1
|
Butelman ER, Goldstein RZ, Nwaneshiudu CA, Girdhar K, Roussos P, Russo SJ, Alia-Klein N. Neuroimmune Mechanisms of Opioid Use Disorder and Recovery: Translatability to Human Studies, and Future Research Directions. Neuroscience 2023; 528:102-116. [PMID: 37562536 PMCID: PMC10720374 DOI: 10.1016/j.neuroscience.2023.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
Opioid use disorder (OUD) is a major current cause of morbidity and mortality. Long-term exposure to short-acting opioids (MOP-r agonists such as heroin or fentanyl) results in complex pathophysiological changes to neuroimmune and neuroinflammatory functions, affected in part by peripheral mechanisms (e.g., cytokines in blood), and by neuroendocrine systems such as the hypothalamic-pituitary-adrenal (HPA) stress axis. There are important findings from preclinical models, but their role in the trajectory and outcomes of OUD in humans is not well understood. The goal of this narrative review is to examine available data on immune and inflammatory functions in persons with OUD, and to identify major areas for future research. Peripheral blood biomarker studies revealed a pro-inflammatory state in persons with OUD in withdrawal or early abstinence, consistent with available postmortem brain studies (which show glial activation) and diffusion tensor imaging studies (indicating white matter disruptions), with gradual abstinence-associated recovery. The mechanistic roles of these neuroimmune and neuroinflammatory changes in the trajectory of OUD (including recovery and medication management) cannot be examined practically with postmortem data. Collection of longitudinal data in larger-scale human cohorts would allow examination of these mechanisms associated with OUD stage and progression. Given the heterogeneity in presentation of OUD, a precision medicine approach integrating multi-omic peripheral biomarkers and comprehensive phenotyping, including neuroimaging, can be beneficial in risk stratification, and individually optimized selection of interventions for individuals who will benefit, and assessments under refractory therapy.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Rita Z Goldstein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chinwe A Nwaneshiudu
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA, Medical Center, Bronx, NY, USA
| | - Scott J Russo
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nelly Alia-Klein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Bálint ER, Fűr G, Kui B, Balla Z, Kormányos ES, Orján EM, Tóth B, Horváth G, Szűcs E, Benyhe S, Ducza E, Pallagi P, Maléth J, Venglovecz V, Hegyi P, Kiss L, Rakonczay Z. Fentanyl but Not Morphine or Buprenorphine Improves the Severity of Necrotizing Acute Pancreatitis in Rats. Int J Mol Sci 2022; 23:1192. [PMID: 35163111 PMCID: PMC8835441 DOI: 10.3390/ijms23031192] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Opioids are widely used for the pain management of acute pancreatitis (AP), but their impact on disease progression is unclear. Therefore, our aim was to study the effects of clinically relevant opioids on the severity of experimental AP. Various doses of fentanyl, morphine, or buprenorphine were administered as pre- and/or post-treatments in rats. Necrotizing AP was induced by the intraperitoneal injection of L-ornithine-HCl or intra-ductal injection of Na-taurocholate, while intraperitoneal caerulein administration caused edematous AP. Disease severity was determined by laboratory and histological measurements. Mu opioid receptor (MOR) expression and function was assessed in control and AP animals. MOR was expressed in both the pancreas and brain. The pancreatic expression and function of MOR were reduced in AP. Fentanyl post-treatment reduced necrotizing AP severity, whereas pre-treatment exacerbated it. Fentanyl did not affect the outcome of edematous AP. Morphine decreased vacuolization in edematous AP, while buprenorphine pre-treatment increased pancreatic edema during AP. The overall effects of morphine on disease severity were negligible. In conclusion, the type, dosing, administration route, and timing of opioid treatment can influence the effects of opioids on AP severity. Fentanyl post-treatment proved to be beneficial in AP. Clinical studies are needed to determine which opioids are best in AP.
Collapse
Affiliation(s)
- Emese Réka Bálint
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Gabriella Fűr
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Balázs Kui
- Department of Medicine, University of Szeged, 6725 Szeged, Hungary; (B.K.); (P.P.); (J.M.); (P.H.)
| | - Zsolt Balla
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Eszter Sára Kormányos
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Erik Márk Orján
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Brigitta Tóth
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Gyöngyi Horváth
- Department of Physiology, University of Szeged, 6725 Szeged, Hungary;
| | - Edina Szűcs
- Institute of Biochemistry, Biological Research Center, 6726 Szeged, Hungary; (E.S.); (S.B.)
| | - Sándor Benyhe
- Institute of Biochemistry, Biological Research Center, 6726 Szeged, Hungary; (E.S.); (S.B.)
| | - Eszter Ducza
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6725 Szeged, Hungary;
| | - Petra Pallagi
- Department of Medicine, University of Szeged, 6725 Szeged, Hungary; (B.K.); (P.P.); (J.M.); (P.H.)
| | - József Maléth
- Department of Medicine, University of Szeged, 6725 Szeged, Hungary; (B.K.); (P.P.); (J.M.); (P.H.)
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6725 Szeged, Hungary;
| | - Péter Hegyi
- Department of Medicine, University of Szeged, 6725 Szeged, Hungary; (B.K.); (P.P.); (J.M.); (P.H.)
- Institute for Translational Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Lóránd Kiss
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| | - Zoltán Rakonczay
- Department of Pathophysiology, University of Szeged, 6725 Szeged, Hungary; (E.R.B.); (G.F.); (Z.B.); (E.S.K.); (E.M.O.); (B.T.)
| |
Collapse
|
3
|
Gornalusse GG, Vojtech LN, Levy CN, Hughes SM, Kim Y, Valdez R, Pandey U, Ochsenbauer C, Astronomo R, McElrath J, Hladik F. Buprenorphine Increases HIV-1 Infection In Vitro but Does Not Reactivate HIV-1 from Latency. Viruses 2021; 13:1472. [PMID: 34452338 PMCID: PMC8402857 DOI: 10.3390/v13081472] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/28/2021] [Accepted: 07/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND medication-assisted treatment (MAT) with buprenorphine is now widely prescribed to treat addiction to heroin and other illicit opioids. There is some evidence that illicit opioids enhance HIV-1 replication and accelerate AIDS pathogenesis, but the effect of buprenorphine is unknown. METHODS we obtained peripheral blood mononuclear cells (PBMCs) from healthy volunteers and cultured them in the presence of morphine, buprenorphine, or methadone. We infected the cells with a replication-competent CCR5-tropic HIV-1 reporter virus encoding a secreted nanoluciferase gene, and measured infection by luciferase activity in the supernatants over time. We also surveyed opioid receptor expression in PBMC, genital epithelial cells and other leukocytes by qPCR and western blotting. Reactivation from latency was assessed in J-Lat 11.1 and U1 cell lines. RESULTS we did not detect expression of classical opioid receptors in leukocytes, but did find nociception/orphanin FQ receptor (NOP) expression in blood and vaginal lymphocytes as well as genital epithelial cells. In PBMCs, we found that at physiological doses, morphine, and methadone had a variable or no effect on HIV infection, but buprenorphine treatment significantly increased HIV-1 infectivity (median: 8.797-fold increase with 20 nM buprenorphine, eight experiments, range: 3.570-691.9, p = 0.0078). Using latently infected cell lines, we did not detect reactivation of latent HIV following treatment with any of the opioid drugs. CONCLUSIONS our results suggest that buprenorphine, in contrast to morphine or methadone, increases the in vitro susceptibility of leukocytes to HIV-1 infection but has no effect on in vitro HIV reactivation. These findings contribute to our understanding how opioids, including those used for MAT, affect HIV infection and reactivation, and can help to inform the choice of MAT for people living with HIV or who are at risk of HIV infection.
Collapse
Affiliation(s)
- Germán Gustavo Gornalusse
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Lucia N. Vojtech
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Claire N. Levy
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Sean M. Hughes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Yeseul Kim
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Rogelio Valdez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
| | - Urvashi Pandey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Christina Ochsenbauer
- School of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Rena Astronomo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
| | - Julie McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Pathobiology, Global Health and Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Florian Hladik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (G.G.G.); (L.N.V.); (C.N.L.); (S.M.H.); (Y.K.); (R.V.); (U.P.); (R.A.); (J.M.)
- Departments of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
4
|
Biondi BE, Mohanty S, Wyk BV, Montgomery RR, Shaw AC, Springer SA. Design and implementation of a prospective cohort study of persons living with and without HIV infection who are initiating medication treatment for opioid use disorder. Contemp Clin Trials Commun 2021; 21:100704. [PMID: 33490708 PMCID: PMC7807244 DOI: 10.1016/j.conctc.2021.100704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/15/2020] [Accepted: 01/01/2021] [Indexed: 02/02/2023] Open
Abstract
Background Opioid use disorder (OUD) negatively impacts the HIV continuum of care for persons living with HIV. Medication treatment for OUD (MOUD) may have differential biological effects in individuals with HIV and OUD. To address the question of modulation of immune responses by MOUDs, we describe state of the art systems biology approaches to carry out the first prospective, longitudinal study of persons with and without HIV infection with OUD initiating MOUD. Methods A prospective cohort study of persons with DSM-5 diagnosed OUD who are living with and without HIV infection and initiating treatment with methadone or buprenorphine is underway to assess biological effects of these medications on immunobiological outcomes. Results We describe the recruitment, laboratory, and statistical methods of this study as well as the protocol details. Of those screened for enrollment into the study, 468 (36%) were eligible and 135 were enrolled thus far. Retention through month 6 has been high at 80%. Conclusions This study will use state of the art systems biology approaches to carry out the first prospective, longitudinal studies of persons living with and without HIV with DSM-5 OUD initiating treatment with MOUD.
Collapse
Affiliation(s)
- Breanne E Biondi
- Department of Internal Medicine, Section of Infectious Diseases, AIDS Program, Yale School of Medicine, New Haven, CT, USA
| | - Subhasis Mohanty
- Department of Internal Medicine, Section of Infectious Diseases, AIDS Program, Yale School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Section of Geriatrics, Yale School of Medicine, New Haven, CT, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Section of Rheumatology, Yale School of Medicine, New Haven, CT, USA
| | - Albert C Shaw
- Department of Internal Medicine, Section of Infectious Diseases, AIDS Program, Yale School of Medicine, New Haven, CT, USA
| | - Sandra A Springer
- Department of Internal Medicine, Section of Infectious Diseases, AIDS Program, Yale School of Medicine, New Haven, CT, USA.,Center for Interdisciplinary Research on AIDS, Yale University School of Public Health, New Haven, CT, USA
| |
Collapse
|
5
|
Harnessing cancer immunotherapy during the unexploited immediate perioperative period. Nat Rev Clin Oncol 2020; 17:313-326. [PMID: 32066936 DOI: 10.1038/s41571-019-0319-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
The immediate perioperative period (days before and after surgery) is hypothesized to be crucial in determining long-term cancer outcomes: during this short period, numerous factors, including excess stress and inflammatory responses, tumour-cell shedding and pro-angiogenic and/or growth factors, might facilitate the progression of pre-existing micrometastases and the initiation of new metastases, while simultaneously jeopardizing immune control over residual malignant cells. Thus, application of anticancer immunotherapy during this critical time frame could potentially improve patient outcomes. Nevertheless, this strategy has rarely been implemented to date. In this Perspective, we discuss apparent contraindications for the perioperative use of cancer immunotherapy, suggest safe immunotherapeutic and other anti-metastatic approaches during this important time frame and specify desired characteristics of such interventions. These characteristics include a rapid onset of immune activation, avoidance of tumour-promoting effects, no or minimal increase in surgical risk, resilience to stress-related factors and minimal induction of stress responses. Pharmacological control of excess perioperative stress-inflammatory responses has been shown to be clinically feasible and could potentially be combined with immune stimulation to overcome the direct pro-metastatic effects of surgery, prevent immune suppression and enhance immunostimulatory responses. Accordingly, we believe that certain types of immunotherapy, together with interventions to abrogate stress-inflammatory responses, should be evaluated in conjunction with surgery and, for maximal effectiveness, could be initiated before administration of adjuvant therapies. Such strategies might improve the overall success of cancer treatment.
Collapse
|
6
|
Barlass U, Dutta R, Cheema H, George J, Sareen A, Dixit A, Yuan Z, Giri B, Meng J, Banerjee S, Banerjee S, Dudeja V, Dawra RK, Roy S, Saluja AK. Morphine worsens the severity and prevents pancreatic regeneration in mouse models of acute pancreatitis. Gut 2018. [PMID: 28642332 DOI: 10.1136/gutjnl-2017-313717] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Opioids such as morphine are widely used for the management of pain associated with acute pancreatitis. Interestingly, opioids are also known to affect the immune system and modulate inflammatory pathways in non-pancreatic diseases. However, the impact of morphine on the progression of acute pancreatitis has never been evaluated. In the current study, we evaluated the impact of morphine on the progression and severity of acute pancreatitis. METHODS Effect of morphine treatment on acute pancreatitis in caerulein, L-arginine and ethanol-palmitoleic acid models was evaluated after induction of the disease. Inflammatory response, gut permeability and bacterial translocation were compared. Experiments were repeated in mu (µ) opioid receptor knockout mice (MORKO) and in wild-type mice in the presence of opioid receptor antagonist naltrexone to evaluate the role of µ-opioid receptors in morphine's effect on acute pancreatitis. Effect of morphine treatment on pathways activated during pancreatic regeneration like sonic Hedgehog and activation of embryonic transcription factors like pdx-1 and ptf-1 were measured by immunofluorescence and quantitative PCR. RESULTS Histological data show that treatment with morphine after induction of acute pancreatitis exacerbates the disease with increased pancreatic neutrophilic infiltration and necrosis in all three models of acute pancreatitis. Morphine also exacerbated acute pancreatitis-induced gut permeabilisation and bacteraemia. These effects were antagonised in the MORKO mice or in the presence of naltrexone suggesting that morphine's effect on severity of acute pancreatitis are mediated through the µ-opioid receptors. Morphine treatment delayed macrophage infiltration, sonic Hedgehog pathway activation and expression of pdx-1 and ptf-1. CONCLUSION Morphine treatment worsens the severity of acute pancreatitis and delays resolution and regeneration. Considering our results, the safety of morphine for analgesia during acute pancreatitis should be re-evaluated in future human studies.
Collapse
Affiliation(s)
- Usman Barlass
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Raini Dutta
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Hassam Cheema
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - John George
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Archana Sareen
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Ajay Dixit
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Zuobiao Yuan
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Bhuwan Giri
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Jingjing Meng
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Santanu Banerjee
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Sulagna Banerjee
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Vikas Dudeja
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Rajinder K Dawra
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Sabita Roy
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| | - Ashok K Saluja
- Sylvester Comprehensive Cancer Center Department of Surgery, University of Miami, Miami, Florida, USA
| |
Collapse
|
7
|
Jaureguiberry-Bravo M, Wilson R, Carvallo L, Berman JW. Opioids and Opioid Maintenance Therapies: Their Impact on Monocyte-Mediated HIV Neuropathogenesis. Curr HIV Res 2017; 14:417-430. [PMID: 27009099 DOI: 10.2174/1570162x14666160324124132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/26/2015] [Accepted: 11/10/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND HIV-1 enters the CNS within two weeks after peripheral infection and results in chronic neuroinflammation that leads to HIV associated neurocognitive disorders (HAND) in more than 50% of infected people. HIV enters the CNS by transmigration of infected monocytes across the blood brain barrier. Intravenous drug abuse is a major risk factor for HIV-1 infection, and opioids have been shown to alter the progression and severity of HAND. Methadone and buprenorphine are opioid derivates that are used as opioid maintenance therapies. They are commonly used to treat opioid dependency in HIV infected substance abusers, but their effects on monocyte migration relevant to the development of cognitive impairment are not well characterized. CONCLUSION Here, we will discuss the effects of opioids and opioid maintenance therapies on the inflammatory functions of monocytes and macrophages that are related to the development of neuroinflammation in the context of HIV infection.
Collapse
Affiliation(s)
| | | | | | - Joan W Berman
- Department, of Pathology and Microbiology and Immunology, F727, Albert Einstein College of Medicine, 1300 Morris Park Ave. Bronx, NY, 10461, USA.
| |
Collapse
|
8
|
Chen YH, Wu KL, Tsai MT, Chien WH, Chen ML, Wang Y. Methadone enhances human influenza A virus replication. Addict Biol 2017; 22:257-271. [PMID: 26350582 DOI: 10.1111/adb.12305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/13/2015] [Accepted: 08/14/2015] [Indexed: 12/24/2022]
Abstract
Growing evidence has indicated that opioids enhance replication of human immunodeficiency virus and hepatitis C virus in target cells. However, it is unknown whether opioids can enhance replication of other clinically important viral pathogens. In this study, the interaction of opioid agonists and human influenza A/WSN/33 (H1N1) virus was examined in human lung epithelial A549 cells. Cells were exposed to morphine, methadone or buprenorphine followed by human H1N1 viral infection. Exposure to methadone differentially enhanced viral propagation, consistent with an increase in virus adsorption, susceptibility to virus infection and viral protein synthesis. In contrast, morphine or buprenorphine did not alter H1N1 replication. Because A549 cells do not express opioid receptors, methadone-enhanced H1N1 replication in human lung cells may not be mediated through these receptors. The interaction of methadone and H1N1 virus was also examined in adult mice. Treatment with methadone significantly increased H1N1 viral replication in lungs. Our data suggest that use of methadone facilitates influenza A viral infection in lungs and might raise concerns regarding the possible consequence of an increased risk of serious influenza A virus infection in people who receive treatment in methadone maintenance programs.
Collapse
Affiliation(s)
- Yun-Hsiang Chen
- Center for Neuropsychiatric Research; National Health Research Institutes; Taiwan
- Department of Life Science; Fu Jen Catholic University; Taiwan
| | - Kuang-Lun Wu
- Center for Neuropsychiatric Research; National Health Research Institutes; Taiwan
| | - Ming-Ta Tsai
- Center for Neuropsychiatric Research; National Health Research Institutes; Taiwan
| | - Wei-Hsien Chien
- Department of Occupational Therapy; Fu Jen Catholic University; Taiwan
| | - Mao-Liang Chen
- Department of Research; Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation; Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research; National Health Research Institutes; Taiwan
| |
Collapse
|
9
|
Wang X, Ma TC, Li JL, Zhou Y, Geller EB, Adler MW, Peng JS, Zhou W, Zhou DJ, Ho WZ. Heroin inhibits HIV-restriction miRNAs and enhances HIV infection of macrophages. Front Microbiol 2015; 6:1230. [PMID: 26583016 PMCID: PMC4632020 DOI: 10.3389/fmicb.2015.01230] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/20/2015] [Indexed: 12/16/2022] Open
Abstract
Although opioids have been extensively studied for their impact on the immune system, limited information is available about the specific actions of opioids on intracellular antiviral innate immunity against HIV infection. Thus, we investigated whether heroin, one of the most abused drugs, inhibits the expression of intracellular HIV restriction microRNA (miRNA) and facilitates HIV replication in macrophages. Heroin treatment of macrophages enhanced HIV replication, which was associated with the downregulation of several HIV restriction miRNAs. These heroin-mediated actions on the miRNAs and HIV could be antagonized by naltrexone, an opioid receptor antagonist. Furthermore, the in vitro negative impact of heroin on HIV-associated miRNAs was confirmed by the in vivo observation that heroin addicts had significantly lower levels of macrophage-derived HIV restriction miRNAs than those in the control subjects. These in vitro and in vivo findings indicate that heroin use compromises intracellular anti-HIV innate immunity, providing a favorable microenvironment for HIV survival in the target cells.
Collapse
Affiliation(s)
- Xu Wang
- School of Basic Medical Sciences, Wuhan University Wuhan, China ; Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA
| | - Tong-Cui Ma
- School of Basic Medical Sciences, Wuhan University Wuhan, China ; Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA ; The Center for Animal Experiment/ABSL-III Laboratory, Wuhan University Wuhan, China
| | - Jie-Liang Li
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA
| | - Yu Zhou
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA
| | - Ellen B Geller
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA
| | - Martin W Adler
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA
| | - Jin-Song Peng
- Wuhan Center for Disease Prevention and Control Wuhan, China
| | - Wang Zhou
- Wuhan Center for Disease Prevention and Control Wuhan, China
| | - Dun-Jin Zhou
- Wuhan Center for Disease Prevention and Control Wuhan, China
| | - Wen-Zhe Ho
- School of Basic Medical Sciences, Wuhan University Wuhan, China ; Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia PA, USA ; The Center for Animal Experiment/ABSL-III Laboratory, Wuhan University Wuhan, China
| |
Collapse
|
10
|
Abstract
OBJECTIVES There are few studies with conflicting results on the effects of in vivo administration of opioids on immune function. The aim of this study was to evaluate the serum levels of interferon (IFN)-γ, interleukin (IL)-4, IL-10, IL-17, and hs-C-reactive protein (hs-CRP) in opium smokers. METHODS The study was conducted between 44 male opium addicts and 44 controls aged 20 to 40 years. The control group was healthy individuals with no lifetime history of substance abuse. All the opium abusers were selected from those who had a history of use of opium, as a regular habit, at least for 1 year, with a daily opium dosage of not less than 2 g. Addicts known to abuse alcohol or other drugs were excluded. Serum samples were collected from all participants and tested for the cytokine and hs-CRP levels by ELISA (enzyme-linked immunosorbent assay) method. Statistical analysis was performed using the Student t test. RESULTS The mean serum levels of IFN-γ, IL-10, and IL-17 in the opium addicts were significantly higher than those observed in the control group. The mean concentration of serum IL-4 in opium addicts did not differ from that in the control group. Systemic IL-10 levels correlated positively and significantly with CRP in opium addicts. CONCLUSIONS Long-term, daily use of opium is associated with higher Th1 (IFN-γ), Tr1 (IL-10), and Th17 (IL-17) cytokines concentration in serum. Interferon-γ and IL-17 are involved in inducing and mediating proinflammatory responses. Our data suggest that an immunoregulatory response is occurring with the upregulation of IL-10.
Collapse
|
11
|
Börner C, Lanciotti S, Koch T, Höllt V, Kraus J. μ opioid receptor agonist-selective regulation of interleukin-4 in T lymphocytes. J Neuroimmunol 2013; 263:35-42. [PMID: 23965172 DOI: 10.1016/j.jneuroim.2013.07.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 07/08/2013] [Accepted: 07/18/2013] [Indexed: 12/13/2022]
Abstract
Opioids are irreplaceable for the treatment of severe pain. However, opioid-induced immunomodulation affects therapies. Here we report that treatment of human T lymphocytes with the opioids fentanyl, methadone, loperamide and beta-endorphin resulted in a strong induction of the cytokine interleukin-4. In contrast, morphine and buprenorphine induced markedly and significantly lower levels of interleukin-4 mRNA and protein. These findings suggest agonist-biased μ opioid receptor signaling in T cells. In the future, better knowledge about agonist-specific immunomodulatory effects of opioids offers the possibility to select drugs for a therapy with more favorable and/or less detrimental side effects in immune cells.
Collapse
Affiliation(s)
- Christine Börner
- Department of Pharmacology and Toxicology, University of Magdeburg, 44 Leipzigerstrasse, 39120 Magdeburg, Germany
| | | | | | | | | |
Collapse
|
12
|
Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids 2011; 45:9-24. [PMID: 22170499 DOI: 10.1007/s00726-011-1163-0] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 11/15/2011] [Indexed: 12/13/2022]
Abstract
Endogenous opioids are synthesized in vivo to modulate pain mechanisms and inflammatory pathways. Endogenous and exogenous opioids mediate analgesia in response to painful stimuli by binding to opioid receptors on neuronal cells. However, wide distribution of opioid receptors on tissues and organ systems outside the CNS, such as the cells of the immune system, indicate that opioids are capable of exerting additional effects in the periphery, such as immunomodulation. The increased prevalence of infections in opioid abuser-based epidemiological studies further highlights the immunosuppressive effects of opioids. In spite of their many debilitating side effects, prescription opioids remain a gold standard for treatment of chronic pain. Therefore, given the prevalence of opioid use and abuse, opioid-mediated immune suppression presents a serious concern in our society today. It is imperative to understand the mechanisms by which exogenous opioids modulate immune processes. In this review, we will discuss the role of opioid receptors and their ligands in mediating immune-suppressive functions. We will summarize recent studies on direct and indirect opioid modulation of the cells of the immune system, as well as the role of opioids in exacerbation of certain disease states.
Collapse
|
13
|
Roy S, Ninkovic J, Banerjee S, Charboneau RG, Das S, Dutta R, Kirchner VA, Koodie L, Ma J, Meng J, Barke RA. Opioid drug abuse and modulation of immune function: consequences in the susceptibility to opportunistic infections. J Neuroimmune Pharmacol 2011; 6:442-65. [PMID: 21789507 PMCID: PMC3601186 DOI: 10.1007/s11481-011-9292-5] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 06/27/2011] [Indexed: 12/13/2022]
Abstract
Infection rate among intravenous drug users (IDU) is higher than the general public, and is the major cause of morbidity and hospitalization in the IDU population. Epidemiologic studies provide data on increased prevalence of opportunistic bacterial infections such as TB and pneumonia, and viral infections such as HIV-1 and hepatitis in the IDU population. An important component in the intravenous drug abuse population and in patients receiving medically indicated chronic opioid treatment is opioid withdrawal. Data on bacterial virulence in the context of opioid withdrawal suggest that mice undergoing withdrawal had shortened survival and increased bacterial load in response to Salmonella infection. As the body of evidence in support of opioid dependency and its immunosuppressive effects is growing, it is imperative to understand the mechanisms by which opioids exert these effects and identify the populations at risk that would benefit the most from the interventions to counteract opioid immunosuppressive effects. Thus, it is important to refine the existing animal model to closely match human conditions and to cross-validate these findings through carefully controlled human studies. Better understanding of the mechanisms will facilitate the search for new therapeutic modalities to counteract adverse effects including increased infection rates. This review will summarize the effects of morphine on innate and adaptive immunity, identify the role of the mu opioid receptor in these functions and the signal transduction activated in the process. The role of opioid withdrawal in immunosuppression and the clinical relevance of these findings will also be discussed.
Collapse
Affiliation(s)
- Sabita Roy
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang X, Zhang T, Ho WZ. Opioids and HIV/HCV infection. J Neuroimmune Pharmacol 2011; 6:477-89. [PMID: 21755286 PMCID: PMC3937260 DOI: 10.1007/s11481-011-9296-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/04/2011] [Indexed: 12/20/2022]
Abstract
Since human immunodeficiency virus (HIV) and hepatitis C virus (HCV) share the same modes of transmission and common risk factors for infection, co-infections with HIV and HCV are frequently found in injection drug users (IDUs). IDUs represent one of the largest reservoirs of HIV as well as HCV in the United States. These two pathogens are also likely to be responsible for the highest infectious disease morbidity and mortality rates among IDUs. IDUs frequently involve the abuse of heroin, the most common abused opiate. Opiates have been suggested to have a cofactor role in the immunopathogenesis of HIV disease, as they have the potential to compromise host immune responses and enhances microbial infections. Although in vitro studies have yielded relatively agreeable data that morphine, the active metabolite of heroin, exacerbate HIV infection/replication, epidemiologic studies as well as in vivo non-human primate investigations on the impact of opiate abuse on HIV disease progression have yielded the conflicting data. Given immunomodulation and immunocompromising effect as well as demonstrated impact to enhance HIV replication in vitro, it is reasonable to believe that opiate abuse is a facilitator in HIV and/or HCV disease progression. However, much remain to be learned about the mechanisms of opiate-mediated broad influence on host immunity and viral expression. Thus, more extensive studies are needed in order to determine the effects of different conditions of opiate abuse and to define the understanding of the role of opiate in modulating HIV and/or HCV disease progression.
Collapse
Affiliation(s)
- Xu Wang
- Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, Hubei 430071, People’s Republic of China. Department of Pathology and Laboratory Medicine, Medical Education and Research Building, Room 1082A, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Ting Zhang
- Division of Infectious Diseases, The Children’s Hospital of Fudan University, Shanghai 200032, People’s Republic of China
| | - Wen-Zhe Ho
- Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan, Hubei 430071, People’s Republic of China. Department of Pathology and Laboratory Medicine, Medical Education and Research Building, Room 1052, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
15
|
Drugs of abuse and HIV infection/replication: implications for mother-fetus transmission. Life Sci 2010; 88:972-9. [PMID: 21056582 DOI: 10.1016/j.lfs.2010.10.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 10/11/2010] [Accepted: 10/27/2010] [Indexed: 12/14/2022]
Abstract
Human immunodeficiency virus (HIV) infection and progression of acquired immunodeficiency syndrome (AIDS) can be modulated by a number of cofactors, including drugs of abuse. Opioids, cocaine, cannabinoids, methamphetamine (METH), alcohol, and other substances of abuse have been implicated as risk factors for HIV infection, as they all have the potential to compromise host immunity and facilitate viral replication. Although epidemiologic evidence regarding the impact of drugs of abuse on HIV disease progression is mixed, in vitro studies as well as studies using in vivo animal models have indicated that drugs of abuse have the ability to enhance HIV infection/replication. Drugs of abuse may also be a risk factor for perinatal transmission of HIV. Because high levels of viral load in maternal blood are associated with increased risk of HIV vertical transmission, it is likely that drugs of abuse play an important role in promoting mother-fetus transmission. Furthermore, because the neonatal immune system differs qualitatively from the adult system, it is possible that maternal exposure to drugs of abuse would exacerbate neonatal immunity defects, facilitating HIV infection of neonate immune cells and promoting HIV vertical transmission. The availability and use of antiretroviral therapy for women infected with HIV increase, there is an increasing interest in determining the impact of drug abuse on efficacy of AIDS Clinical Trials Group (ACTG)-standardized treatment regimens for woman infected with HIV in the context of HIV vertical transmission.
Collapse
|
16
|
Easten KH, Harry RA, Purcell WM, McLeod JD. Nociceptin-induced modulation of human T cell function. Peptides 2009; 30:926-34. [PMID: 19428771 DOI: 10.1016/j.peptides.2009.01.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 01/21/2009] [Accepted: 01/22/2009] [Indexed: 01/21/2023]
Abstract
There is an accumulating evidence for the immunoregulatory role of the neuropeptide, nociceptin/orphanin FQ (N/OFQ) however its role on T cell function requires elucidation. This study has demonstrated an inhibitory role for N/OFQ on SEB-activated T cell function. N/OFQ decreases T cell proliferation, which is abrogated when the costimulatory receptors CD80 and CD86 are blocked. In addition, evidence suggests that the immunoregulatory cytokines TGF-beta, IFN-gamma and nitric oxide (NO) are involved in the N/OFQ effect. N/OFQ also, through involvement of IFN and NO, induces the expression of the immunosuppressive modulator indoleamine 2,3-dioxygenase (IDO), suggesting a central role for IDO in the N/OFQ effect on T cell proliferation. The data presented in this report indicate a multi-faceted mechanism of action used by N/OFQ to modulate T cell function.
Collapse
Affiliation(s)
- Kate H Easten
- Faculty of Health and Life Sciences, Centre for Research in Biomedicine, University of the West of England, Coldharbour lane, Frenchay, Bristol, UK
| | | | | | | |
Collapse
|
17
|
Effects of chronic morphine and morphine withdrawal on gene expression in rat peripheral blood mononuclear cells. Neuropharmacology 2008; 55:1347-54. [DOI: 10.1016/j.neuropharm.2008.08.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 08/04/2008] [Accepted: 08/19/2008] [Indexed: 12/30/2022]
|
18
|
Wan Q, Wang X, Wang YJ, Song L, Wang SH, Ho WZ. Morphine suppresses intracellular interferon-alpha expression in neuronal cells. J Neuroimmunol 2008; 199:1-9. [PMID: 18562017 DOI: 10.1016/j.jneuroim.2008.04.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 04/09/2008] [Accepted: 04/14/2008] [Indexed: 11/30/2022]
Abstract
Interferon alpha (IFN-alpha) not only plays a key role in innate host immunity against infections but also is involved in the cellular functions of the central nervous system (CNS). In this study, we examined the impact of morphine on IFN-alpha expression in human neuronal cells (NT2-N). Similar to human immune cells, NT2-N cells also expressed IFN-alpha at both mRNA and protein levels. IFN-alpha expression in NT2-N cells, however, was inhibited by morphine. Naltrexone antagonized the inhibitory effect of morphine on IFN-alpha expression in NT2-N cells. The specific mu opioid receptor antagonist, Cys2, Tyr3, Arg5, Pen7-amide (CTAP), also blocked the morphine action on intracellular IFN-alpha expression. Investigation of the mechanisms involved in the morphine action showed that although morphine had little effect on the expression of key IFN regulatory factors (IRFs), morphine inhibited IFN-alpha promoter activation and suppressed the expression and phosphorylation of signal transducer and activator of transcription 1 (STAT1) in the neuronal cells. These findings provide direct in vitro evidence that opioids may impair neuronal cell-mediated innate protection in the CNS.
Collapse
Affiliation(s)
- Qi Wan
- Division of Allergy and Immunology, Joseph Stokes, Jr. Research Institute at The Children's Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | | | | | | | | | | |
Collapse
|
19
|
Bonnet MP, Beloeil H, Benhamou D, Mazoit JX, Asehnoune K. The μ Opioid Receptor Mediates Morphine-Induced Tumor Necrosis Factor and Interleukin-6 Inhibition in Toll-Like Receptor 2-Stimulated Monocytes. Anesth Analg 2008; 106:1142-9, table of contents. [DOI: 10.1213/ane.0b013e318165de89] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Modulation of immune function by morphine: implications for susceptibility to infection. J Neuroimmune Pharmacol 2007; 1:77-89. [PMID: 18040793 DOI: 10.1007/s11481-005-9009-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
21
|
Qian L, Tan KS, Wei SJ, Wu HM, Xu Z, Wilson B, Lu RB, Hong JS, Flood PM. Microglia-Mediated Neurotoxicity Is Inhibited by Morphine through an Opioid Receptor-Independent Reduction of NADPH Oxidase Activity. THE JOURNAL OF IMMUNOLOGY 2007; 179:1198-209. [PMID: 17617613 DOI: 10.4049/jimmunol.179.2.1198] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies have shown that morphine modulates the function of glia cells through both opioid receptor dependent and independent mechanisms. However, the mechanism by which morphine regulates neuronal disorders through the alteration of microglia activity remains unclear. In this study, using rat primary mesencephalic neuron-glia cultures, we report that both l-morphine and its synthetic stereoenantiomer, d-morphine, an ineffective opioid receptor agonist, significantly reduced LPS- or 1-methyl-4-phenylpyridinium-induced dopaminergic neurotoxicity with similar efficacy, indicating a nonopioid receptor-mediated effect. In addition, using reconstituted neuron and glia cultures, subpicomolar concentrations of morphine were found to be neuroprotective only in the presence of microglia, and significantly inhibited the production of inflammatory mediators from LPS-stimulated microglia cells. Mechanistic studies showed that both l- and d- morphine failed to protect dopaminergic neurons in cultures from NADPH oxidase (PHOX) knockout mice and significantly reduced LPS-induced PHOX cytosolic subunit p47(phox) translocation to the cell membrane by inhibiting ERK phosphorylation. Taken together, our results demonstrate that morphine, even at subpicomolar concentrations, exerts potent anti-inflammatory and neuroprotective effects either through the inhibition of direct microglial activation by LPS or through the inhibition of reactive microgliosis elicited by 1-methyl-4-phenylpyridinium. Furthermore, our study reveals that inhibition of PHOX is a novel site of action for the mu-opioid receptor-independent effect of morphine.
Collapse
Affiliation(s)
- Li Qian
- Comprehensive Center for Inflammatory Disorders, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang CQ, Li Y, Douglas SD, Wang X, Metzger DS, Zhang T, Ho WZ. Morphine withdrawal enhances hepatitis C virus replicon expression. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 167:1333-40. [PMID: 16251417 PMCID: PMC1603791 DOI: 10.1016/s0002-9440(10)61220-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We previously demonstrated that morphine enhances hepatitis C virus (HCV) replication in human hepatic cells. Here we describe the impact of morphine withdrawal (MW), a recurrent event during the course of opioid abuse, on HCV replicon expression in human hepatic cells. MW enhanced both viral RNA and protein expression in HCV replicon cells. Blocking opioid receptors by treatment with naloxone after morphine cessation (precipitated withdrawal, PW) induced greater HCV replicon expression than MW. Investigation of the mechanism responsible for MW- or PW-mediated HCV enhancement showed that both MW and PW inhibited the expression of endogenous interferon-alpha (IFN-alpha) in the hepatic cells. This down-regulation of intracellular IFN-alpha expression was due to the negative impact of MW or PW on IFN-alpha promoter activation and on the expression of IFN regulatory factor 7 (IRF-7), a strong transactivator of the IFN-alpha promoter. In addition, both MW and PW inhibited the anti-HCV ability of recombinant IFN-alpha in the hepatic cells. These in vitro observations support the concept that opioid abuse favors HCV persistence in hepatic cells by suppressing IFN-alpha-mediated intracellular innate immunity and contributes to the development of chronic HCV infection.
Collapse
Affiliation(s)
- Chuan-Qing Wang
- Department of Pediatrics, Division of Allergy and Immunology, The Children's Hospital of Philadelphia, and the Department of Psychiatry, University of Pennsylvania School of Medicine, 34th St. and Civic Center Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Thorpe LE, Frederick M, Pitt J, Cheng I, Watts DH, Buschur S, Green K, Zorrilla C, Landesman SH, Hershow RC. Effect of hard-drug use on CD4 cell percentage, HIV RNA level, and progression to AIDS-defining class C events among HIV-infected women. J Acquir Immune Defic Syndr 2005; 37:1423-30. [PMID: 15483472 DOI: 10.1097/01.qai.0000127354.78706.5d] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In vitro and animal studies suggest that cocaine and heroin increase HIV replication and suppress immune function, whereas epidemiologic studies are inconclusive regarding their effect on HIV infection progression. The authors prospectively examined the association between illicit-drug use and 4 outcome measures (CD4 cell percentage, HIV RNA level, survival to class C diagnosis of HIV infection, and death) in a national cohort of HIV-infected women. Women enrolled between 1989 and 1995 were followed for 5 years and repeatedly interviewed about illicit ("hard")--drug use. Up to 3 periodic urine screens validated self-reported use. Outcomes were compared between hard-drug users (women using cocaine, heroin, methadone, or injecting drugs) and nonusers, adjusting for age, antiretroviral therapy, number of pregnancies, smoking, and baseline CD4 cell percentage. Of 1148 women, 40% reported baseline hard-drug use during pregnancy. In multivariate analyses, hard-drug use was not associated with change in CD4 cell percentage (P = 0.84), HIV RNA level (P = 0.48), or all-cause mortality (relative hazard = 1.10; 95% confidence interval, 0.61-1.98). Hard-drug users did, however, exhibit a higher risk of developing class C diagnoses (relative hazard = 1.65; 95% confidence interval, 1.00-2.72), especially herpes, pulmonary tuberculosis, and recurrent pneumonia. Hard-drug-using women may have a higher risk for nonfatal opportunistic infections.
Collapse
Affiliation(s)
- Lorna E Thorpe
- New York City Department of Health and Mental Hygiene, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Neu N, Leighty R, Adeniyi-Jones S, Diaz C, Handelsman E, Kaufman G, Paul ME, Rich K, Mofenson L, Pitt J. Immune parameters and morbidity in hard drug and human immunodeficiency virus-exposed but uninfected infants. Pediatrics 2004; 113:1260-6. [PMID: 15121939 DOI: 10.1542/peds.113.5.1260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To examine the association of maternal hard drug use (injection drugs, cocaine, and opiates) on lymphocyte subsets and clinical morbidity in uninfected infants who are born to human immunodeficiency virus-infected mothers who were enrolled in the Women and Infants Transmission Study (1990-2000). METHODS Maternal hard drug use was identified by self-report and/or urine toxicology. Infant evaluations occurred at birth and at 1, 2, 4, 6, 9, 12, 18, and 24 months of age. RESULTS A total of 401 (28%) of the 1436 uninfected infants were born to drug-using mothers. Maternal CD4 lymphocyte percentage and RNA at delivery were not significantly different between drug users and nonusers. Infants who were born to drug-using mothers had lower mean gestational age (37.8 vs 38.5 weeks) and birth weight (2.9 vs 3.1 kg). Infants with intrauterine drug exposure had lower CD4 lymphocyte percentage over the first 4 months of life after adjusting for covariates and higher natural killer lymphocyte percentage. When the analysis was stratified by time period of entry, the incidence of clinical events was not different between infants who were born to drug users versus nonusers. CONCLUSION Maternal hard drug use is associated with immunologic changes in infants early in life, although these changes did not seem to be associated with increased risk of infections.
Collapse
Affiliation(s)
- Natalie Neu
- Columbia University, New York, New York 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang J, Barke RA, Charboneau R, Loh HH, Roy S. Morphine negatively regulates interferon-gamma promoter activity in activated murine T cells through two distinct cyclic AMP-dependent pathways. J Biol Chem 2003; 278:37622-31. [PMID: 12842891 DOI: 10.1074/jbc.m301224200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To explore the mechanism by which morphine promotes the incidence of HIV infection, we evaluated the regulatory role of morphine on the interferon-gamma (IFN-gamma) promoter in activated T cells from wild type and mu-opioid receptor knockout mice. Our results show that morphine inhibited anti-CD3/CD28-stimulated IFN-gamma promoter activity in a dose-dependent manner. Chronic morphine treatment of T cells increased intracellular cAMP. To evaluate the role of cAMP in morphine's modulatory function, the effects of dibutyryl cyclic AMP and forskolin were investigated. Both dibutyryl cyclic AMP and forskolin treatment inhibited IFN-gamma promoter activity. Treatment with pertussis toxin, but not with a protein kinase A inhibitor, antagonized morphine's inhibitory effects. Morphine inhibited phosphorylation of ERK1/2 and p38 MAPK; in addition, morphine treatment in the presence of either ERK1/2 or p38 MAPK inhibitor (PD98059 or SB203580) resulted in an additive inhibition of IFN-gamma promoter activity. The transcription factor activator protein-1, NF-kappaB, and nuclear factor of activated T cells (NFAT) were negatively regulated by morphine. Overexpression of NF-kappaB p65 rescued the inhibitory effect of morphine on IFN-gamma promoter activity. However, only when NFATc1 was co-overexpressed with c-fos was the inhibitory effect of morphine on IFN-gamma promoter counteracted. The inhibitory effects of morphine were not observed in T cells obtained from mu-opioid receptor knockout mice, suggesting that morphine modulation of IFN-gamma promoter activity is mediated through the mu-opioid receptor. In summary, our data indicate that morphine modulation of IFN-gamma promoter activity is mediated through two distinct cAMP-dependent pathways, the NF-kappaB signaling pathway and the ERK1/2, p38 MAPK, AP-1/NFAT pathway.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
26
|
Li Y, Zhang T, Douglas SD, Lai JP, Xiao WD, Pleasure DE, Ho WZ. Morphine enhances hepatitis C virus (HCV) replicon expression. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1167-75. [PMID: 12937158 PMCID: PMC1868275 DOI: 10.1016/s0002-9440(10)63476-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Little information is available regarding whether substance abuse enhances hepatitis C virus (HCV) replication and promotes HCV disease progression. We investigated whether morphine alters HCV mRNA expression in HCV replicon-containing liver cells. Morphine significantly increased HCV mRNA expression, an effect which could be abolished by either of the opioid receptor antagonists, naltrexone or beta-funaltrexamine. Investigation of the mechanism responsible for this enhancement of HCV replicon expression demonstrated that morphine activated NF-kappaB promoter and that caffeic acid phenethyl ester, a specific inhibitor of the activation of NF-kappaB, blocked morphine-activated HCV RNA expression. In addition, morphine compromised the anti-HCV effect of interferon alpha (IFN-alpha). Our in vitro data indicate that morphine may play an important role as a positive regulator of HCV replication in human hepatic cells and may compromise IFN-alpha therapy.
Collapse
Affiliation(s)
- Yuan Li
- Division of Immunologic and Infectious Diseases, Department of Pediatrics, Stokes Research Institute, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, 34th Street & Civic Center Boulevard, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The use of recreational drugs of abuse has generated serious health concerns. There is a long-recognized relationship between addictive drugs and increased levels of infections. Studies of the mechanisms of actions of these drugs became more urgent with the advent of AIDS and its correlation with abused substances. The nature and mechanisms of immunomodulation by marijuana, opiates, cocaine, nicotine, and alcohol are described in this review. Recent studies of the effects of opiates or marijuana on the immune system have demonstrated that they are receptor mediated, occurring both directly via specific receptors on immune cells and indirectly through similar receptors on cells of the nervous system. Findings are also discussed that demonstrate that cocaine and nicotine have similar immunomodulatory effects, which are also apparently receptor mediated. Finally, the nature and mechanisms of immunomodulation by alcohol are described. Although no specific alcohol receptors have been identified, it is widely recognized that alcohol enhances susceptibility to opportunistic microbes. The review covers recent studies of the effects of these drugs on immunity and on increased susceptibility to infectious diseases, including AIDS.
Collapse
Affiliation(s)
- Herman Friedman
- Department of Medical Microbiology and Immunology, College of Medicine, University of South Florida, Tampa, Florida 33612, USA.
| | | | | |
Collapse
|
28
|
Tomassini N, Renaud FL, Roy S, Loh HH. Mu and delta receptors mediate morphine effects on phagocytosis by murine peritoneal macrophages. J Neuroimmunol 2003; 136:9-16. [PMID: 12620638 DOI: 10.1016/s0165-5728(02)00463-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Studies with selective opioid agonists show that mu- and delta(2)-opioid receptors, but not kappa, are involved in opioid inhibition of phagocytosis in elicited murine macrophages. All mu and delta(2) agonists tested had similar maximal effects on phagocytosis, and all dose-response curves suggest positive cooperativity. In addition, mu and delta antagonists antagonized the effect of both mu and delta agonists. Furthermore, in mu-opioid receptor knockout mice (MORKO), we observed a decrease in potency and maximal effect for a delta agonist. These data suggest that mu and delta receptors are not only involved in the modulation of phagocytosis in macrophages, but they also affect each other's activity by an unknown cooperative mechanism.
Collapse
MESH Headings
- Animals
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/pharmacology
- Narcotic Antagonists/pharmacology
- Peritoneum/cytology
- Peritoneum/drug effects
- Peritoneum/immunology
- Phagocytosis/drug effects
- Phagocytosis/immunology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/deficiency
Collapse
Affiliation(s)
- Nilka Tomassini
- Biology Department, University of Puerto Rico, POB 23360 Río Piedras Campus, San Juan 00931-3360, Puerto Rico
| | | | | | | |
Collapse
|
29
|
Hultén C, Grönlund U, Hirvonen J, Tulamo RM, Suominen MM, Marhaug G, Forsberg M. Dynamics in serum of the inflammatory markers serum amyloid A (SAA), haptoglobin, fibrinogen and alpha2-globulins during induced noninfectious arthritis in the horse. Equine Vet J 2002; 34:699-704. [PMID: 12455841 DOI: 10.2746/042516402776250405] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Despite the importance of noninfectious joint diseases in equine medicine, little is known about the acute phase response which may be elicited if the local inflammatory process of noninfectious arthritis is sufficiently strong, Therefore the aim of this study was to monitor the systemic inflammatory response during experimentally-induced noninfectious arthritis by studying the dynamics in serum of the acute phase proteins serum amyloid A (SAA), haptoglobin, fibrinogen and alpha2-globulins. Twenty-four Standardbred horses, age 3-7 years, found healthy on thorough clinical, radiological, haematological and serum biochemical examination, were injected aseptically into the right midcarpal joint with amphotericin B. Blood samples were drawn before induction of arthritis (0 h), and at 8, 16, 24, 36 and 48 h postinduction and then on Days 3, 4, 5 and 15 postinduction. All horses developed lameness with joint effusion and joint heat as well as increased respiratory rate, heart rate and body temperature. The lameness started to decline after 24-36 h and, in most animals, systemic signs disappeared on Day 2 postinjection. The concentration of the acute phase proteins increased following induction of arthritis. The SAA concentrations were higher than baseline concentrations from 16 h postinduction and were maximal at 36-48 h (227 times baseline concentration). The haptoglobin concentrations were higher than baseline concentrations from 24 h and were maximal at 48-96 h (1.14 times baseline concentration). The maximal concentrations of fibrinogen were seen between 36-72 h postinjection and increased on average 0.87 times from baseline concentrations. The fibrinogen concentrations were higher than baseline concentrations from 24 h postinjection. Alpha2-globulins concentrations showed a minor increase and increased 0.55 times from baseline concentrations. The markers had returned to baseline concentrations by Day 15. Our results demonstrate that amphotericin B-induced arthritis in a single joint gives rise to a systemic acute phase response measurable as increased concentrations in serum SAA, haptoglobin, fibrinogen and alpha2-globulins during the first 2 weeks of the condition and, thereby, that such an increase need not be indicative of infectious arthritis. Further research should be aimed at determining whether chronic noninfectious arthritis in the horse gives rise to increased acute phase protein concentrations in serum.
Collapse
Affiliation(s)
- C Hultén
- Department of Clinical Chemistry, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, Uppsala
| | | | | | | | | | | | | |
Collapse
|
30
|
Azuma Y, Ohura K. Endomorphin-2 modulates productions of TNF-alpha, IL-1beta, IL-10, and IL-12, and alters functions related to innate immune of macrophages. Inflammation 2002; 26:223-32. [PMID: 12238565 DOI: 10.1023/a:1019766602138] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We evaluate immunological effects of opioid peptide endomorphin-2 on the production of cytokines related to inflammation and Th1/Th2 balance, and functions related to innate immune of rat peritoneal macrophages. Endomorphin-2 inhibited TNF-alpha, IL-10, and IL-12 productions, but potentiated IL-1beta production by macrophages. Moreover, endomorphin-2 potentiated macrophage adhesion to fibronectin, and the expression of adhesion molecule Mac-1 on macrophages. In contrast, endomorphin-2 suppressed phagocytosis of opsonized E. coli by macrophages, without affecting phagocytosis of non-opsonized E. coli. In addition, endomorphin-2 inhibited macrophage chemotaxis, and the production of superoxide anion by macrophages. These results suggest that endomorphin-2 may alter macrophage functions such as cytokine productions and functions related to innate immune.
Collapse
Affiliation(s)
- Yasutaka Azuma
- Department of Pharmacology, Osaka Dental University, Hirakata, Japan.
| | | |
Collapse
|
31
|
Azuma Y, Ohura K. Endomorphins 1 and 2 inhibit IL-10 and IL-12 production and innate immune functions, and potentiate NF-kappaB DNA binding in THP-1 differentiated to macrophage-like cells. Scand J Immunol 2002; 56:260-9. [PMID: 12193227 DOI: 10.1046/j.1365-3083.2002.01128.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We evaluated immunological effects of opioid peptides endomorphins 1 and 2 on the production of interleukin-10 (IL-10) and IL-12 cytokines, functions related to innate immunity and NF-kappaB DNA binding in human cell line THP-1. Endomorphins 1 and 2 inhibited lipopolysaccharide (LPS)-stimulated IL-10 and IL-12 production in THP-1 differentiated to macrophage-like cells by phorbol 12-myristate 13-acetate (PMA). Similarly, they suppressed LPS-stimulated IL-10 and IL-12 production in THP-1 matured to monocytes by 1alpha,25-dihydroxyvitamin D3. In addition, endomorphins 1 and 2 led to marked potentiation of NF-kappaB binding in THP-1 differentiated to macrophage-like cells. Furthermore, these endomorphins further potentiated LPS-induced NF-kappaB binding. Moreover, they inhibited chemotaxis, phagocytosis of Escherichia coli and PMA-stimulated production of hydrogen peroxide in THP-1 differentiated to macrophage-like cells. These results suggest that endomorphins 1 and 2 may inhibit THP-1 functions, such as cytokine production and functions related to innate immune, and potentiate NF-kappaB DNA binding in THP-1.
Collapse
Affiliation(s)
- Y Azuma
- Department of Pharmacology, Osaka Dental University, Hirakata, Osaka, Japan.
| | | |
Collapse
|
32
|
Inui Y, Azuma Y, Ohura K. Differential alteration of functions of rat peritoneal macrophages responsive to endogenous opioid peptide endomorphin-1. Int Immunopharmacol 2002; 2:1133-42. [PMID: 12349950 DOI: 10.1016/s1567-5769(02)00065-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Endomorphin-1 is a recently isolated endogenous opioid peptide, and potent and selective high affinity mu-opioid receptor agonist. We evaluate the role of endomorphin-1 on macrophage functions. Endomorphin-1 potentiated macrophage adhesion and the expression of adhesion molecule Mac-1 on macrophages. However, endomorphin-1 did not alter phagocytosis of Escherichia coli by macrophages. Moreover, endomorphin-1 inhibited macrophage chemotaxis and the production of superoxide anion by macrophages. On the contrary, endomorphin-1 inhibited TNF-alpha production by macrophages stimulated with both LPS and PMA, respectively. Similarly, endomorphin-1 suppressed IL-10 and IL-12 productions in response to LPS. In contrast, endomorphin-1 potentiated IL-1beta production by macrophages stimulated with PMA. These results suggest that endomorphin-1 may alter macrophage functions such as cytokine productions and functions related to natural host defense.
Collapse
Affiliation(s)
- Yoshiro Inui
- Department of Pharmacology, Osaka Dental University, Hirakata, Japan
| | | | | |
Collapse
|
33
|
Singhal PC, Bhaskaran M, Patel J, Patel K, Kasinath BS, Duraisamy S, Franki N, Reddy K, Kapasi AA. Role of p38 mitogen-activated protein kinase phosphorylation and Fas-Fas ligand interaction in morphine-induced macrophage apoptosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4025-33. [PMID: 11937560 DOI: 10.4049/jimmunol.168.8.4025] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we evaluated the molecular mechanisms involved in morphine-induced macrophage apoptosis. Both morphine and TGF-beta promoted P38 mitogen-activated protein kinase (MAPK) phosphorylation, and this phosphorylation was inhibited by SB 202190 as well as by SB 203580. Anti-TGF-beta Ab as well as naltrexone (an opiate receptor antagonist) inhibited morphine-induced macrophage P38 MAPK phosphorylation. Anti-TGF-beta Ab also attenuated morphine-induced p53 as well as inducible NO synthase expression; in contrast, N(G)-nitro-L-arginine methyl ester, an inhibitor of NO synthase, inhibited morphine-induced P38 MAPK phosphorylation and Bax expression. Morphine also enhanced the expression of both Fas and Fas ligand (FasL), whereas anti-FasL Ab prevented morphine-induced macrophage apoptosis. Moreover, naltrexone inhibited morphine-induced FasL expression. In addition, macrophages either deficient in FasL or lacking p53 showed resistance to the effect of morphine. Inhibitors of both caspase-8 and caspase-9 partially prevented the apoptotic effect of morphine on macrophages. In addition, caspase-3 inhibitor prevented morphine-induced macrophage apoptosis. These findings suggest that morphine-induced macrophage apoptosis proceeds through opiate receptors via P38 MAPK phosphorylation. Both TGF-beta and inducible NO synthase play an important role in morphine-induced downstream signaling, which seems to activate proteins involved in both extrinsic (Fas and FasL) and intrinsic (p53 and Bax) cell death pathways.
Collapse
Affiliation(s)
- Pravin C Singhal
- Immunology and Inflammation Center, North Shore-Long Island Jewish Research Institute and Division of Kidney Diseases and Hypertension, Long Island Jewish Medical Center, New Hyde Park, NY 11040, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Li Y, Wang X, Tian S, Guo CJ, Douglas SD, Ho WZ. Methadone enhances human immunodeficiency virus infection of human immune cells. J Infect Dis 2002; 185:118-22. [PMID: 11756991 PMCID: PMC4009627 DOI: 10.1086/338011] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2001] [Revised: 09/24/2001] [Indexed: 11/03/2022] Open
Abstract
Opiate abuse has been postulated to be a cofactor in the immunopathogenesis of acquired immunodeficiency syndrome (AIDS). This study evaluated whether methadone, a drug widely prescribed for the treatment of drug abusers with opioid dependence, affects human immunodeficiency virus (HIV) infection of human immune cells. When added to human fetal microglia and blood monocyte-derived macrophage cultures, methadone significantly enhanced HIV infection of these cells. This enhancement was associated with the up-regulation of expression of CCR5, a primary coreceptor for macrophage-tropic HIV entry into macrophages. Most importantly, the addition of methadone to the cultures of latently infected peripheral blood mononuclear cells from HIV-infected patients enhanced viral activation and replication. Although the in vivo relevance of these findings remains to be determined, the data underscore the necessity of further studies to define the role of opioids, including methadone, in the immunopathogenesis of HIV infection and AIDS.
Collapse
Affiliation(s)
- Yuan Li
- Division of Immunologic and Infectious Diseases, Joseph Stokes Jr. Research Institute of The Children's Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
It is now clear that opioid receptors participate in the function of the cells of the immune system, and evidence suggests that opioids modulate both innate and acquired immune responses. We review literature here which establishes that mu-, kappa-, and delta-opioid compounds alter resistance to a variety of infectious agents, including the Human Immunodeficiency Virus (HIV). The nature of the immunomodulatory activity of the opioids has been the subject of a great deal of research over the last ten years. There is increasing evidence that effects of opioids on the immune response are mediated at several levels. Modulation of the inflammatory response appears to be a target of these compounds, including effects on phagocytic activity, as well as the response of cells to various chemoattractant molecules. Moreover, findings from several laboratories have demonstrated the impact of opioid treatment on antibody responses, and the molecular basis for this effect is likely due, at least in part, to the modulation of both cytokine and cytokine receptor expression. Future research should provide a clearer understanding of the cellular and molecular targets of opioid action within the immune system.
Collapse
Affiliation(s)
- L McCarthy
- Department of Microbiology and Immunology, Temple University School of Medicine, 3400 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
36
|
Welters ID, Menzebach A, Goumon Y, Langefeld TW, Teschemacher H, Hempelmann G, Stefano GB. Morphine suppresses complement receptor expression, phagocytosis, and respiratory burst in neutrophils by a nitric oxide and mu(3) opiate receptor-dependent mechanism. J Neuroimmunol 2000; 111:139-45. [PMID: 11063831 DOI: 10.1016/s0165-5728(00)00401-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We investigated whether morphine and fentanyl influence surface receptor expression, phagocytic activity and superoxide anion generation of neutrophils in a whole blood flow cytometric assay. Morphine suppressed complement and Fcgamma receptor expression and neutrophil function in a concentration- and time-dependent manner. Morphine-induced changes were similar to those caused by the nitric oxide (NO) donor S-nitroso-N-acetyl-penicillamine and were abolished by preincubation with the NO synthase inhibitor N-nitro-L-arginine as well as naloxone. Fentanyl had no immunosuppressive effects. These results suggest that these neutrophil functions are inhibited by morphine-stimulated NO release mediated by the mu(3) opiate receptor subtype found on immunocytes.
Collapse
Affiliation(s)
- I D Welters
- Department of Anaesthesiology and Intensive Care Medicine, Justus-Liebig-University Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Small CB, McGowan JP, Klein RS, Schnipper SM, Chang CJ, Rosenstreich DL. Serum IgE levels in patients with human immunodeficiency virus infection. Ann Allergy Asthma Immunol 1998; 81:75-80. [PMID: 9690576 DOI: 10.1016/s1081-1206(10)63112-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Increased serum IgE levels are associated with advanced HIV infection. The magnitude of the increase has varied greatly between studies which generally did not assess potential confounding factors. OBJECTIVE To determine whether the increased serum IgE levels reported with HIV infection is affected by demographic or behavioral factors, we studied injection drug users, women, and minority ethnic and racial groups with HIV infection, for whom little data now exist. METHODS A prospective cross-sectional study of ambulatory patients with or at risk for HIV infection was performed. We enrolled 83 injection drug users and 56 non-drug users seropositive for HIV and 43 seronegative at-risk individuals from an Infectious Diseases clinic and a longitudinal study of HIV infection in injection drug users in the Bronx, New York City. Fifteen HIV-seronegative non-atopic controls were also studied. Total serum IgE levels were measured by a solid phase fluorescent assay and lymphocyte phenotypes were measured by monoclonal antibodies. RESULTS On multiple linear regression analysis, HIV infection (P=.01) and advanced HIV disease (P < or =.01) were independently associated with increased serum IgE levels, controlling for gender, race, age, and use of injection drugs. In both HIV-seronegative and seropositive individuals, female gender was independently associated with lower IgE levels (P < or = .001). We did not find an independent effect of race or injection drug use on IgE levels. CONCLUSIONS Increased serum IgE levels were associated with HIV infection, the highest levels existing in those with advanced HIV disease. Women had lower IgE levels than men, independent of HIV status. Active or past drug use, race, and age were not found to be independently associated with serum IgE levels. Further studies are necessary to elucidate the mechanisms underlying the increased serum IgE levels seen with HIV infection and its associated immunodeficiency, and to substantiate and explore the decreased levels found in women.
Collapse
Affiliation(s)
- C B Small
- Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York 10467-2490, USA
| | | | | | | | | | | |
Collapse
|
38
|
Eisenstein TK, Hilburger ME. Opioid modulation of immune responses: effects on phagocyte and lymphoid cell populations. J Neuroimmunol 1998; 83:36-44. [PMID: 9610671 DOI: 10.1016/s0165-5728(97)00219-1] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The literature describing effects of morphine on cells of the immune system points to the clear conclusion that morphine given in vivo suppresses a variety of immune responses that involve the major cell types in the immune system, including natural killer (NK) cells, T cells, B cells, macrophages and polymorphonuclear leukocytes (PMNs). Depression of NK cell activity has been reported in humans, monkeys and rodents. Similarly, responses of T cells are depressed by morphine, as assessed by inhibition of induction of delayed-type hypersensitivity reactions and cytotoxic T-cell activity, modulation of T-cell antigen expression, and depression of responses to T-cell mitogens. Effects on T cells have been reported in humans, monkeys and rodents. Effects of morphine on B-cell activity have mainly been tested in rodents using assays of antibody formation, which also require macrophages and T cells, preventing a conclusion as to the cell type being affected. Consistent effects on phagocytic cell function have been reported in rodents given morphine. In contrast, studies on immunomodulatory effects of morphine added to cells of the immune system in vitro have shown robust effects on some of these cell types, but not others. There is a rich literature demonstrating downregulation of phagocytic cell function by morphine, particularly for human peripheral blood mononuclear cells (PBMCs) and PMNs. Phagocytosis, chemotactic responses, interleukin production, and generation of activated oxygen intermediates and arachidonic acid products have all been reported to be inhibited. On the contrary, the literature does not support direct effects of morphine on NK cell function, is inconclusive concerning effects on B cells, and provides limited evidence for effects on T cells. The divergence between the in vivo and in vitro data suggests that effects on some cells in the immune system observed after in vivo morphine are probably not direct, but mediated. In aggregate, the literature supports the existence of an in vivo neural-immune circuit through which morphine acts to depress the function of all cells of the immune system. Further, there is strong evidence that morphine can directly depress the function of macrophages and PMNs, and modulate expression of one type of T-cell surface marker. There is, however, little evidence for direct effects of morphine on NK cells and B cells. A further complication emerges from reports of immunopotentiation of immune function in in vitro assays using endogenous opioids. The possibility of different receptors for endogenous and exogenous opioids or of interactions among the activated opioid receptors may account for these opposing effects.
Collapse
Affiliation(s)
- T K Eisenstein
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | |
Collapse
|
39
|
|
40
|
Reply. Am J Obstet Gynecol 1997. [DOI: 10.1016/s0002-9378(97)70249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
An Evaluation of the Antiinflammatory Effects of Intraarticular Synthetic beta-Endorphin in the Canine Model. Anesth Analg 1996. [DOI: 10.1213/00000539-199601000-00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
42
|
Martinez JH, Mondragón CE, Céspedes A. An evaluation of the antiinflammatory effects of intraarticular synthetic beta-endorphin in the canine model. Anesth Analg 1996; 82:177-81. [PMID: 8712398 DOI: 10.1097/00000539-199601000-00033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We sought to evaluate the antiinflammatory effects of synthetic human beta-endorphin (SHB) when injected into the canine knee joint. Sixteen healthy dogs, aged 1-2 yr, were selected. SHB was injected pre- and postinjury into each knee. The sample size was n = 32 after a randomized factorial arrangement; 2 x 4 with four cases per treatment being performed. Factors considered were: Factor A with two levels: A1 = Preinjury and A2 = Postinjury; Factor B (SHB dose) with four levels: B1 = Control, B2 = 250 micrograms, B3 = 500 micrograms, B4 = 1000 micrograms. The control group received 0.9% NaCl solution. Anesthesia was induced with intravenous thiopental, 14 mg/kg, and acepromazine, 0.5 mg/kg. Injury was produced with an intraarticular injection of 4 mL HCl 0.5 M, which was left in situ for 20 min. Inflammation was measured using the 610 nm absorbency of Evans blue extravasate in biopsy specimens. Histopathologic studies were performed on each knee. We found that beta-endorphin has a clear, dose-related, antiinflammatory effect, reducing the tissue extravasation of Evans blue and its absorbency, especially with large doses. This finding was consistent with the histopathologic observations. We conclude that SHB has an antiinflammatory effect. It is still not clear which mechanisms inhibit polymorphonuclear cell adhesion to vascular endothelium or cell and plasmatic protein extravasation.
Collapse
Affiliation(s)
- J H Martinez
- Veterinary and Zootechnist School, Tolima University, Ibagué, Columbia
| | | | | |
Collapse
|
43
|
Gavériaux C, Peluso J, Simonin F, Laforet J, Kieffer B. Identification of kappa- and delta-opioid receptor transcripts in immune cells. FEBS Lett 1995; 369:272-6. [PMID: 7649271 DOI: 10.1016/0014-5793(95)00766-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To investigate the role of opioids as direct modulators of the immune response, we have searched for expression of the recently cloned delta, mu and kappa opioid receptors in immune cells. We have devised a reverse transcriptase-polymerase chain reaction strategy which specifically detects a region spanning putative transmembrane regions 2 to 7 for each transcript in both human and mouse immune cells. In human peripheral blood lymphocyte and monocyte preparations, delta was undetectable while the kappa transcript was present. The analysis of human cell lines revealed low but significant levels of delta opioid receptor transcripts in T, B or monocyte cell lines while the kappa transcript was found in B cell lines only. Investigation of murine cells showed the presence of transcript for the delta receptor in splenocytes and in some T and B cell lines. Unexpectedly, no expression of the mu receptor was detected. Sequence analysis of PCR products demonstrated nucleotide identity between immune and neuronal transcripts, indicating that they derive from the same genes. In conclusion, our results lead to the identification of kappa and delta opioid receptor transcripts in immune cells.
Collapse
MESH Headings
- Animals
- Base Sequence
- Brain/immunology
- Cell Line
- Cloning, Molecular
- Humans
- Leukocytes/metabolism
- Lymph Nodes/immunology
- Mice
- Molecular Sequence Data
- Polymerase Chain Reaction/methods
- RNA, Messenger/biosynthesis
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/genetics
- Sequence Analysis, DNA
- Spleen/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C Gavériaux
- Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | | | | | | | | |
Collapse
|
44
|
Thomas PT, House RV, Bhargava HN. Direct cellular immunomodulation produced by diacetylmorphine (heroin) or methadone. GENERAL PHARMACOLOGY 1995; 26:123-30. [PMID: 7713351 DOI: 10.1016/0306-3623(94)00162-g] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
1. Abuse of the narcotic drug diacetylmorphine (heroin), as well as methadone, a drug for treating heroin addiction, has been associated with alterations in immune function in humans. The current study was performed to assess the direct (in vitro) immunomodulatory effect of exposure to these drugs, in view of the very limited studies reported thus far on this effect. 2. Murine splenocytes or peritoneal macrophages were cultured in vitro at concentrations of 0.0001-100 microM heroin or methadone. B-cell function was assessed by quantitating cellular proliferation in response to stimulation with an antigen analog; T-cell regulatory function was assessed by culturing splenocytes with or without drugs in the presence of anti-CD3 antibody and subsequently quantitating cytokine production; and T-cell effector function was evaluated by culturing lymphocytes with or without drugs during a 5-day induction culture followed by assessment of specific cytotoxic T-lymphocyte (CTL) activity. Natural immunity was assessed by quantitating basal and IL-2 augmented natural killer (NK) cell function, and macrophage function was assessed by cytokine production. 3. In vitro exposure to heroin resulted in decreased B-cell proliferation at concentrations of 1-100 microM, and methadone had a similar effect at concentrations of 0.1-100 microM. 4. Production of IL-2 was suppressed by 0.1-100 microM of heroin, whereas exposure to methadone appeared to result in a generalized modulation, with suppression of IL-2 at most concentrations. In contrast, IL-4 production was only affected at the 100 microM concentration of both drugs.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- P T Thomas
- Life Sciences Department, IIT Research Institute, Chicago, IL 60616, USA
| | | | | |
Collapse
|
45
|
Mazzone A, Mazzucchelli I, Fossati G, Gritti D, Fea M, Ricevuti G. Granulocyte defects and opioid receptors in chronic exposure to heroin or methadone in humans. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1994; 16:959-67. [PMID: 7868301 DOI: 10.1016/0192-0561(94)90049-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In order to elucidate better the immunological effect of opioid abuse in the absence of HIV infection as a confounding factor, granulocyte function was investigated in three groups of HIV-negative subjects, including 20 active parenteral heroin abusers (H), 20 long-term methadone-maintained former opiate abusers (M) and 20 healthy controls (C). Chemotaxis to N-formyl methionyl-leucyl-phenylalanine (fMLP), casein and activated plasma were markedly and similarly reduced (approx. 50%) in both H and M groups, as was true for superoxide production after fMLP and PMA stimulation, 47% decrease of C values. Polymorphonuclear (PMN) of H and M subjects also exhibited a very marked and similar reduction in the expression of CD11b/CD18 integrin receptors after fMLP treatment, with values that were less than 10% of those in controls, as observed by flow cytometry. In parallel, PMN of H and M individuals presented an approximately four-fold increase in opioid receptors numbers compared to controls, a significant inverse correlation existing between the increase in opiate receptors and defective chemotaxis. The possible mechanism underlying the observed changes in PMN of H and M individuals is discussed.
Collapse
Affiliation(s)
- A Mazzone
- Department of Internal Medicine and Therapeutics, University of Pavia, IRCCS San Matteo Hospital, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Carballo-Diéguez A, Sahs J, Goetz R, el Sadr W, Sorell S, Gorman J. The effect of methadone on immunological parameters among HIV-positive and HIV-negative drug users. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 1994; 20:317-29. [PMID: 7977217 DOI: 10.3109/00952999409106017] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Our objective was to assess the effects of methadone use on immune parameters. A convenience sample of men and women drug injectors who knew their HIV serostatus were enrolled in a longitudinal observational study of HIV illness. During analysis of baseline data, differences were noted in immune parameters among Methadone users. Study participants were recruited in Manhattan, New York, from a methadone maintenance clinic, and infectious disease clinic of an inner city hospital, and a drug-free community center. The participants were 220 men and women, current or former drug injectors, approximately half of them HIV-antibody positive and the rest HIV-antibody negative. Candidates with opportunistic infections and secondary neoplasms were excluded. Methadone users were compared to nonmethadone users for absolute and percentage counts of CD4, CD8, and activated T lymphocytes; CD4/CD8 ratio; an HIV symptom check list; and medical staging. The results discussed in this paper were formulated after data collection was complete. Our data indicate that methadone treatment, while not significantly affecting absolute CD4 lymphocyte count, is associated with a lower CD4 percentage and CD4/CD8 cell ratio, and with a higher CD8 absolute count and percentage. These differences are present regardless of HIV serostatus. Our findings should be interpreted with caution since we did not set out to investigate the effects of methadone on the immune system. Nevertheless, if it is corroborated that methadone has a detrimental effect on the immune system, finding alternatives to methadone-maintenance treatment for drug injectors will be a necessity.
Collapse
Affiliation(s)
- A Carballo-Diéguez
- HIV Center for Clinical and Behavioral Studies, New York State Psychiatric Institute and Columbia University, New York
| | | | | | | | | | | |
Collapse
|
47
|
Como-Lesko N, Primavera LH, Szeszko PR. Marijuana usage in relation to harmfulness ratings, perceived likelihood of negative consequences, and defense mechanisms in high school students. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 1994; 20:301-15. [PMID: 7977216 DOI: 10.3109/00952999409106016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This study investigated high school students' marijuana usage patterns in relation to their harmfulness ratings of 15 licit and illicit drugs, perceived negative consequences from using marijuana, and types of defense mechanisms employed. Subjects were classified into one of five pattern-of-use groups based on marijuana usage: principled nonusers, nonusers, light users, moderate users, and heavy users. Principled nonusers (individuals who have never used marijuana and would not do so if it was legalized) rated marijuana, hashish, cocaine, and alcohol as significantly more harmful than heavy users. A cluster analysis of the drugs' harmfulness ratings best fit a three cluster solution and were named medicinal drugs, recreational drugs, and hard drugs. In general, principled nonusers rated negative consequences from using marijuana as significantly more likely to occur than other groups. Principled nonusers and heavy users utilized reversal from the Defense Mechanism Inventory, which includes repression and denial, significantly more than nonusers, indicating some trait common to the two extreme pattern-of-use groups.
Collapse
|
48
|
Mazzone A, Ricevuti G, Pasotti D, Fossati G, Mazzucchelli I, Cavigliano P, Lecchini S, Frigo G, Notario A. Role of increase of opioid receptors in granulocyte CD11b/CD18 dysfunction. Ann N Y Acad Sci 1993; 685:770-3. [PMID: 8103315 DOI: 10.1111/j.1749-6632.1993.tb35941.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- A Mazzone
- Department of Internal Medicine and Therapeutics, University of Pavia, IRCCS San Matteo Hospital, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chao CC, Molitor TW, Close K, Hu S, Peterson PK. Morphine inhibits the release of tumor necrosis factor in human peripheral blood mononuclear cell cultures. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1993; 15:447-53. [PMID: 8389331 DOI: 10.1016/0192-0561(93)90057-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Opiates modulate a variety of immune responses of peripheral blood mononuclear cells (PBMC). When PBMC were treated with morphine for 24 h, cells released less (P < 0.05) bioactive TNF, a cytokine important in host defense, in the following 24-h incubation period when stimulated with lipopolysaccharide and phytohemagglutinin. Morphine alone did not significantly alter the release of TNF from PBMC cultures. Pretreatment of PBMC cultures for 1 h with naloxone blocked (P < 0.05) the inhibitory effect of morphine on the release of TNF upon stimulation with phytohemagglutinin, but not with lipopolysaccharide, suggesting the involvement of an opioid receptor. The mechanism of morphine-induced suppression of TNF release appears to be counteracted by the effect of this opiate on the release of transforming growth factor (TGF)-beta, since antibodies to this immunoregulatory cytokine further enhanced morphine-related inhibition of TNF release. Taken together, these findings indicate that morphine suppresses the release of bioactive TNF from PBMC and that TGF-beta plays a modulatory role in this inhibitory process.
Collapse
Affiliation(s)
- C C Chao
- Neuroimmunobiology and Host Defense Laboratory, Minneapolis Medical Research Foundation, Minnesota 55404
| | | | | | | | | |
Collapse
|
50
|
Zhou Y, Scamurra R, Molitor TW, Murtaugh MP. Characterization of transforming growth factor-beta 1 gene expression in porcine immune cells. Mol Immunol 1992; 29:965-70. [PMID: 1386143 DOI: 10.1016/0161-5890(92)90135-k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have investigated the regulation of transforming growth factor beta 1 gene expression in a variety of porcine immune cell populations, including peripheral blood mononuclear cells (PBMC), peripheral blood monocytes, alveolar macrophages and lymphoid cells from various swine lymphoid tissues. Using porcine transforming growth factor beta 1 cDNA probes in Northern blot assays, messages of 2.5 and 3.5 kb TGF beta 1 mRNA were detected in the cells investigated. A variety of mitogenic and immunomodulatory substances were examined for their ability to induce TGF beta 1 mRNA expression. These include phorbol 12-myristate 13-acetate (PMA), phytohemagglutinin (PHA), concanavalin A (Con A), lipopolysaccharide (LPS), dexamethasone (Dex), tumor necrosis factor (TNF) and interleukin (IL)-1 alpha. While low level constitutive expression of TGF beta 1 mRNA was detected from all cells investigated, PMA treatment of PBMC and alveolar macrophages resulted in a more than 10-fold increase in the steady-state level of TGF beta 1 mRNA within 2 hr of PMA addition. Also, the effect of opiate drugs, methadone (Md) and morphine (Mor), on TGF beta 1 gene expression was determined. Cells treated with opiates expressed the same levels of TGF beta 1 mRNA as untreated cells. Since TGF beta 1 biological activity can be induced by opiates, the regulation of TGF beta 1 gene expression likely involves mechanisms that do not cause changes in mRNA levels.
Collapse
Affiliation(s)
- Y Zhou
- Department of Veterinary Pathobiology, University of Minnesota, St. Paul 55108
| | | | | | | |
Collapse
|