6):661–72.PubMedPubMedCentralCrossRef
65.
go back to reference Watson CW, Sundermann E, Helm J, Paolillo EW, Hong S, Ellis RJ, Letendre S, Marcotte TD, Heaton RK, Morgan EE, Grant I. A longitudinal study of cannabis use and risk for cognitive and functional decline among older adults with HIV. AIDS and behavior. 2023;27(10):3401–3413. https://doi.org/10.1007/s10461-023-04056-6. Watson CW, Sundermann E, Helm J, Paolillo EW, Hong S, Ellis RJ, Letendre S, Marcotte TD, Heaton RK, Morgan EE, Grant I. A longitudinal study of cannabis use and risk for cognitive and functional decline among older adults with HIV. AIDS and behavior. 2023;27(10):3401–3413. https://​doi.​org/​10.​1007/​s10461-023-04056-6.
66.
go back to reference Thames AD, Kuhn TP, Williamson TJ, Jones JD, Mahmood Z, Hammond A. Marijuana effects on changes in brain structure and cognitive function among HIV+ and HIV- adults. Drug Alcohol Depend. 2017;170:120–7.PubMedCrossRef Thames AD, Kuhn TP, Williamson TJ, Jones JD, Mahmood Z, Hammond A. Marijuana effects on changes in brain structure and cognitive function among HIV+ and HIV- adults. Drug Alcohol Depend. 2017;170:120–7.PubMedCrossRef
67.
go back to reference Murdoch DM, Barfield R, Chan C, Towe SL, Bell RP, Volkheimer A, et al. Neuroimaging and immunological features of neurocognitive function related to substance use in people with HIV. J Neurovirol. 2023;29(1):78–93.PubMedCrossRef Murdoch DM, Barfield R, Chan C, Towe SL, Bell RP, Volkheimer A, et al. Neuroimaging and immunological features of neurocognitive function related to substance use in people with HIV. J Neurovirol. 2023;29(1):78–93.PubMedCrossRef
68.
go back to reference Byrd DA, Fellows RP, Morgello S, Franklin D, Heaton RK, Deutsch R, et al. Neurocognitive impact of substance use in HIV infection. J Acquir Immune Defic Syndr. 2011;58(2):154–62.PubMedPubMedCentralCrossRef Byrd DA, Fellows RP, Morgello S, Franklin D, Heaton RK, Deutsch R, et al. Neurocognitive impact of substance use in HIV infection. J Acquir Immune Defic Syndr. 2011;58(2):154–62.PubMedPubMedCentralCrossRef
69.
go back to reference Rogers JM, Iudicello JE, Marcondes MCG, Morgan EE, Cherner M, Ellis RJ, Letendre SL, Heaton RK, Grant I. The combined effects of cannabis, methamphetamine, and HIV on neurocognition. Viruses. 2023;15(3):674. https://doi.org/10.3390/v15030674. Rogers JM, Iudicello JE, Marcondes MCG, Morgan EE, Cherner M, Ellis RJ, Letendre SL, Heaton RK, Grant I. The combined effects of cannabis, methamphetamine, and HIV on neurocognition. Viruses. 2023;15(3):674. https://​doi.​org/​10.​3390/​v15030674.
70.
go back to reference Wang HA, Liang HJ, Ernst TM, Oishi K, Chang L. Microstructural brain abnormalities in HIV+ individuals with or without chronic marijuana use. J Neuroinflammation. 2020;17(1):230.PubMedPubMedCentralCrossRef Wang HA, Liang HJ, Ernst TM, Oishi K, Chang L. Microstructural brain abnormalities in HIV+ individuals with or without chronic marijuana use. J Neuroinflammation. 2020;17(1):230.PubMedPubMedCentralCrossRef
71.
go back to reference Kallianpur KJ, Birn R, Ndhlovu LC, Souza SA, Mitchell B, Paul R, et al. Impact of cannabis use on brain structure and function in suppressed HIV infection. J Behav Brain Sci. 2020;10(8):344–70.PubMedPubMedCentralCrossRef Kallianpur KJ, Birn R, Ndhlovu LC, Souza SA, Mitchell B, Paul R, et al. Impact of cannabis use on brain structure and function in suppressed HIV infection. J Behav Brain Sci. 2020;10(8):344–70.PubMedPubMedCentralCrossRef
72.
go back to reference Schantell M, Springer SD, Arif Y, Sandal ME, Willett MP, Johnson HJ, et al. Regular cannabis use modulates the impact of HIV on the neural dynamics serving cognitive control. J Psychopharmacol. 2022;36(12):1324–37.PubMedPubMedCentralCrossRef Schantell M, Springer SD, Arif Y, Sandal ME, Willett MP, Johnson HJ, et al. Regular cannabis use modulates the impact of HIV on the neural dynamics serving cognitive control. J Psychopharmacol. 2022;36(12):1324–37.PubMedPubMedCentralCrossRef
73.
go back to reference Flannery JS, Riedel MC, Hill-Bowen LD, Poudel R, Bottenhorn KL, Salo T, et al. Altered large-scale brain network interactions associated with HIV infection and error processing. Netw Neurosci. 2022;6(3):791–815.PubMedPubMedCentralCrossRef Flannery JS, Riedel MC, Hill-Bowen LD, Poudel R, Bottenhorn KL, Salo T, et al. Altered large-scale brain network interactions associated with HIV infection and error processing. Netw Neurosci. 2022;6(3):791–815.PubMedPubMedCentralCrossRef
74.
go back to reference Gomez D, Power C, Gill MJ, Fujiwara E. Determinants of risk-taking in HIV-associated neurocognitive disorders. Neuropsychology. 2017;31(7):798–810.PubMedCrossRef Gomez D, Power C, Gill MJ, Fujiwara E. Determinants of risk-taking in HIV-associated neurocognitive disorders. Neuropsychology. 2017;31(7):798–810.PubMedCrossRef
75.
go back to reference Attonito JM, Devieux JG, Lerner BD, Hospital MM, Rosenberg R. Exploring substance use and HIV treatment factors associated with neurocognitive impairment among people living with HIV/AIDS. Front Public Health. 2014;2:105.PubMedPubMedCentralCrossRef Attonito JM, Devieux JG, Lerner BD, Hospital MM, Rosenberg R. Exploring substance use and HIV treatment factors associated with neurocognitive impairment among people living with HIV/AIDS. Front Public Health. 2014;2:105.PubMedPubMedCentralCrossRef
76.
go back to reference Christopher-Hayes NJ, Lew BJ, Wiesman AI, Schantell M, O’Neill J, May PE, et al. Cannabis use impacts pre-stimulus neural activity in the visual cortices of people with HIV. Hum Brain Mapp. 2021;42(16):5446–57.PubMedPubMedCentralCrossRef Christopher-Hayes NJ, Lew BJ, Wiesman AI, Schantell M, O’Neill J, May PE, et al. Cannabis use impacts pre-stimulus neural activity in the visual cortices of people with HIV. Hum Brain Mapp. 2021;42(16):5446–57.PubMedPubMedCentralCrossRef
77.
go back to reference Crook CL, Savin MJ, Byrd D, Summers AC, Guzman VA, Morris EP, et al. The neurocognitive effects of a past cannabis use disorder in a diverse sample of people living with HIV. AIDS Care. 2021;33(11):1482–91.PubMedCrossRef Crook CL, Savin MJ, Byrd D, Summers AC, Guzman VA, Morris EP, et al. The neurocognitive effects of a past cannabis use disorder in a diverse sample of people living with HIV. AIDS Care. 2021;33(11):1482–91.PubMedCrossRef
78.
go back to reference Okafor CN, Plankey MW, Li M, Chen X, Surkan PJ, Shoptaw S, et al. Association of marijuana use with changes in cognitive processing speed and flexibility for 17 years in HIV-seropositive and HIV-seronegative men. Subst Use Misuse. 2019;54(4):525–37.PubMedPubMedCentralCrossRef Okafor CN, Plankey MW, Li M, Chen X, Surkan PJ, Shoptaw S, et al. Association of marijuana use with changes in cognitive processing speed and flexibility for 17 years in HIV-seropositive and HIV-seronegative men. Subst Use Misuse. 2019;54(4):525–37.PubMedPubMedCentralCrossRef
79.
go back to reference Flannery JS, Riedel MC, Salo T, Hill-Bowen LD, Poudel R, Adams AR, et al. Interactive effects of HIV infection and cannabis use on insula subregion functional connectivity. J Neuroimmune Pharmacol. 2022;17(1–2):289–304.PubMedCrossRef Flannery JS, Riedel MC, Salo T, Hill-Bowen LD, Poudel R, Adams AR, et al. Interactive effects of HIV infection and cannabis use on insula subregion functional connectivity. J Neuroimmune Pharmacol. 2022;17(1–2):289–304.PubMedCrossRef
80.
go back to reference Flannery JS, Riedel MC, Salo T, Poudel R, Laird AR, Gonzalez R, et al. HIV infection is linked with reduced error-related default mode network suppression and poorer medication management abilities. Prog Neuropsychopharmacol Biol Psychiatry. 2021;111:110398.PubMedPubMedCentralCrossRef Flannery JS, Riedel MC, Salo T, Poudel R, Laird AR, Gonzalez R, et al. HIV infection is linked with reduced error-related default mode network suppression and poorer medication management abilities. Prog Neuropsychopharmacol Biol Psychiatry. 2021;111:110398.PubMedPubMedCentralCrossRef
81.
go back to reference Chang L, Cloak C, Yakupov R, Ernst T. Combined and independent effects of chronic marijuana use and HIV on brain metabolites. J Neuroimmune Pharmacol. 2006;1(1):65–76.PubMedPubMedCentralCrossRef Chang L, Cloak C, Yakupov R, Ernst T. Combined and independent effects of chronic marijuana use and HIV on brain metabolites. J Neuroimmune Pharmacol. 2006;1(1):65–76.PubMedPubMedCentralCrossRef
82.
go back to reference Meade CS, Bell RP, Towe SL, Chen NK, Hobkirk AL, Huettel SA. Synergistic effects of marijuana abuse and HIV infection on neural activation during a cognitive interference task. Addict Biol. 2019;24(6):1235–44.PubMedCrossRef Meade CS, Bell RP, Towe SL, Chen NK, Hobkirk AL, Huettel SA. Synergistic effects of marijuana abuse and HIV infection on neural activation during a cognitive interference task. Addict Biol. 2019;24(6):1235–44.PubMedCrossRef
83.
go back to reference Skalski LM, Towe SL, Sikkema KJ, Meade CS. Memory impairment in HIV-infected individuals with early and late initiation of regular marijuana use. AIDS Behav. 2018;22(5):1596–605.PubMedPubMedCentralCrossRef Skalski LM, Towe SL, Sikkema KJ, Meade CS. Memory impairment in HIV-infected individuals with early and late initiation of regular marijuana use. AIDS Behav. 2018;22(5):1596–605.PubMedPubMedCentralCrossRef
84.
go back to reference Iudicello JE, Woods SP, Cattie JE, Doyle K, Grant I, Group HIVNRP. Risky decision-making in HIV-associated neurocognitive disorders (HAND). Clin Neuropsychol. 2013;27(2):256–75.PubMedCrossRef Iudicello JE, Woods SP, Cattie JE, Doyle K, Grant I, Group HIVNRP. Risky decision-making in HIV-associated neurocognitive disorders (HAND). Clin Neuropsychol. 2013;27(2):256–75.PubMedCrossRef
85.
go back to reference Fridberg DJ, Queller S, Ahn WY, Kim W, Bishara AJ, Busemeyer JR, et al. Cognitive mechanisms underlying risky decision-making in chronic cannabis users. J Math Psychol. 2010;54(1):28–38.PubMedPubMedCentralCrossRef Fridberg DJ, Queller S, Ahn WY, Kim W, Bishara AJ, Busemeyer JR, et al. Cognitive mechanisms underlying risky decision-making in chronic cannabis users. J Math Psychol. 2010;54(1):28–38.PubMedPubMedCentralCrossRef
86.
go back to reference Jacobs IR, Xu C, Hermes DJ, League AF, Xu C, Nath B, et al. Inhibitory control deficits associated with upregulation of CB(1)R in the HIV-1 Tat transgenic mouse model of HAND. J Neuroimmune Pharmacol. 2019;14(4):661–78.PubMedPubMedCentralCrossRef Jacobs IR, Xu C, Hermes DJ, League AF, Xu C, Nath B, et al. Inhibitory control deficits associated with upregulation of CB(1)R in the HIV-1 Tat transgenic mouse model of HAND. J Neuroimmune Pharmacol. 2019;14(4):661–78.PubMedPubMedCentralCrossRef
87.
go back to reference League AF, Gorman BL, Hermes DJ, Johnson CT, Jacobs IR, Yadav-Samudrala BJ, et al. Monoacylglycerol lipase inhibitor MJN110 reduces neuronal hyperexcitability, restores dendritic arborization complexity, and regulates reward-related behavior in presence of HIV-1 Tat. Front Neurol. 2021;12:651272.PubMedPubMedCentralCrossRef League AF, Gorman BL, Hermes DJ, Johnson CT, Jacobs IR, Yadav-Samudrala BJ, et al. Monoacylglycerol lipase inhibitor MJN110 reduces neuronal hyperexcitability, restores dendritic arborization complexity, and regulates reward-related behavior in presence of HIV-1 Tat. Front Neurol. 2021;12:651272.PubMedPubMedCentralCrossRef
88.
go back to reference Kesby JP, Fields JA, Chang A, Coban H, Achim CL, Semenova S, et al. Effects of HIV-1 TAT protein and methamphetamine exposure on visual discrimination and executive function in mice. Behav Brain Res. 2018;349:73–9.PubMedPubMedCentralCrossRef Kesby JP, Fields JA, Chang A, Coban H, Achim CL, Semenova S, et al. Effects of HIV-1 TAT protein and methamphetamine exposure on visual discrimination and executive function in mice. Behav Brain Res. 2018;349:73–9.PubMedPubMedCentralCrossRef
89.
go back to reference Roberts BZ, He YV, Chatha M, Minassian A, Grant I, Young JW. HIV Transgenic rats demonstrate superior task acquisition and intact reversal learning in the within-session probabilistic reversal learning task. Cogn Affect Behav Neurosci. 2021;21(6):1207–21.PubMedPubMedCentralCrossRef Roberts BZ, He YV, Chatha M, Minassian A, Grant I, Young JW. HIV Transgenic rats demonstrate superior task acquisition and intact reversal learning in the within-session probabilistic reversal learning task. Cogn Affect Behav Neurosci. 2021;21(6):1207–21.PubMedPubMedCentralCrossRef
90.
go back to reference Haney M, Rabkin J, Gunderson E, Foltin RW. Dronabinol and marijuana in HIV(+) marijuana smokers: acute effects on caloric intake and mood. Psychopharmacology. 2005;181(1):170–8.PubMedCrossRef Haney M, Rabkin J, Gunderson E, Foltin RW. Dronabinol and marijuana in HIV(+) marijuana smokers: acute effects on caloric intake and mood. Psychopharmacology. 2005;181(1):170–8.PubMedCrossRef
91.
go back to reference Bedi G, Foltin RW, Gunderson EW, Rabkin J, Hart CL, Comer SD, et al. Efficacy and tolerability of high-dose dronabinol maintenance in HIV-positive marijuana smokers: a controlled laboratory study. Psychopharmacology. 2010;212(4):675–86.PubMedPubMedCentralCrossRef Bedi G, Foltin RW, Gunderson EW, Rabkin J, Hart CL, Comer SD, et al. Efficacy and tolerability of high-dose dronabinol maintenance in HIV-positive marijuana smokers: a controlled laboratory study. Psychopharmacology. 2010;212(4):675–86.PubMedPubMedCentralCrossRef
92.
go back to reference Haney M, Gunderson EW, Rabkin J, Hart CL, Vosburg SK, Comer SD, et al. Dronabinol and marijuana in HIV-positive marijuana smokers. Caloric intake, mood, and sleep. J Acquir Immune Defic Syndr. 2007;45(5):545–54.PubMedCrossRef Haney M, Gunderson EW, Rabkin J, Hart CL, Vosburg SK, Comer SD, et al. Dronabinol and marijuana in HIV-positive marijuana smokers. Caloric intake, mood, and sleep. J Acquir Immune Defic Syndr. 2007;45(5):545–54.PubMedCrossRef
93.
go back to reference Lorkiewicz SA, Ventura AS, Heeren TC, Winter MR, Walley AY, Sullivan M, et al. Lifetime marijuana and alcohol use, and cognitive dysfunction in people with human immunodeficiency virus infection. Subst Abus. 2018;39(1):116–23.PubMedCrossRef Lorkiewicz SA, Ventura AS, Heeren TC, Winter MR, Walley AY, Sullivan M, et al. Lifetime marijuana and alcohol use, and cognitive dysfunction in people with human immunodeficiency virus infection. Subst Abus. 2018;39(1):116–23.PubMedCrossRef
94.
go back to reference Cash MC, Cunnane K, Fan C, Romero-Sandoval EA. Mapping cannabis potency in medical and recreational programs in the United States. PLoS ONE. 2020;15(3):e0230167.PubMedPubMedCentralCrossRef Cash MC, Cunnane K, Fan C, Romero-Sandoval EA. Mapping cannabis potency in medical and recreational programs in the United States. PLoS ONE. 2020;15(3):e0230167.PubMedPubMedCentralCrossRef
95.
go back to reference Pennypacker SD, Cunnane K, Cash MC, Romero-Sandoval EA. Potency and therapeutic THC and CBD ratios: U.S. cannabis markets overshoot. Front Pharmacol. 2022;13:921493.PubMedPubMedCentralCrossRef Pennypacker SD, Cunnane K, Cash MC, Romero-Sandoval EA. Potency and therapeutic THC and CBD ratios: U.S. cannabis markets overshoot. Front Pharmacol. 2022;13:921493.PubMedPubMedCentralCrossRef
97.
go back to reference Crane NA, Schuster RM, Fusar-Poli P, Gonzalez R. Effects of cannabis on neurocognitive functioning: recent advances, neurodevelopmental influences, and sex differences. Neuropsychol Rev. 2013;23(2):117–37.PubMedCrossRef Crane NA, Schuster RM, Fusar-Poli P, Gonzalez R. Effects of cannabis on neurocognitive functioning: recent advances, neurodevelopmental influences, and sex differences. Neuropsychol Rev. 2013;23(2):117–37.PubMedCrossRef
98.
go back to reference Woods SP, Doyle KL, Morgan EE, Naar-King S, Outlaw AY, Nichols SL, et al. Task importance affects event-based prospective memory performance in adults with HIV-associated neurocognitive disorders and HIV-infected young adults with problematic substance use. J Int Neuropsychol Soc. 2014;20(6):652–62.PubMedPubMedCentralCrossRef Woods SP, Doyle KL, Morgan EE, Naar-King S, Outlaw AY, Nichols SL, et al. Task importance affects event-based prospective memory performance in adults with HIV-associated neurocognitive disorders and HIV-infected young adults with problematic substance use. J Int Neuropsychol Soc. 2014;20(6):652–62.PubMedPubMedCentralCrossRef
99.
go back to reference Winsauer PJ, Molina PE, Amedee AM, Filipeanu CM, McGoey RR, Troxclair DA, et al. Tolerance to chronic delta-9-tetrahydrocannabinol (Delta(9)-THC) in rhesus macaques infected with simian immunodeficiency virus. Exp Clin Psychopharmacol. 2011;19(2):154–72.PubMedPubMedCentralCrossRef Winsauer PJ, Molina PE, Amedee AM, Filipeanu CM, McGoey RR, Troxclair DA, et al. Tolerance to chronic delta-9-tetrahydrocannabinol (Delta(9)-THC) in rhesus macaques infected with simian immunodeficiency virus. Exp Clin Psychopharmacol. 2011;19(2):154–72.PubMedPubMedCentralCrossRef
100.
go back to reference Evans EB, Wenger GR. Effects of drugs of abuse on acquisition of behavioral chains in squirrel monkeys. Psychopharmacology. 1992;107(1):55–60.PubMedCrossRef Evans EB, Wenger GR. Effects of drugs of abuse on acquisition of behavioral chains in squirrel monkeys. Psychopharmacology. 1992;107(1):55–60.PubMedCrossRef
101.
go back to reference Schulze GE, McMillan DE, Bailey JR, Scallet A, Ali SF, Slikker W Jr, et al. Acute effects of delta-9-tetrahydrocannabinol in rhesus monkeys as measured by performance in a battery of complex operant tests. J Pharmacol Exp Ther. 1988;245(1):178–86.PubMed Schulze GE, McMillan DE, Bailey JR, Scallet A, Ali SF, Slikker W Jr, et al. Acute effects of delta-9-tetrahydrocannabinol in rhesus monkeys as measured by performance in a battery of complex operant tests. J Pharmacol Exp Ther. 1988;245(1):178–86.PubMed
102.
go back to reference Winsauer PJ, Lambert P, Moerschbaecher JM. Cannabinoid ligands and their effects on learning and performance in rhesus monkeys. Behav Pharmacol. 1999;10(5):497–511.PubMedCrossRef Winsauer PJ, Lambert P, Moerschbaecher JM. Cannabinoid ligands and their effects on learning and performance in rhesus monkeys. Behav Pharmacol. 1999;10(5):497–511.PubMedCrossRef
103.
go back to reference Beardsley PM, Scimeca JA, Martin BR. Studies on the agonistic activity of delta 9–11-tetrahydrocannabinol in mice, dogs and rhesus monkeys and its interactions with delta 9-tetrahydrocannabinol. J Pharmacol Exp Ther. 1987;241(2):521–6.PubMed Beardsley PM, Scimeca JA, Martin BR. Studies on the agonistic activity of delta 9–11-tetrahydrocannabinol in mice, dogs and rhesus monkeys and its interactions with delta 9-tetrahydrocannabinol. J Pharmacol Exp Ther. 1987;241(2):521–6.PubMed
104.
go back to reference Kamien JB, Bickel WK, Higgins ST, Hughes JR. The effects of delta(9)-tetrahydrocannabinol on repeated acquisition and performance of response sequences and on self-reports in humans. Behav Pharmacol. 1994;5(1):71–8.PubMedCrossRef Kamien JB, Bickel WK, Higgins ST, Hughes JR. The effects of delta(9)-tetrahydrocannabinol on repeated acquisition and performance of response sequences and on self-reports in humans. Behav Pharmacol. 1994;5(1):71–8.PubMedCrossRef
105.
go back to reference Wang L, Zeng Y, Zhou Y, Yu J, Liang M, Qin L, et al. Win 55,212–2 improves neural injury induced by HIV-1 glycoprotein 120 in rats by exciting CB2R. Brain Res Bull. 2022;182:67–79.PubMedCrossRef Wang L, Zeng Y, Zhou Y, Yu J, Liang M, Qin L, et al. Win 55,212–2 improves neural injury induced by HIV-1 glycoprotein 120 in rats by exciting CB2R. Brain Res Bull. 2022;182:67–79.PubMedCrossRef
106.
go back to reference Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11(1):47–60.PubMedCrossRef Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11(1):47–60.PubMedCrossRef
107.
go back to reference Gonzalez R, Schuster RM, Vassileva J, Martin EM. Impact of HIV and a history of marijuana dependence on procedural learning among individuals with a history of substance dependence. J Clin Exp Neuropsychol. 2011;33(7):735–52.PubMedPubMedCentralCrossRef Gonzalez R, Schuster RM, Vassileva J, Martin EM. Impact of HIV and a history of marijuana dependence on procedural learning among individuals with a history of substance dependence. J Clin Exp Neuropsychol. 2011;33(7):735–52.PubMedPubMedCentralCrossRef
108.
go back to reference ElSohly MA, Chandra S, Radwan M, Majumdar CG, Church JC. A comprehensive review of cannabis potency in the United States in the last decade. Biol Psychiatry Cogn Neurosci Neuroimaging. 2021;6(6):603–6.PubMed ElSohly MA, Chandra S, Radwan M, Majumdar CG, Church JC. A comprehensive review of cannabis potency in the United States in the last decade. Biol Psychiatry Cogn Neurosci Neuroimaging. 2021;6(6):603–6.PubMed
109.
go back to reference Ellis RJ, Badiee J, Vaida F, Letendre S, Heaton RK, Clifford D, et al. CD4 nadir is a predictor of HIV neurocognitive impairment in the era of combination antiretroviral therapy. AIDS. 2011;25(14):1747–51.PubMedCrossRef Ellis RJ, Badiee J, Vaida F, Letendre S, Heaton RK, Clifford D, et al. CD4 nadir is a predictor of HIV neurocognitive impairment in the era of combination antiretroviral therapy. AIDS. 2011;25(14):1747–51.PubMedCrossRef
110.
go back to reference Garvey L, Surendrakumar V, Winston A. Low rates of neurocognitive impairment are observed in neuro-asymptomatic HIV-infected subjects on effective antiretroviral therapy. HIV Clin Trials. 2011;12(6):333–8.PubMedCrossRef Garvey L, Surendrakumar V, Winston A. Low rates of neurocognitive impairment are observed in neuro-asymptomatic HIV-infected subjects on effective antiretroviral therapy. HIV Clin Trials. 2011;12(6):333–8.PubMedCrossRef
111.
go back to reference Hassanzadeh-Behbahani S, Shattuck KF, Bronshteyn M, Dawson M, Diaz M, Kumar P, et al. Low CD4 nadir linked to widespread cortical thinning in adults living with HIV. Neuroimage Clin. 2020;25:102155.PubMedCrossRef Hassanzadeh-Behbahani S, Shattuck KF, Bronshteyn M, Dawson M, Diaz M, Kumar P, et al. Low CD4 nadir linked to widespread cortical thinning in adults living with HIV. Neuroimage Clin. 2020;25:102155.PubMedCrossRef
112.
go back to reference Valcour V, Yee P, Williams AE, Shiramizu B, Watters M, Selnes O, et al. Lowest ever CD4 lymphocyte count (CD4 nadir) as a predictor of current cognitive and neurological status in human immunodeficiency virus type 1 infection–The Hawaii Aging with HIV Cohort. J Neurovirol. 2006;12(5):387–91.PubMedCrossRef Valcour V, Yee P, Williams AE, Shiramizu B, Watters M, Selnes O, et al. Lowest ever CD4 lymphocyte count (CD4 nadir) as a predictor of current cognitive and neurological status in human immunodeficiency virus type 1 infection–The Hawaii Aging with HIV Cohort. J Neurovirol. 2006;12(5):387–91.PubMedCrossRef
113.
go back to reference Munoz-Moreno JA, Fumaz CR, Ferrer MJ, Prats A, Negredo E, Garolera M, et al. Nadir CD4 cell count predicts neurocognitive impairment in HIV-infected patients. AIDS Res Hum Retrovir. 2008;24(10):1301–7.PubMedCrossRef Munoz-Moreno JA, Fumaz CR, Ferrer MJ, Prats A, Negredo E, Garolera M, et al. Nadir CD4 cell count predicts neurocognitive impairment in HIV-infected patients. AIDS Res Hum Retrovir. 2008;24(10):1301–7.PubMedCrossRef
114.
go back to reference Young JW. Development of cross-species translational paradigms for psychiatric research in the Research Domain Criteria era. Neurosci Biobehav Rev. 2023;148:105119.PubMedCrossRef Young JW. Development of cross-species translational paradigms for psychiatric research in the Research Domain Criteria era. Neurosci Biobehav Rev. 2023;148:105119.PubMedCrossRef
116.
go back to reference Aguilar-Valles A, Sanchez E, de Gortari P, Balderas I, Ramirez-Amaya V, Bermudez-Rattoni F, et al. Analysis of the stress response in rats trained in the water-maze: differential expression of corticotropin-releasing hormone, CRH-R1, glucocorticoid receptors and brain-derived neurotrophic factor in limbic regions. Neuroendocrinology. 2005;82(5–6):306–19.PubMedCrossRef Aguilar-Valles A, Sanchez E, de Gortari P, Balderas I, Ramirez-Amaya V, Bermudez-Rattoni F, et al. Analysis of the stress response in rats trained in the water-maze: differential expression of corticotropin-releasing hormone, CRH-R1, glucocorticoid receptors and brain-derived neurotrophic factor in limbic regions. Neuroendocrinology. 2005;82(5–6):306–19.PubMedCrossRef
117.
go back to reference Harrison FE, Hosseini AH, McDonald MP. Endogenous anxiety and stress responses in water maze and Barnes maze spatial memory tasks. Behav Brain Res. 2009;198(1):247–51.PubMedCrossRef Harrison FE, Hosseini AH, McDonald MP. Endogenous anxiety and stress responses in water maze and Barnes maze spatial memory tasks. Behav Brain Res. 2009;198(1):247–51.PubMedCrossRef
118.
go back to reference Raber J, Toggas SM, Lee S, Bloom FE, Epstein CJ, Mucke L. Central nervous system expression of HIV-1 Gp120 activates the hypothalamic-pituitary-adrenal axis: evidence for involvement of NMDA receptors and nitric oxide synthase. Virology. 1996;226(2):362–73.PubMedCrossRef Raber J, Toggas SM, Lee S, Bloom FE, Epstein CJ, Mucke L. Central nervous system expression of HIV-1 Gp120 activates the hypothalamic-pituitary-adrenal axis: evidence for involvement of NMDA receptors and nitric oxide synthase. Virology. 1996;226(2):362–73.PubMedCrossRef
119.
go back to reference Viveros MP, Marco EM, File SE. Endocannabinoid system and stress and anxiety responses. Pharmacol Biochem Behav. 2005;81(2):331–42.PubMedCrossRef Viveros MP, Marco EM, File SE. Endocannabinoid system and stress and anxiety responses. Pharmacol Biochem Behav. 2005;81(2):331–42.PubMedCrossRef
120.
go back to reference Young JW, Light GA, Marston HM, Sharp R, Geyer MA. The 5-choice continuous performance test: evidence for a translational test of vigilance for mice. PLoS ONE. 2009;4(1):e4227.PubMedPubMedCentralCrossRef Young JW, Light GA, Marston HM, Sharp R, Geyer MA. The 5-choice continuous performance test: evidence for a translational test of vigilance for mice. PLoS ONE. 2009;4(1):e4227.PubMedPubMedCentralCrossRef
121.
go back to reference Bari A, Theobald DE, Caprioli D, Mar AC, Aidoo-Micah A, Dalley JW, et al. Serotonin modulates sensitivity to reward and negative feedback in a probabilistic reversal learning task in rats. Neuropsychopharmacology. 2010;35(6):1290–301.PubMedPubMedCentralCrossRef Bari A, Theobald DE, Caprioli D, Mar AC, Aidoo-Micah A, Dalley JW, et al. Serotonin modulates sensitivity to reward and negative feedback in a probabilistic reversal learning task in rats. Neuropsychopharmacology. 2010;35(6):1290–301.PubMedPubMedCentralCrossRef
122.
go back to reference Culbreth AJ, Gold JM, Cools R, Barch DM. Impaired activation in cognitive control regions predicts reversal learning in schizophrenia. Schizophr Bull. 2016;42(2):484–93.PubMedCrossRef Culbreth AJ, Gold JM, Cools R, Barch DM. Impaired activation in cognitive control regions predicts reversal learning in schizophrenia. Schizophr Bull. 2016;42(2):484–93.PubMedCrossRef
123.
go back to reference Reddy LF, Waltz JA, Green MF, Wynn JK, Horan WP. Probabilistic reversal learning in schizophrenia: stability of deficits and potential causal mechanisms. Schizophr Bull. 2016;42(4):942–51.PubMedPubMedCentralCrossRef Reddy LF, Waltz JA, Green MF, Wynn JK, Horan WP. Probabilistic reversal learning in schizophrenia: stability of deficits and potential causal mechanisms. Schizophr Bull. 2016;42(4):942–51.PubMedPubMedCentralCrossRef
124.
go back to reference Linke J, King AV, Rietschel M, Strohmaier J, Hennerici M, Gass A, et al. Increased medial orbitofrontal and amygdala activation: evidence for a systems-level endophenotype of bipolar I disorder. Am J Psychiatry. 2012;169(3):316–25.PubMedCrossRef Linke J, King AV, Rietschel M, Strohmaier J, Hennerici M, Gass A, et al. Increased medial orbitofrontal and amygdala activation: evidence for a systems-level endophenotype of bipolar I disorder. Am J Psychiatry. 2012;169(3):316–25.PubMedCrossRef
125.
go back to reference Mukherjee D, Filipowicz ALS, Vo K, Satterthwaite TD, Kable JW. Reward and punishment reversal-learning in major depressive disorder. J Abnorm Psychol. 2020;129(8):810–23.PubMedCrossRef Mukherjee D, Filipowicz ALS, Vo K, Satterthwaite TD, Kable JW. Reward and punishment reversal-learning in major depressive disorder. J Abnorm Psychol. 2020;129(8):810–23.PubMedCrossRef
126.
go back to reference Amitai N, Young JW, Higa K, Sharp RF, Geyer MA, Powell SB. Isolation rearing effects on probabilistic learning and cognitive flexibility in rats. Cogn Affect Behav Neurosci. 2014;14(1):388–406.PubMedPubMedCentralCrossRef Amitai N, Young JW, Higa K, Sharp RF, Geyer MA, Powell SB. Isolation rearing effects on probabilistic learning and cognitive flexibility in rats. Cogn Affect Behav Neurosci. 2014;14(1):388–406.PubMedPubMedCentralCrossRef
127.
go back to reference Milienne-Petiot M, Kesby JP, Graves M, van Enkhuizen J, Semenova S, Minassian A, et al. The effects of reduced dopamine transporter function and chronic lithium on motivation, probabilistic learning, and neurochemistry in mice: modeling bipolar mania. Neuropharmacology. 2017;113(Pt A):260–70.PubMedCrossRef Milienne-Petiot M, Kesby JP, Graves M, van Enkhuizen J, Semenova S, Minassian A, et al. The effects of reduced dopamine transporter function and chronic lithium on motivation, probabilistic learning, and neurochemistry in mice: modeling bipolar mania. Neuropharmacology. 2017;113(Pt A):260–70.PubMedCrossRef
128.
go back to reference Roberts BZ, Young JW, He YV, Cope ZA, Shilling PD, Feifel D. Oxytocin improves probabilistic reversal learning but not effortful motivation in Brown Norway rats. Neuropharmacology. 2019;150:15–26.PubMedPubMedCentralCrossRef Roberts BZ, Young JW, He YV, Cope ZA, Shilling PD, Feifel D. Oxytocin improves probabilistic reversal learning but not effortful motivation in Brown Norway rats. Neuropharmacology. 2019;150:15–26.PubMedPubMedCentralCrossRef
129.
go back to reference Tranter MM, Aggarwal S, Young JW, Dillon DG, Barnes SA. Reinforcement learning deficits exhibited by postnatal PCP-treated rats enable deep neural network classification. Neuropsychopharmacology. 2023;48(9):1377–85.PubMedCrossRef Tranter MM, Aggarwal S, Young JW, Dillon DG, Barnes SA. Reinforcement learning deficits exhibited by postnatal PCP-treated rats enable deep neural network classification. Neuropsychopharmacology. 2023;48(9):1377–85.PubMedCrossRef
130.
go back to reference Moradi AR, Miraghaei MA, Parhon H, Jabbari H, Jobson L. Posttraumatic stress disorder, depression, executive functioning, and autobiographical remembering in individuals with HIV and in carers of those with HIV in Iran. AIDS Care. 2013;25(3):281–8.PubMedCrossRef Moradi AR, Miraghaei MA, Parhon H, Jabbari H, Jobson L. Posttraumatic stress disorder, depression, executive functioning, and autobiographical remembering in individuals with HIV and in carers of those with HIV in Iran. AIDS Care. 2013;25(3):281–8.PubMedCrossRef
131.
go back to reference Chang L, Lim A, Lau E, Alicata D. Chronic tobacco-smoking on psychopathological symptoms, impulsivity and cognitive deficits in HIV-infected individuals. J Neuroimmune Pharmacol. 2017;12(3):389–401.PubMedPubMedCentralCrossRef Chang L, Lim A, Lau E, Alicata D. Chronic tobacco-smoking on psychopathological symptoms, impulsivity and cognitive deficits in HIV-infected individuals. J Neuroimmune Pharmacol. 2017;12(3):389–401.PubMedPubMedCentralCrossRef
132.
go back to reference Kanmogne GD, Fonsah JY, Tang B, Doh RF, Kengne AM, Umlauf A, et al. Effects of HIV on executive function and verbal fluency in Cameroon. Sci Rep. 2018;8(1):17794.PubMedPubMedCentralCrossRef Kanmogne GD, Fonsah JY, Tang B, Doh RF, Kengne AM, Umlauf A, et al. Effects of HIV on executive function and verbal fluency in Cameroon. Sci Rep. 2018;8(1):17794.PubMedPubMedCentralCrossRef
133.
go back to reference Sanford R, Fellows LK, Ances BM, Collins DL. Association of brain structure changes and cognitive function with combination antiretroviral therapy in HIV-positive individuals. JAMA Neurol. 2018;75(1):72–9.PubMedCrossRef Sanford R, Fellows LK, Ances BM, Collins DL. Association of brain structure changes and cognitive function with combination antiretroviral therapy in HIV-positive individuals. JAMA Neurol. 2018;75(1):72–9.PubMedCrossRef
134.
go back to reference Alakkas A, Ellis RJ, Watson CW, Umlauf A, Heaton RK, Letendre S, et al. White matter damage, neuroinflammation, and neuronal integrity in HAND. J Neurovirol. 2019;25(1):32–41.PubMedCrossRef Alakkas A, Ellis RJ, Watson CW, Umlauf A, Heaton RK, Letendre S, et al. White matter damage, neuroinflammation, and neuronal integrity in HAND. J Neurovirol. 2019;25(1):32–41.PubMedCrossRef
135.
go back to reference Patel SH, Kolson DL, Glosser G, Matozzo I, Ge Y, Babb JS, et al. Correlation between percentage of brain parenchymal volume and neurocognitive performance in HIV-infected patients. AJNR Am J Neuroradiol. 2002;23(4):543–9.PubMedPubMedCentral Patel SH, Kolson DL, Glosser G, Matozzo I, Ge Y, Babb JS, et al. Correlation between percentage of brain parenchymal volume and neurocognitive performance in HIV-infected patients. AJNR Am J Neuroradiol. 2002;23(4):543–9.PubMedPubMedCentral
136.
go back to reference Cohen RA, Harezlak J, Schifitto G, Hana G, Clark U, Gongvatana A, et al. Effects of nadir CD4 count and duration of human immunodeficiency virus infection on brain volumes in the highly active antiretroviral therapy era. J Neurovirol. 2010;16(1):25–32.PubMedPubMedCentralCrossRef Cohen RA, Harezlak J, Schifitto G, Hana G, Clark U, Gongvatana A, et al. Effects of nadir CD4 count and duration of human immunodeficiency virus infection on brain volumes in the highly active antiretroviral therapy era. J Neurovirol. 2010;16(1):25–32.PubMedPubMedCentralCrossRef
137.
go back to reference Thames AD, Foley JM, Wright MJ, Panos SE, Ettenhofer M, Ramezani A, et al. Basal ganglia structures differentially contribute to verbal fluency: evidence from human immunodeficiency virus (HIV)-infected adults. Neuropsychologia. 2012;50(3):390–5.PubMedCrossRef Thames AD, Foley JM, Wright MJ, Panos SE, Ettenhofer M, Ramezani A, et al. Basal ganglia structures differentially contribute to verbal fluency: evidence from human immunodeficiency virus (HIV)-infected adults. Neuropsychologia. 2012;50(3):390–5.PubMedCrossRef
138.
go back to reference Bonnet F, Amieva H, Marquant F, Bernard C, Bruyand M, Dauchy FA, et al. Cognitive disorders in HIV-infected patients: are they HIV-related? AIDS. 2013;27(3):391–400.PubMedCrossRef Bonnet F, Amieva H, Marquant F, Bernard C, Bruyand M, Dauchy FA, et al. Cognitive disorders in HIV-infected patients: are they HIV-related? AIDS. 2013;27(3):391–400.PubMedCrossRef
139.
go back to reference Kuper M, Rabe K, Esser S, Gizewski ER, Husstedt IW, Maschke M, et al. Structural gray and white matter changes in patients with HIV. J Neurol. 2011;258(6):1066–75.PubMedCrossRef Kuper M, Rabe K, Esser S, Gizewski ER, Husstedt IW, Maschke M, et al. Structural gray and white matter changes in patients with HIV. J Neurol. 2011;258(6):1066–75.PubMedCrossRef
140.
go back to reference Watson C, Busovaca E, Foley JM, Allen IE, Schwarz CG, Jahanshad N, et al. White matter hyperintensities correlate to cognition and fiber tract integrity in older adults with HIV. J Neurovirol. 2017;23(3):422–9.PubMedPubMedCentralCrossRef Watson C, Busovaca E, Foley JM, Allen IE, Schwarz CG, Jahanshad N, et al. White matter hyperintensities correlate to cognition and fiber tract integrity in older adults with HIV. J Neurovirol. 2017;23(3):422–9.PubMedPubMedCentralCrossRef
141.
go back to reference Su T, Wit FW, Caan MW, Schouten J, Prins M, Geurtsen GJ, et al. White matter hyperintensities in relation to cognition in HIV-infected men with sustained suppressed viral load on combination antiretroviral therapy. AIDS. 2016;30(15):2329–39.PubMedCrossRef Su T, Wit FW, Caan MW, Schouten J, Prins M, Geurtsen GJ, et al. White matter hyperintensities in relation to cognition in HIV-infected men with sustained suppressed viral load on combination antiretroviral therapy. AIDS. 2016;30(15):2329–39.PubMedCrossRef
142.
go back to reference Connolly CG, Bischoff-Grethe A, Jordan SJ, Woods SP, Ellis RJ, Paulus MP, et al. Altered functional response to risky choice in HIV infection. PLoS ONE. 2014;9(10):e111583.PubMedPubMedCentralCrossRef Connolly CG, Bischoff-Grethe A, Jordan SJ, Woods SP, Ellis RJ, Paulus MP, et al. Altered functional response to risky choice in HIV infection. PLoS ONE. 2014;9(10):e111583.PubMedPubMedCentralCrossRef
143.
go back to reference Chang L, Speck O, Miller EN, Braun J, Jovicich J, Koch C, et al. Neural correlates of attention and working memory deficits in HIV patients. Neurology. 2001;57(6):1001–7.PubMedCrossRef Chang L, Speck O, Miller EN, Braun J, Jovicich J, Koch C, et al. Neural correlates of attention and working memory deficits in HIV patients. Neurology. 2001;57(6):1001–7.PubMedCrossRef
144.
go back to reference Ernst T, Chang L, Jovicich J, Ames N, Arnold S. Abnormal brain activation on functional MRI in cognitively asymptomatic HIV patients. Neurology. 2002;59(9):1343–9.PubMedCrossRef Ernst T, Chang L, Jovicich J, Ames N, Arnold S. Abnormal brain activation on functional MRI in cognitively asymptomatic HIV patients. Neurology. 2002;59(9):1343–9.PubMedCrossRef
145.
go back to reference Ernst T, Yakupov R, Nakama H, Crocket G, Cole M, Watters M, et al. Declined neural efficiency in cognitively stable human immunodeficiency virus patients. Ann Neurol. 2009;65(3):316–25.PubMedPubMedCentralCrossRef Ernst T, Yakupov R, Nakama H, Crocket G, Cole M, Watters M, et al. Declined neural efficiency in cognitively stable human immunodeficiency virus patients. Ann Neurol. 2009;65(3):316–25.PubMedPubMedCentralCrossRef
146.
go back to reference Chang L, Tomasi D, Yakupov R, Lozar C, Arnold S, Caparelli E, et al. Adaptation of the attention network in human immunodeficiency virus brain injury. Ann Neurol. 2004;56(2):259–72.PubMedCrossRef Chang L, Tomasi D, Yakupov R, Lozar C, Arnold S, Caparelli E, et al. Adaptation of the attention network in human immunodeficiency virus brain injury. Ann Neurol. 2004;56(2):259–72.PubMedCrossRef
147.
go back to reference Melrose RJ, Tinaz S, Castelo JM, Courtney MG, Stern CE. Compromised fronto-striatal functioning in HIV: an fMRI investigation of semantic event sequencing. Behav Brain Res. 2008;188(2):337–47.PubMedCrossRef Melrose RJ, Tinaz S, Castelo JM, Courtney MG, Stern CE. Compromised fronto-striatal functioning in HIV: an fMRI investigation of semantic event sequencing. Behav Brain Res. 2008;188(2):337–47.PubMedCrossRef
148.
go back to reference Maki PM, Cohen MH, Weber K, Little DM, Fornelli D, Rubin LH, et al. Impairments in memory and hippocampal function in HIV-positive vs HIV-negative women: a preliminary study. Neurology. 2009;72(19):1661–8.PubMedPubMedCentralCrossRef Maki PM, Cohen MH, Weber K, Little DM, Fornelli D, Rubin LH, et al. Impairments in memory and hippocampal function in HIV-positive vs HIV-negative women: a preliminary study. Neurology. 2009;72(19):1661–8.PubMedPubMedCentralCrossRef
149.
go back to reference Wiesman AI, O’Neill J, Mills MS, Robertson KR, Fox HS, Swindells S, et al. Aberrant occipital dynamics differentiate HIV-infected patients with and without cognitive impairment. Brain. 2018;141(6):1678–90.PubMedPubMedCentralCrossRef Wiesman AI, O’Neill J, Mills MS, Robertson KR, Fox HS, Swindells S, et al. Aberrant occipital dynamics differentiate HIV-infected patients with and without cognitive impairment. Brain. 2018;141(6):1678–90.PubMedPubMedCentralCrossRef
150.
go back to reference Hall SA, Lalee Z, Bell RP, Towe SL, Meade CS. Synergistic effects of HIV and marijuana use on functional brain network organization. Prog Neuropsychopharmacol Biol Psychiatry. 2021;104:110040.PubMedCrossRef Hall SA, Lalee Z, Bell RP, Towe SL, Meade CS. Synergistic effects of HIV and marijuana use on functional brain network organization. Prog Neuropsychopharmacol Biol Psychiatry. 2021;104:110040.PubMedCrossRef
151.
go back to reference Chang L, Ernst T, Witt MD, Ames N, Gaiefsky M, Miller E. Relationships among brain metabolites, cognitive function, and viral loads in antiretroviral-naive HIV patients. Neuroimage. 2002;17(3):1638–48.PubMedCrossRef Chang L, Ernst T, Witt MD, Ames N, Gaiefsky M, Miller E. Relationships among brain metabolites, cognitive function, and viral loads in antiretroviral-naive HIV patients. Neuroimage. 2002;17(3):1638–48.PubMedCrossRef
152.
go back to reference Vitiello B, Goodkin K, Ashtana D, Shapshak P, Atkinson JH, Heseltine PN, et al. HIV-1 RNA concentration and cognitive performance in a cohort of HIV-positive people. AIDS. 2007;21(11):1415–22.PubMedCrossRef Vitiello B, Goodkin K, Ashtana D, Shapshak P, Atkinson JH, Heseltine PN, et al. HIV-1 RNA concentration and cognitive performance in a cohort of HIV-positive people. AIDS. 2007;21(11):1415–22.PubMedCrossRef
153.
go back to reference Zhang Y, Qiao L, Ding W, Wei F, Zhao Q, Wang X, et al. An initial screening for HIV-associated neurocognitive disorders of HIV-1 infected patients in China. J Neurovirol. 2012;18(2):120–6.PubMedCrossRef Zhang Y, Qiao L, Ding W, Wei F, Zhao Q, Wang X, et al. An initial screening for HIV-associated neurocognitive disorders of HIV-1 infected patients in China. J Neurovirol. 2012;18(2):120–6.PubMedCrossRef
154.
go back to reference Ellis RJ, Peterson SN, Li Y, Schrier R, Iudicello J, Letendre S, Morgan E, Tang B, Grant I, Cherner M. Recent cannabis use in HIV is associated with reduced inflammatory markers in CSF and blood. Neurology(R) neuroimmunology & neuroinflammation, 2020;7(5):e809. https://doi.org/10.1212/NXI.0000000000000809.  Ellis RJ, Peterson SN, Li Y, Schrier R, Iudicello J, Letendre S, Morgan E, Tang B, Grant I, Cherner M. Recent cannabis use in HIV is associated with reduced inflammatory markers in CSF and blood. Neurology(R) neuroimmunology & neuroinflammation, 2020;7(5):e809. https://​doi.​org/​10.​1212/​NXI.​0000000000000809​.
155.
go back to reference Marcellin F, Lions C, Rosenthal E, Roux P, Sogni P, Wittkop L, et al. No significant effect of cannabis use on the count and percentage of circulating CD4 T-cells in HIV-HCV co-infected patients (ANRS CO13-HEPAVIH French cohort). Drug Alcohol Rev. 2017;36(2):227–38.PubMedCrossRef Marcellin F, Lions C, Rosenthal E, Roux P, Sogni P, Wittkop L, et al. No significant effect of cannabis use on the count and percentage of circulating CD4 T-cells in HIV-HCV co-infected patients (ANRS CO13-HEPAVIH French cohort). Drug Alcohol Rev. 2017;36(2):227–38.PubMedCrossRef
156.
go back to reference Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, et al. Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann Intern Med. 2003;139(4):258–66.PubMedCrossRef Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, et al. Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann Intern Med. 2003;139(4):258–66.PubMedCrossRef
157.
go back to reference Bredt BM, Higuera-Alhino D, Shade SB, Hebert SJ, McCune JM, Abrams DI. Short-term effects of cannabinoids on immune phenotype and function in HIV-1-infected patients. J Clin Pharmacol. 2002;42(S1):82S-S89.PubMedCrossRef Bredt BM, Higuera-Alhino D, Shade SB, Hebert SJ, McCune JM, Abrams DI. Short-term effects of cannabinoids on immune phenotype and function in HIV-1-infected patients. J Clin Pharmacol. 2002;42(S1):82S-S89.PubMedCrossRef
158.
go back to reference Manuzak JA, Gott TM, Kirkwood JS, Coronado E, Hensley-McBain T, Miller C, et al. 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):1872–82.PubMedPubMedCentralCrossRef Manuzak JA, Gott TM, Kirkwood JS, Coronado E, Hensley-McBain T, Miller C, et al. 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):1872–82.PubMedPubMedCentralCrossRef
159.
go back to reference Burlacu R, Umlauf A, Marcotte TD, Soontornniyomkij B, Diaconu CC, Bulacu-Talnariu A, et al. Plasma CXCL10 correlates with HAND in HIV-infected women. J Neurovirol. 2020;26(1):23–31.PubMedCrossRef Burlacu R, Umlauf A, Marcotte TD, Soontornniyomkij B, Diaconu CC, Bulacu-Talnariu A, et al. Plasma CXCL10 correlates with HAND in HIV-infected women. J Neurovirol. 2020;26(1):23–31.PubMedCrossRef
160.
go back to reference Portilla I, Reus S, Leon R, van-der Hofstadt C, Sanchez J, Lopez N, et al. Neurocognitive impairment in well-controlled HIV-infected patients: a cross-sectional study. AIDS Res Hum Retrovir. 2019;35(7):634–41.PubMedCrossRef Portilla I, Reus S, Leon R, van-der Hofstadt C, Sanchez J, Lopez N, et al. Neurocognitive impairment in well-controlled HIV-infected patients: a cross-sectional study. AIDS Res Hum Retrovir. 2019;35(7):634–41.PubMedCrossRef
161.
go back to reference Yin L, Dinasarapu AR, Borkar SA, Chang KF, De Paris K, Kim-Chang JJ, et al. Anti-inflammatory effects of recreational marijuana in virally suppressed youth with HIV-1 are reversed by use of tobacco products in combination with marijuana. Retrovirology. 2022;19(1):10.PubMedPubMedCentralCrossRef Yin L, Dinasarapu AR, Borkar SA, Chang KF, De Paris K, Kim-Chang JJ, et al. Anti-inflammatory effects of recreational marijuana in virally suppressed youth with HIV-1 are reversed by use of tobacco products in combination with marijuana. Retrovirology. 2022;19(1):10.PubMedPubMedCentralCrossRef
162.
go back to reference Eugenin EA, Osiecki K, Lopez L, Goldstein H, Calderon TM, Berman JW. CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigration of human immunodeficiency virus (HIV)-infected leukocytes across the blood-brain barrier: a potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci. 2006;26(4):1098–106.PubMedPubMedCentralCrossRef Eugenin EA, Osiecki K, Lopez L, Goldstein H, Calderon TM, Berman JW. CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigration of human immunodeficiency virus (HIV)-infected leukocytes across the blood-brain barrier: a potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci. 2006;26(4):1098–106.PubMedPubMedCentralCrossRef
163.
go back to reference Anderson AM, Jang JH, Easley KA, Fuchs D, Gisslen M, Zetterberg H, et al. Cognitive and neuronal link with inflammation: a longitudinal study in people with and without HIV infection. J Acquir Immune Defic Syndr. 2020;85(5):617–25.PubMedPubMedCentralCrossRef Anderson AM, Jang JH, Easley KA, Fuchs D, Gisslen M, Zetterberg H, et al. Cognitive and neuronal link with inflammation: a longitudinal study in people with and without HIV infection. J Acquir Immune Defic Syndr. 2020;85(5):617–25.PubMedPubMedCentralCrossRef
164.
go back to reference Sanchez-Sanchez JL, Giudici KV, Guyonnet S, Delrieu J, Li Y, Bateman RJ, et al. Plasma MCP-1 and changes on cognitive function in community-dwelling older adults. Alzheimers Res Ther. 2022;14(1):5.PubMedPubMedCentralCrossRef Sanchez-Sanchez JL, Giudici KV, Guyonnet S, Delrieu J, Li Y, Bateman RJ, et al. Plasma MCP-1 and changes on cognitive function in community-dwelling older adults. Alzheimers Res Ther. 2022;14(1):5.PubMedPubMedCentralCrossRef
165.
go back to reference Lee WJ, Liao YC, Wang YF, Lin IF, Wang SJ, Fuh JL. Plasma MCP-1 and cognitive decline in patients with Alzheimer’s disease and mild cognitive impairment: a two-year follow-up study. Sci Rep. 2018;8(1):1280.PubMedPubMedCentralCrossRef Lee WJ, Liao YC, Wang YF, Lin IF, Wang SJ, Fuh JL. Plasma MCP-1 and cognitive decline in patients with Alzheimer’s disease and mild cognitive impairment: a two-year follow-up study. Sci Rep. 2018;8(1):1280.PubMedPubMedCentralCrossRef
166.
go back to reference Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J Leukoc Biol. 2012;91(3):401–15.PubMedPubMedCentralCrossRef Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J Leukoc Biol. 2012;91(3):401–15.PubMedPubMedCentralCrossRef
167.
168.
go back to reference Calcagno A, Atzori C, Romito A, Vai D, Audagnotto S, Stella ML, et al. Blood brain barrier impairment is associated with cerebrospinal fluid markers of neuronal damage in HIV-positive patients. J Neurovirol. 2016;22(1):88–92.PubMedCrossRef Calcagno A, Atzori C, Romito A, Vai D, Audagnotto S, Stella ML, et al. Blood brain barrier impairment is associated with cerebrospinal fluid markers of neuronal damage in HIV-positive patients. J Neurovirol. 2016;22(1):88–92.PubMedCrossRef
169.
go back to reference Ellis RJ, Peterson S, Cherner M, Morgan E, Schrier R, Tang B, et al. Beneficial effects of cannabis on blood-brain barrier function in human immunodeficiency virus. Clin Infect Dis. 2021;73(1):124–9.PubMedCrossRef Ellis RJ, Peterson S, Cherner M, Morgan E, Schrier R, Tang B, et al. Beneficial effects of cannabis on blood-brain barrier function in human immunodeficiency virus. Clin Infect Dis. 2021;73(1):124–9.PubMedCrossRef
171.
go back to reference Hinkin CH, Castellon SA, Levine AJ, Barclay TR, Singer EJ. Neurocognition in individuals co-infected with HIV and hepatitis C. J Addict Dis. 2008;27(2):11–7.PubMedPubMedCentralCrossRef Hinkin CH, Castellon SA, Levine AJ, Barclay TR, Singer EJ. Neurocognition in individuals co-infected with HIV and hepatitis C. J Addict Dis. 2008;27(2):11–7.PubMedPubMedCentralCrossRef
172.
go back to reference Namagga JK, Rukundo GZ, Niyonzima V, Voss J. Depression and HIV associated neurocognitive disorders among HIV infected adults in rural southwestern Uganda: a cross-sectional quantitative study. BMC Psychiatry. 2021;21(1):350.PubMedPubMedCentralCrossRef Namagga JK, Rukundo GZ, Niyonzima V, Voss J. Depression and HIV associated neurocognitive disorders among HIV infected adults in rural southwestern Uganda: a cross-sectional quantitative study. BMC Psychiatry. 2021;21(1):350.PubMedPubMedCentralCrossRef
173.
go back to reference Gutierrez J, Porras TN, Yoo-Jeong M, Khasiyev F, Igwe KC, Laing KK, et al. Cerebrovascular contributions to neurocognitive disorders in people living with HIV. J Acquir Immune Defic Syndr. 2021;88(1):79–85.PubMedPubMedCentralCrossRef Gutierrez J, Porras TN, Yoo-Jeong M, Khasiyev F, Igwe KC, Laing KK, et al. Cerebrovascular contributions to neurocognitive disorders in people living with HIV. J Acquir Immune Defic Syndr. 2021;88(1):79–85.PubMedPubMedCentralCrossRef
174.
go back to reference Wright EJ, Grund B, Robertson K, Brew BJ, Roediger M, Bain MP, et al. Cardiovascular risk factors associated with lower baseline cognitive performance in HIV-positive persons. Neurology. 2010;75(10):864–73.PubMedPubMedCentralCrossRef Wright EJ, Grund B, Robertson K, Brew BJ, Roediger M, Bain MP, et al. Cardiovascular risk factors associated with lower baseline cognitive performance in HIV-positive persons. Neurology. 2010;75(10):864–73.PubMedPubMedCentralCrossRef
175.
go back to reference Becker JT, Kingsley L, Mullen J, Cohen B, Martin E, Miller EN, et al. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology. 2009;73(16):1292–9.PubMedPubMedCentralCrossRef Becker JT, Kingsley L, Mullen J, Cohen B, Martin E, Miller EN, et al. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology. 2009;73(16):1292–9.PubMedPubMedCentralCrossRef
176.
go back to reference Foley J, Ettenhofer M, Wright MJ, Siddiqi I, Choi M, Thames AD, et al. Neurocognitive functioning in HIV-1 infection: effects of cerebrovascular risk factors and age. Clin Neuropsychol. 2010;24(2):265–85.PubMedPubMedCentralCrossRef Foley J, Ettenhofer M, Wright MJ, Siddiqi I, Choi M, Thames AD, et al. Neurocognitive functioning in HIV-1 infection: effects of cerebrovascular risk factors and age. Clin Neuropsychol. 2010;24(2):265–85.PubMedPubMedCentralCrossRef
177.
go back to reference Avraham HK, Jiang S, Fu Y, Rockenstein E, Makriyannis A, Zvonok A, et al. The cannabinoid CB(2) receptor agonist AM1241 enhances neurogenesis in GFAP/Gp120 transgenic mice displaying deficits in neurogenesis. Br J Pharmacol. 2014;171(2):468–79.PubMedCrossRef Avraham HK, Jiang S, Fu Y, Rockenstein E, Makriyannis A, Zvonok A, et al. The cannabinoid CB(2) receptor agonist AM1241 enhances neurogenesis in GFAP/Gp120 transgenic mice displaying deficits in neurogenesis. Br J Pharmacol. 2014;171(2):468–79.PubMedCrossRef
178.
go back to reference Kim HJ, Shin AH, Thayer SA. Activation of cannabinoid type 2 receptors inhibits HIV-1 envelope glycoprotein gp120-induced synapse loss. Mol Pharmacol. 2011;80(3):357–66.PubMedPubMedCentralCrossRef Kim HJ, Shin AH, Thayer SA. Activation of cannabinoid type 2 receptors inhibits HIV-1 envelope glycoprotein gp120-induced synapse loss. Mol Pharmacol. 2011;80(3):357–66.PubMedPubMedCentralCrossRef
179.
180.
go back to reference Zhang X, Thayer SA. Monoacylglycerol lipase inhibitor JZL184 prevents HIV-1 gp120-induced synapse loss by altering endocannabinoid signaling. Neuropharmacology. 2018;128:269–81.PubMedCrossRef Zhang X, Thayer SA. Monoacylglycerol lipase inhibitor JZL184 prevents HIV-1 gp120-induced synapse loss by altering endocannabinoid signaling. Neuropharmacology. 2018;128:269–81.PubMedCrossRef
181.
go back to reference Esposito G, Ligresti A, Izzo AA, Bisogno T, Ruvo M, Di Rosa M, et al. The endocannabinoid system protects rat glioma cells against HIV-1 Tat protein-induced cytotoxicity Mechanism and regulation. J Biol Chem. 2002;277(52):50348–54.PubMedCrossRef Esposito G, Ligresti A, Izzo AA, Bisogno T, Ruvo M, Di Rosa M, et al. The endocannabinoid system protects rat glioma cells against HIV-1 Tat protein-induced cytotoxicity Mechanism and regulation. J Biol Chem. 2002;277(52):50348–54.PubMedCrossRef
182.
go back to reference Avraham HK, Jiang S, Fu Y, Rockenstein E, Makriyannis A, Wood J, et al. Impaired neurogenesis by HIV-1-Gp120 is rescued by genetic deletion of fatty acid amide hydrolase enzyme. Br J Pharmacol. 2015;172(19):4603–14.PubMedPubMedCentralCrossRef Avraham HK, Jiang S, Fu Y, Rockenstein E, Makriyannis A, Wood J, et al. Impaired neurogenesis by HIV-1-Gp120 is rescued by genetic deletion of fatty acid amide hydrolase enzyme. Br J Pharmacol. 2015;172(19):4603–14.PubMedPubMedCentralCrossRef
183.
go back to reference Hermes DJ, Yadav-Samudrala BJ, Xu C, Paniccia JE, Meeker RB, Armstrong ML, et al. GPR18 drives FAAH inhibition-induced neuroprotection against HIV-1 Tat-induced neurodegeneration. Exp Neurol. 2021;341:113699.PubMedPubMedCentralCrossRef Hermes DJ, Yadav-Samudrala BJ, Xu C, Paniccia JE, Meeker RB, Armstrong ML, et al. GPR18 drives FAAH inhibition-induced neuroprotection against HIV-1 Tat-induced neurodegeneration. Exp Neurol. 2021;341:113699.PubMedPubMedCentralCrossRef
184.
go back to reference Hermes DJ, Xu C, Poklis JL, Niphakis MJ, Cravatt BF, Mackie K, et al. Neuroprotective effects of fatty acid amide hydrolase catabolic enzyme inhibition in a HIV-1 Tat model of neuroAIDS. Neuropharmacology. 2018;141:55–65.PubMedPubMedCentralCrossRef Hermes DJ, Xu C, Poklis JL, Niphakis MJ, Cravatt BF, Mackie K, et al. Neuroprotective effects of fatty acid amide hydrolase catabolic enzyme inhibition in a HIV-1 Tat model of neuroAIDS. Neuropharmacology. 2018;141:55–65.PubMedPubMedCentralCrossRef
185.
go back to reference Bisogno T, De Petrocellis L, Di Marzo V. Fatty acid amide hydrolase, an enzyme with many bioactive substrates Possible therapeutic implications. Curr Pharm Des. 2002;8(7):533–47.PubMedCrossRef Bisogno T, De Petrocellis L, Di Marzo V. Fatty acid amide hydrolase, an enzyme with many bioactive substrates Possible therapeutic implications. Curr Pharm Des. 2002;8(7):533–47.PubMedCrossRef
186.
go back to reference Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996;384(6604):83–7.PubMedCrossRef Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996;384(6604):83–7.PubMedCrossRef
187.
go back to reference Xu C, Hermes DJ, Mackie K, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Cannabinoids occlude the HIV-1 Tat-induced decrease in GABAergic neurotransmission in prefrontal cortex slices. J Neuroimmune Pharmacol. 2016;11(2):316–31.PubMedPubMedCentralCrossRef Xu C, Hermes DJ, Mackie K, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Cannabinoids occlude the HIV-1 Tat-induced decrease in GABAergic neurotransmission in prefrontal cortex slices. J Neuroimmune Pharmacol. 2016;11(2):316–31.PubMedPubMedCentralCrossRef
188.
go back to reference Xu C, Yadav-Samudrala BJ, Xu C, Nath B, Mistry T, Jiang W, Niphakis MJ, Cravatt BF, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Inhibitory neurotransmission is sex-dependently affected by tat expression in transgenic mice and suppressed by the fatty acid amide hydrolase enzyme inhibitor pf3845 via cannabinoid type-1 receptor mechanisms. Cells, 2022;11(5):857. https://doi.org/10.3390/cells11050857. Xu C, Yadav-Samudrala BJ, Xu C, Nath B, Mistry T, Jiang W, Niphakis MJ, Cravatt BF, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Inhibitory neurotransmission is sex-dependently affected by tat expression in transgenic mice and suppressed by the fatty acid amide hydrolase enzyme inhibitor pf3845 via cannabinoid type-1 receptor mechanisms. Cells, 2022;11(5):857. https://​doi.​org/​10.​3390/​cells11050857.
190.
go back to reference Little PJ, Compton DR, Johnson MR, Melvin LS, Martin BR. Pharmacology and stereoselectivity of structurally novel cannabinoids in mice. J Pharmacol Exp Ther. 1988;247(3):1046–51.PubMed Little PJ, Compton DR, Johnson MR, Melvin LS, Martin BR. Pharmacology and stereoselectivity of structurally novel cannabinoids in mice. J Pharmacol Exp Ther. 1988;247(3):1046–51.PubMed
191.
go back to reference Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009;23(1):133–44.PubMedCrossRef Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009;23(1):133–44.PubMedCrossRef
192.
go back to reference Wang XF, Galaj E, Bi GH, Zhang C, He Y, Zhan J, et al. Different receptor mechanisms underlying phytocannabinoid- versus synthetic cannabinoid-induced tetrad effects: opposite roles of CB(1) /CB(2) versus GPR55 receptors. Br J Pharmacol. 2020;177(8):1865–80.PubMedPubMedCentralCrossRef Wang XF, Galaj E, Bi GH, Zhang C, He Y, Zhan J, et al. Different receptor mechanisms underlying phytocannabinoid- versus synthetic cannabinoid-induced tetrad effects: opposite roles of CB(1) /CB(2) versus GPR55 receptors. Br J Pharmacol. 2020;177(8):1865–80.PubMedPubMedCentralCrossRef
193.
go back to reference van Egmond N, Straub VM, van der Stelt M. Targeting endocannabinoid signaling: FAAH and MAG lipase inhibitors. Annu Rev Pharmacol Toxicol. 2021;61:441–63.PubMedCrossRef van Egmond N, Straub VM, van der Stelt M. Targeting endocannabinoid signaling: FAAH and MAG lipase inhibitors. Annu Rev Pharmacol Toxicol. 2021;61:441–63.PubMedCrossRef
194.
go back to reference Li GL, Winter H, Arends R, Jay GW, Le V, Young T, et al. Assessment of the pharmacology and tolerability of PF-04457845, an irreversible inhibitor of fatty acid amide hydrolase-1, in healthy subjects. Br J Clin Pharmacol. 2012;73(5):706–16.PubMedCrossRef Li GL, Winter H, Arends R, Jay GW, Le V, Young T, et al. Assessment of the pharmacology and tolerability of PF-04457845, an irreversible inhibitor of fatty acid amide hydrolase-1, in healthy subjects. Br J Clin Pharmacol. 2012;73(5):706–16.PubMedCrossRef
195.
go back to reference Mayo LM, Asratian A, Linde J, Morena M, Haataja R, Hammar V, et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol Psychiatry. 2020;87(6):538–47.PubMedCrossRef Mayo LM, Asratian A, Linde J, Morena M, Haataja R, Hammar V, et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol Psychiatry. 2020;87(6):538–47.PubMedCrossRef
196.
go back to reference Huggins JP, Smart TS, Langman S, Taylor L, Young T. An efficient randomised, placebo-controlled clinical trial with the irreversible fatty acid amide hydrolase-1 inhibitor PF-04457845, which modulates endocannabinoids but fails to induce effective analgesia in patients with pain due to osteoarthritis of the knee. Pain. 2012;153(9):1837–46.PubMedCrossRef Huggins JP, Smart TS, Langman S, Taylor L, Young T. An efficient randomised, placebo-controlled clinical trial with the irreversible fatty acid amide hydrolase-1 inhibitor PF-04457845, which modulates endocannabinoids but fails to induce effective analgesia in patients with pain due to osteoarthritis of the knee. Pain. 2012;153(9):1837–46.PubMedCrossRef
197.
go back to reference D’Souza DC, Cortes-Briones J, Creatura G, Bluez G, Thurnauer H, Deaso E, et al. Efficacy and safety of a fatty acid amide hydrolase inhibitor (PF-04457845) in the treatment of cannabis withdrawal and dependence in men: a double-blind, placebo-controlled, parallel group, phase 2a single-site randomised controlled trial. Lancet Psychiatry. 2019;6(1):35–45.PubMedCrossRef D’Souza DC, Cortes-Briones J, Creatura G, Bluez G, Thurnauer H, Deaso E, et al. Efficacy and safety of a fatty acid amide hydrolase inhibitor (PF-04457845) in the treatment of cannabis withdrawal and dependence in men: a double-blind, placebo-controlled, parallel group, phase 2a single-site randomised controlled trial. Lancet Psychiatry. 2019;6(1):35–45.PubMedCrossRef
198.
go back to reference Desai N, Burns L, Gong Y, Zhi K, Kumar A, Summers N, et al. An update on drug-drug interactions between antiretroviral therapies and drugs of abuse in HIV systems. Expert Opin Drug Metab Toxicol. 2020;16(11):1005–18.PubMedPubMedCentralCrossRef Desai N, Burns L, Gong Y, Zhi K, Kumar A, Summers N, et al. An update on drug-drug interactions between antiretroviral therapies and drugs of abuse in HIV systems. Expert Opin Drug Metab Toxicol. 2020;16(11):1005–18.PubMedPubMedCentralCrossRef
199.
go back to reference Tucker JS, Burnam MA, Sherbourne CD, Kung FY, Gifford AL. Substance use and mental health correlates of nonadherence to antiretroviral medications in a sample of patients with human immunodeficiency virus infection. Am J Med. 2003;114(7):573–80.PubMedCrossRef Tucker JS, Burnam MA, Sherbourne CD, Kung FY, Gifford AL. Substance use and mental health correlates of nonadherence to antiretroviral medications in a sample of patients with human immunodeficiency virus infection. Am J Med. 2003;114(7):573–80.PubMedCrossRef
200.
go back to reference Hicks PL, Mulvey KP, Chander G, Fleishman JA, Josephs JS, Korthuis PT, et al. The impact of illicit drug use and substance abuse treatment on adherence to HAART. AIDS Care. 2007;19(9):1134–40.PubMedPubMedCentralCrossRef Hicks PL, Mulvey KP, Chander G, Fleishman JA, Josephs JS, Korthuis PT, et al. The impact of illicit drug use and substance abuse treatment on adherence to HAART. AIDS Care. 2007;19(9):1134–40.PubMedPubMedCentralCrossRef
201.
go back to reference Bonn-Miller MO, Oser ML, Bucossi MM, Trafton JA. Cannabis use and HIV antiretroviral therapy adherence and HIV-related symptoms. J Behav Med. 2014;37(1):1–10.PubMedCrossRef Bonn-Miller MO, Oser ML, Bucossi MM, Trafton JA. Cannabis use and HIV antiretroviral therapy adherence and HIV-related symptoms. J Behav Med. 2014;37(1):1–10.PubMedCrossRef
202.
go back to reference Zhang Y, Wilson TE, Adedimeji A, Merenstein D, Milam J, Cohen J, et al. The impact of substance use on adherence to antiretroviral therapy among HIV-infected women in the United States. AIDS Behav. 2018;22(3):896–908.PubMedPubMedCentralCrossRef Zhang Y, Wilson TE, Adedimeji A, Merenstein D, Milam J, Cohen J, et al. The impact of substance use on adherence to antiretroviral therapy among HIV-infected women in the United States. AIDS Behav. 2018;22(3):896–908.PubMedPubMedCentralCrossRef
203.
go back to reference Manuzak JA, Granche J, Tassiopoulos K, Rower JE, Knox JR, Williams DW, et al. Cannabis use is associated with decreased antiretroviral therapy adherence among older adults with HIV. Open Forum Infect Dis. 2023;10(1):ofac699.PubMedPubMedCentralCrossRef Manuzak JA, Granche J, Tassiopoulos K, Rower JE, Knox JR, Williams DW, et al. Cannabis use is associated with decreased antiretroviral therapy adherence among older adults with HIV. Open Forum Infect Dis. 2023;10(1):ofac699.PubMedPubMedCentralCrossRef
204.
go back to reference Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.PubMedPubMedCentralCrossRef Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.PubMedPubMedCentralCrossRef
205.
go back to reference Jordan Walter T, Pocuca N, Young JW, Geyer MA, Minassian A, Perry W. The relationship between cannabis use and cognition in people with bipolar disorder: a systematic scoping review. Psychiatry Res. 2021;297:113695.PubMedCrossRef Jordan Walter T, Pocuca N, Young JW, Geyer MA, Minassian A, Perry W. The relationship between cannabis use and cognition in people with bipolar disorder: a systematic scoping review. Psychiatry Res. 2021;297:113695.PubMedCrossRef
206.
go back to reference Pocuca N, Walter TJ, Minassian A, Young JW, Geyer MA, Perry W. The effects of cannabis use on cognitive function in healthy aging: a systematic scoping review. Arch Clin Neuropsychol. 2021;36(5):673–85.PubMedCrossRef Pocuca N, Walter TJ, Minassian A, Young JW, Geyer MA, Perry W. The effects of cannabis use on cognitive function in healthy aging: a systematic scoping review. Arch Clin Neuropsychol. 2021;36(5):673–85.PubMedCrossRef
Metadata
Title
The Impact of Cannabis Use on Cognition in People with HIV: Evidence of Function-Dependent Effects and Mechanisms from Clinical and Preclinical Studies
Authors
Samantha M. Ayoub
Breanna M. Holloway
Alannah H. Miranda
Benjamin Z. Roberts
Jared W. Young
Arpi Minassian
Ronald J. Ellis
Publication date
11-04-2024
Publisher
Springer US
Published in
Current HIV/AIDS Reports
Print ISSN: 1548-3568
Electronic ISSN: 1548-3576
DOI
https://doi.org/10.1007/s11904-024-00698-w
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.