1
|
Grant I, Krupitsky E, Vetrova M, Umlauf A, Heaton RK, Hauger RL, Toussova O, Franklin DR, Letendre SL, Woody G, Blokhina E, Lioznov D, Zvartau E. Effects of Opioid Withdrawal on Psychobiology in People Living with HIV. Viruses 2024; 16:92. [PMID: 38257791 PMCID: PMC10818595 DOI: 10.3390/v16010092] [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: 10/23/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE Many persons with opioid use disorders (OUDs) have HIV disease and experience clinically significant stress after they enroll in abstinence-based treatment and undergo medically assisted withdrawal. We examined whether opioid withdrawal affects virologic control, inflammatory markers, cognition, and mood in persons with an OUD and HIV, and explored whether measures of withdrawal stress, such as activation of the HPA axis, contribute to alterations in immune function, cognition, and mood. METHOD AND PARTICIPANTS Study participants were 53 persons with HIV who were admitted for OUD treatment at the City Addiction Hospital in Saint Petersburg, Russian Federation. Participants were examined at admission, at the anticipated peak of withdrawal 3 to 7 days after the last day of a clonidine-based withdrawal process lasting 7 to 14 days, and 3 to 4 weeks after completing withdrawal. At these times, participants received medical exams and were evaluated for symptoms of withdrawal, as well as cognition and mood. Viral load, plasma cortisol, DHEA sulfate ester (DHEA-S), interleukin-6 (IL-6), and soluble CD14 (sCD14) were determined. Multivariable models examined the relationships between markers of HPA activation and the other parameters over time. RESULTS HPA activation as indexed by cortisol/DHEA-S ratio increased during withdrawal, as did markers of immune activation, IL-6 and sCD14. There were no significant associations between viral load and indicators of HPA activation. In longitudinal analyses, higher cortisol/DHEA sulfate was related to worse cognition overall, and more mood disturbance. Increase in IL-6 was associated with worse cognitive performance on a learning task. There were no significant associations with sCD14. CONCLUSIONS Worsening of cognition and measures of mood disturbance during withdrawal were associated with activation of the HPA axis and some measures of inflammation. Whether repeated episodes of opioid withdrawal have a cumulative impact on long-term HIV outcomes and neurocognition is a topic for further investigation.
Collapse
Affiliation(s)
- Igor Grant
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
| | - Evgeny Krupitsky
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
- Department of Addictions, Bekhterev National Medical Research Center for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
| | - Marina Vetrova
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
| | - Anya Umlauf
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
| | - Robert K. Heaton
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
| | - Richard L. Hauger
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Olga Toussova
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
| | - Donald R. Franklin
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
| | - Scott L. Letendre
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California San Diego, San Diego, CA 92103, USA; (A.U.); (R.K.H.); (R.L.H.); (D.R.F.); (S.L.L.)
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | - George Woody
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Elena Blokhina
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
| | - Dmitry Lioznov
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
| | - Edwin Zvartau
- Department of Pharmacology, Pavlov State Medical University, 197022 Saint Petersburg, Russia; (E.K.); (M.V.); (O.T.); (E.B.); (D.L.); (E.Z.)
| |
Collapse
|
2
|
Ketchem JM, Bowman EJ, Isales CM. Male sex hormones, aging, and inflammation. Biogerontology 2023; 24:1-25. [PMID: 36596999 PMCID: PMC9810526 DOI: 10.1007/s10522-022-10002-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2023]
Abstract
Adequate levels of androgens (eugonadism), and specifically testosterone, are vital compounds for male quality of life, longevity, and positive health outcomes. Testosterone exerts its effects by binding to the androgen receptor, which is expressed in numerous tissues throughout the body. Significant research has been conducted on the impact of this steroid hormone on skeletal, muscle and adipose tissues and on the cardiovascular, immune, and nervous systems. Testosterone levels have also been studied in relation to the impact of diseases, aging, nutrition and the environment on its circulating levels. Conversely, the impact of testosterone on health has also been evaluated with respect to its cardiac and vascular protective effects, body composition, autoimmunity and all-cause mortality. The male aging process results in decreasing testosterone levels over time. The exact mechanisms and impact of these changes in testosterone levels with age on health- and life-span are still not completely clear. Further research is needed to determine the optimal testosterone and androgen levels to protect from chronic age-related conditions such as frailty and osteoporosis.
Collapse
Affiliation(s)
- Justin M. Ketchem
- grid.410427.40000 0001 2284 9329Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | | | - Carlos M. Isales
- grid.410427.40000 0001 2284 9329Departments of Medicine, Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912 USA
| |
Collapse
|
3
|
Kim D, Kiprov DD, Luellen C, Lieb M, Liu C, Watanabe E, Mei X, Cassaleto K, Kramer J, Conboy MJ, Conboy IM. Old plasma dilution reduces human biological age: a clinical study. GeroScience 2022; 44:2701-2720. [PMID: 35999337 PMCID: PMC9398900 DOI: 10.1007/s11357-022-00645-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/10/2022] [Indexed: 01/07/2023] Open
Abstract
This work extrapolates to humans the previous animal studies on blood heterochronicity and establishes a novel direct measurement of biological age. Our results support the hypothesis that, similar to mice, human aging is driven by age-imposed systemic molecular excess, the attenuation of which reverses biological age, defined in our work as a deregulation (noise) of 10 novel protein biomarkers. The results on biological age are strongly supported by the data, which demonstrates that rounds of therapeutic plasma exchange (TPE) promote a global shift to a younger systemic proteome, including youthfully restored pro-regenerative, anticancer, and apoptotic regulators and a youthful profile of myeloid/lymphoid markers in circulating cells, which have reduced cellular senescence and lower DNA damage. Mechanistically, the circulatory regulators of the JAK-STAT, MAPK, TGF-beta, NF-κB, and Toll-like receptor signaling pathways become more youthfully balanced through normalization of TLR4, which we define as a nodal point of this molecular rejuvenation. The significance of our findings is confirmed through big-data gene expression studies.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Connor Luellen
- Biophysics, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Michael Lieb
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Etsuko Watanabe
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Xiaoyue Mei
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Joel Kramer
- Brain Aging Center, UCSF, San Francisco, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
4
|
Qrareya AN, Mahdi F, Kaufman MJ, Ashpole NM, Paris JJ. Age-related neuroendocrine, cognitive, and behavioral co-morbidities are promoted by HIV-1 Tat expression in male mice. Aging (Albany NY) 2022; 14:5345-5365. [PMID: 35830469 PMCID: PMC9320553 DOI: 10.18632/aging.204166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
In the U.S. about half of the HIV-infected individuals are aged 50 and older. In men living with HIV, secondary hypogonadism is common and occurs earlier than in seronegative men, and its prevalence increases with age. While the mechanisms(s) are unknown, the HIV-1 trans-activator of transcription (Tat) protein disrupts neuroendocrine function in mice partly by dysregulating mitochondria and neurosteroidogenesis. We hypothesized that conditional Tat expression in middle-aged male transgenic mice [Tat(+)] would promote age-related comorbidities compared to age-matched controls [Tat(−)]. We expected Tat to alter steroid hormone milieu consistent with behavioral deficits. Middle-aged Tat(+) mice had lower circulating testosterone and progesterone than age-matched controls and greater circulating corticosterone and central allopregnanolone than other groups. Young Tat(+) mice had greater circulating progesterone and estradiol-to-testosterone ratios. Older age or Tat exposure increased anxiety-like behavior (open field; elevated plus-maze), increased cognitive errors (radial arm water maze), and reduced grip strength. Young Tat(+), or middle-aged Tat(−), males had higher mechanical nociceptive thresholds than age-matched counterparts. Steroid levels correlated with behaviors. Thus, Tat may contribute to HIV-accelerated aging.
Collapse
Affiliation(s)
- Alaa N Qrareya
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Fakhri Mahdi
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Marc J Kaufman
- Department of Psychiatry, McLean Imaging Center, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Nicole M Ashpole
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.,Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.,Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA
| |
Collapse
|
5
|
Souza-Teodoro LH, Andrade LHS, Carvalho LA. Could be dehydroepiandrosterone (DHEA) a novel target for depression? JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022. [DOI: 10.1016/j.jadr.2022.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
6
|
Salahuddin MF, Qrareya AN, Mahdi F, Moss E, Akins NS, Li J, Le HV, Paris JJ. Allopregnanolone and neuroHIV: Potential benefits of neuroendocrine modulation in the era of antiretroviral therapy. J Neuroendocrinol 2022; 34:e13047. [PMID: 34651359 PMCID: PMC8866218 DOI: 10.1111/jne.13047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
Forty years into the HIV pandemic, approximately 50% of infected individuals still suffer from a constellation of neurological disorders collectively known as 'neuroHIV.' Although combination antiretroviral therapy (cART) has been a tremendous success, in its present form, it cannot eradicate HIV. Reservoirs of virus reside within the central nervous system, serving as sources of HIV virotoxins that damage mitochondria and promote neurotoxicity. Although understudied, there is evidence that HIV or the HIV regulatory protein, trans-activator of transcription (Tat), can dysregulate neurosteroid formation potentially contributing to endocrine dysfunction. People living with HIV commonly suffer from endocrine disorders, including hypercortisolemia accompanied by paradoxical adrenal insufficiency upon stress. Age-related comorbidities often onset sooner and with greater magnitude among people living with HIV and are commonly accompanied by hypogonadism. In the post-cART era, these derangements of the hypothalamic-pituitary-adrenal and -gonadal axes are secondary (i.e., relegated to the brain) and indicative of neuroendocrine dysfunction. We review the clinical and preclinical evidence for neuroendocrine dysfunction in HIV, the capacity for hormone therapeutics to play an ameliorative role and the future steroid-based therapeutics that may have efficacy as novel adjunctives to cART.
Collapse
Affiliation(s)
- Mohammed F. Salahuddin
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Alaa N. Qrareya
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Fakhri Mahdi
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Emaya Moss
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Nicholas S. Akins
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Jing Li
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
- Research Institute of Pharmaceutical SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Hoang V. Le
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
- Research Institute of Pharmaceutical SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| | - Jason J. Paris
- Department of BioMolecular SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
- Research Institute of Pharmaceutical SciencesSchool of PharmacyUniversity of MississippiUniversityMSUSA
| |
Collapse
|
7
|
Tang J, Chen LR, Chen KH. The Utilization of Dehydroepiandrosterone as a Sexual Hormone Precursor in Premenopausal and Postmenopausal Women: An Overview. Pharmaceuticals (Basel) 2021; 15:46. [PMID: 35056103 PMCID: PMC8781653 DOI: 10.3390/ph15010046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 12/03/2022] Open
Abstract
Dehydroepiandrosterone (DHEA), and its metabolite, dehydroepiandrosterone sulfate ester (DHEAS), are the most abundant circulating steroid hormones, and are synthesized in the zona reticularis of the adrenal cortex, in the gonads, and in the brain. The precise physiological role of DHEA and DHEAS is not yet fully understood, but these steroid hormones can act as androgens, estrogens, and neurosteroids, and perform many roles in the human body. Since both levels decline with age, use of DHEA supplements have gained more attention due to being advertised as an antidote to aging in postmenopausal women, who may have concerns on age-related diseases and overall well-being. However, current research has not reached an overall consensus on the effects of DHEA on postmenopausal women. This overview is a summary of the current literature, addressing the metabolic pathway for DHEA synthesis and utilization, as well as the effects of DHEA on premenopausal and postmenopausal women with disease states and other factors. As for the therapeutic effects on menopausal syndrome and other age-related diseases, several studies have found that DHEA supplementations can alleviate vasomotor symptoms, preserve the integrity of the immune system, reduce bone loss, and increase muscle mass. Intravaginal DHEA has shown significant beneficial effects in menopausal women with severe vulvovaginal symptoms. On the other hand, DHEA supplements have not shown definitive effects in cardiovascular disease, adrenal insufficiency, insulin sensitivity, and cognition. Due to inadequate sample sizes and treatment durations of current studies, it is difficult to assess the safety and efficacy of DHEA and draw reliable conclusions for the physiological role, the optimal dosage, and the effects on premenopausal and postmenopausal women; therefore, the study of DHEA warrants future investigation. Further research into the roles of these steroid hormones may bring us closer to a therapeutic option in the future.
Collapse
Affiliation(s)
- Justine Tang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
8
|
Infection of Glia by Human Pegivirus Suppresses Peroxisomal and Antiviral Signaling Pathways. J Virol 2021; 95:e0107421. [PMID: 34524914 DOI: 10.1128/jvi.01074-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human pegivirus (HPgV) infects peripheral leukocytes but was recently shown to be a neurotropic virus associated with leukoencephalitis in humans. In the present study, we investigated the neural cell tropism of HPgV as well as its effects on host immune responses. HPgV wild type (WT) and a mutant virus with a deletion in the HPgV NS2 gene (ΔNS2) were able to productively infect human astrocytes and microglia but not neurons or an oligodendrocyte-derived cell line. Of note, the ΔNS2 virus replicated better than WT pegivirus in astrocytes, with both viruses being able to subsequently infect and spread in fresh human astrocyte cultures. Infection of human glia by HPgV WT and ΔNS2 viruses resulted in suppression of peroxisome-associated genes, including PEX11B, ABCD1, PEX7, ABCD3, PEX3, and PEX5L, during peak viral production, which was accompanied by reduced expression of IFNB, IRF3, IRF1, and MAVS, particularly in ΔNS2-infected cells. These data were consistent with analyses of brain tissue from patients infected with HPgV in which we observed suppression of peroxisome and type I interferon gene transcripts, including PEX11B, ABCD3, IRF1, and IRF3, with concurrent loss of PMP70 immunoreactivity in glia. Our data indicate that human astrocytes and microglia are permissive to HPgV infection, resulting in peroxisome injury and suppressed antiviral signaling that is influenced by viral diversity. IMPORTANCE Human pegiviruses are detected in 1 to 5% of the general population, principally infecting leukocytes, although their effects on human health remain uncertain. Here, we show that human pegivirus infects specific neural cell types in culture and human brain and, like other neurotropic flaviviruses, causes suppression of peroxisome and antiviral signaling pathways, which could favor ongoing viral infection and perhaps confer susceptibility to the development of neurological disease.
Collapse
|
9
|
Lavaee F, Ranjbar Z, Jalalian M, Amiri MA. Comparison of the sex hormones' serum level in women with recurrent aphthous stomatitis and healthy population: a cross-sectional study. BMC Oral Health 2021; 21:551. [PMID: 34706694 PMCID: PMC8549300 DOI: 10.1186/s12903-021-01812-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Background In this study, we aimed to evaluate the sex hormonal serum level in patients with recurrent aphthous stomatitis and compare them with healthy participants.
Methods This cross-sectional study was done on patients with recurrent aphthous stomatitis who had referred to Shiraz Dental Faculty, Oral and Maxillofacial Medicine Department during 2018–2019. The non -menopause women with recurrence of at least 3 lesions per year were enrolled in this study. The mean serum level of FSH, LH, PRL (prolactin), testosterone, DHT (Dihydrotestosterone), DHEA-S (Dehydroepiandrosterone sulfate), estradiol and progesterone of 30 participants in each group of case and control were measured and compared. The data were analyzed by SPSS version 18 and independent T-test, Mann–Whitney U test, Spearman’s correlation coefficient test, Chi-square test and Fisher’s test. Results The mean serum level of DHEA-S in patients with recurrent aphthous stomatitis (RAS) was significantly lower than the control group (p value = 0.02). In addition to DHEA-S, the mean serum level of testosterone was lower in the evaluation group although this difference was not significant (p value = 0.057). Considering the effect of age on the mean serum level of sex hormones, our results revealed that only DHEA-S mean serum level was decreased by increasing the age of participants in patients with RAS (p value = 0.018). The number of participants with abnormal range of testosterone (p value < 0.0001) and progesterone (p value = 0.037) serum level was significantly more in patients with RAS. The frequency of RAS in a year did not show a significant relationship with the serum level of the evaluated hormones. Conclusion The patients with RAS had a lower serum level of DHEA-S. The mean serum level of testosterone and progesterone was significantly abnormal in RAS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-021-01812-9.
Collapse
Affiliation(s)
- Fatemeh Lavaee
- Oral and Dental Disease Research Center, Oral and Maxillofacial Disease Department, Shiraz University of Medical Sciences, Ghasr-dasht Street, Shiraz, Iran
| | - Zahra Ranjbar
- Oral and Dental Disease Research Center, Oral and Maxillofacial Disease Department, Shiraz University of Medical Sciences, Ghasr-dasht Street, Shiraz, Iran.
| | - Mina Jalalian
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Amiri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Li H, McLaurin KA, Illenberger JM, Mactutus CF, Booze RM. Microglial HIV-1 Expression: Role in HIV-1 Associated Neurocognitive Disorders. Viruses 2021; 13:924. [PMID: 34067600 PMCID: PMC8155894 DOI: 10.3390/v13050924] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022] Open
Abstract
The persistence of HIV-1 viral reservoirs in the brain, despite treatment with combination antiretroviral therapy (cART), remains a critical roadblock for the development of a novel cure strategy for HIV-1. To enhance our understanding of viral reservoirs, two complementary studies were conducted to (1) evaluate the HIV-1 mRNA distribution pattern and major cell type expressing HIV-1 mRNA in the HIV-1 transgenic (Tg) rat, and (2) validate our findings by developing and critically testing a novel biological system to model active HIV-1 infection in the rat. First, a restricted, region-specific HIV-1 mRNA distribution pattern was observed in the HIV-1 Tg rat. Microglia were the predominant cell type expressing HIV-1 mRNA in the HIV-1 Tg rat. Second, we developed and critically tested a novel biological system to model key aspects of HIV-1 by infusing F344/N control rats with chimeric HIV (EcoHIV). In vitro, primary cultured microglia were treated with EcoHIV revealing prominent expression within 24 h of infection. In vivo, EcoHIV expression was observed seven days after stereotaxic injections. Following EcoHIV infection, microglia were the major cell type expressing HIV-1 mRNA, results that are consistent with observations in the HIV-1 Tg rat. Within eight weeks of infection, EcoHIV rats exhibited neurocognitive impairments and synaptic dysfunction, which may result from activation of the NogoA-NgR3/PirB-RhoA signaling pathway and/or neuroinflammation. Collectively, these studies enhance our understanding of HIV-1 viral reservoirs in the brain and offer a novel biological system to model HIV-associated neurocognitive disorders and associated comorbidities (i.e., drug abuse) in rats.
Collapse
Affiliation(s)
| | | | | | | | - Rosemarie M. Booze
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (H.L.); (K.A.M.); (J.M.I.); (C.F.M.)
| |
Collapse
|
11
|
Gabai G, Mongillo P, Giaretta E, Marinelli L. Do Dehydroepiandrosterone (DHEA) and Its Sulfate (DHEAS) Play a Role in the Stress Response in Domestic Animals? Front Vet Sci 2020; 7:588835. [PMID: 33195624 PMCID: PMC7649144 DOI: 10.3389/fvets.2020.588835] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
In animal husbandry, stress is often associated with poor health and welfare. Stress occurs when a physiological control system detects a state of real or presumptive threat to the animal's homeostasis or a failure to control a fitness-critical variable. The definition of stress has mostly relied on glucocorticoids measurement, even though glucocorticoids represent one stress-response system, the hypothalamus-pituitary-adrenocortical axis, which is not precise enough as it is also related to metabolic regulation and activated in non-stressful situations (pleasure, excitement, and arousal). The mammal adrenal can synthesize the androgenic steroid dehydroepiandrosterone (DHEA) and its sulfate metabolite (DHEAS), which have been associated to the stress response in several studies performed mostly in humans and laboratory animals. Although the functions of these steroids are not fully understood, available data suggest their antagonistic effects on glucocorticoids and, in humans, their secretion is affected by stress. This review explores the scientific literature on DHEA and DHEAS release in domestic animals in response to stressors of different nature (inflammatory, physical, or social) and duration, and the extra-adrenal contribution to circulating DHEA. Then, the potential use of DHEA in conjunction with cortisol to improve the definition of the stress phenotype in farmed animals is discussed. Although the focus of this review is on farmed animals, examples from other species are reported when available.
Collapse
Affiliation(s)
- Gianfranco Gabai
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Paolo Mongillo
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Elisa Giaretta
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Lieta Marinelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| |
Collapse
|
12
|
Almagro L, Lemos R, Makowski K, Rodríguez H, Ortiz O, Cáceres W, Herranz MÁ, Molero D, Martínez‐Álvarez R, Suárez M, Martín N. [60]Fullerene Hybrids Bearing “Steroid Wings”: A Joined Experimental and Theoretical Investigation. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Luis Almagro
- Laboratorio de Síntesis Orgánica Facultad de Química Universidad de la Habana 10400 La Habana Cuba
| | - Reinier Lemos
- Laboratorio de Síntesis Orgánica Facultad de Química Universidad de la Habana 10400 La Habana Cuba
| | - Kamil Makowski
- School of Chemical Sciences and Engineering Yachay Tech University 100119 Urququi Ecuador
| | - Hortensia Rodríguez
- School of Chemical Sciences and Engineering Yachay Tech University 100119 Urququi Ecuador
| | - Orlando Ortiz
- Laboratorio de Síntesis Orgánica Facultad de Química Universidad de la Habana 10400 La Habana Cuba
| | - William Cáceres
- Laboratorio de Síntesis Orgánica Facultad de Química Universidad de la Habana 10400 La Habana Cuba
| | - M. Ángeles Herranz
- Departamento de Química Orgánica Facultad de Ciencias Químicas Universidad Complutense de Madrid 28040 Madrid Spain
| | - Dolores Molero
- CAI RMN Universidad Complutense de Madrid 28040 Madrid Spain
| | - Roberto Martínez‐Álvarez
- Departamento de Química Orgánica Facultad de Ciencias Químicas Universidad Complutense de Madrid 28040 Madrid Spain
| | - Margarita Suárez
- Laboratorio de Síntesis Orgánica Facultad de Química Universidad de la Habana 10400 La Habana Cuba
| | - Nazario Martín
- Departamento de Química Orgánica Facultad de Ciencias Químicas Universidad Complutense de Madrid 28040 Madrid Spain
| |
Collapse
|
13
|
Pregnane steroidogenesis is altered by HIV-1 Tat and morphine: Physiological allopregnanolone is protective against neurotoxic and psychomotor effects. Neurobiol Stress 2020; 12:100211. [PMID: 32258256 PMCID: PMC7109513 DOI: 10.1016/j.ynstr.2020.100211] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/08/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pregnane steroids, particularly allopregnanolone (AlloP), are neuroprotective in response to central insult. While unexplored in vivo, AlloP may confer protection against the neurological dysfunction associated with human immunodeficiency virus type 1 (HIV-1). The HIV-1 regulatory protein, trans-activator of transcription (Tat), is neurotoxic and its expression in mice increases anxiety-like behavior; an effect that can be ameliorated by progesterone, but not when 5α-reduction is blocked. Given that Tat's neurotoxic effects involve mitochondrial dysfunction and can be worsened with opioid exposure, we hypothesized that Tat and/or combined morphine would perturb steroidogenesis in mice, promoting neuronal death, and that exogenous AlloP would rescue these effects. Like other models of neural injury, conditionally inducing HIV-1 Tat in transgenic mice significantly increased the central synthesis of pregnenolone and progesterone's 5α-reduced metabolites, including AlloP, while decreasing central deoxycorticosterone (independent of changes in plasma). Morphine significantly increased brain and plasma concentrations of several steroids (including progesterone, deoxycorticosterone, corticosterone, and their metabolites) likely via activation of the hypothalamic-pituitary-adrenal stress axis. Tat, but not morphine, caused glucocorticoid resistance in primary splenocytes. In neurons, Tat depolarized mitochondrial membrane potential and increased cell death. Physiological concentrations of AlloP (0.1, 1, or 10 nM) reversed these effects. High-concentration AlloP (100 nM) was neurotoxic in combination with morphine. Tat induction in transgenic mice potentiated the psychomotor effects of acute morphine, while exogenous AlloP (1.0 mg/kg, but not 0.5 mg/kg) was ameliorative. Data demonstrate that steroidogenesis is altered by HIV-1 Tat or morphine and that physiological AlloP attenuates resulting neurotoxic and psychomotor effects.
Collapse
|
14
|
Powrie YSL, Smith C. Central intracrine DHEA synthesis in ageing-related neuroinflammation and neurodegeneration: therapeutic potential? J Neuroinflammation 2018; 15:289. [PMID: 30326923 PMCID: PMC6192186 DOI: 10.1186/s12974-018-1324-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023] Open
Abstract
It is a well-known fact that DHEA declines on ageing and that it is linked to ageing-related neurodegeneration, which is characterised by gradual cognitive decline. Although DHEA is also associated with inflammation in the periphery, the link between DHEA and neuroinflammation in this context is less clear. This review drew from different bodies of literature to provide a more comprehensive picture of peripheral vs central endocrine shifts with advanced age—specifically in terms of DHEA. From this, we have formulated the hypothesis that DHEA decline is also linked to neuroinflammation and that increased localised availability of DHEA may have both therapeutic and preventative benefit to limit neurodegeneration. We provide a comprehensive discussion of literature on the potential for extragonadal DHEA synthesis by neuroglial cells and reflect on the feasibility of therapeutic manipulation of localised, central DHEA synthesis.
Collapse
Affiliation(s)
- Y S L Powrie
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa
| | - C Smith
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
15
|
Quinn T, Greaves R, Badoer E, Walker D. DHEA in Prenatal and Postnatal Life: Implications for Brain and Behavior. VITAMINS AND HORMONES 2018; 108:145-174. [PMID: 30029725 DOI: 10.1016/bs.vh.2018.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dehydroepiandrosterone (DHEA) and its sulfated congener (DHEAS) are the principal C19 steroid produced by the adrenal gland in many mammals, including humans. It is secreted in high concentrations during fetal life, but synthesis decreases after birth until, in humans and some other primates, there is a prepubertal surge of DHEA production by the adrenal gland-a phenomenon known as adrenarche. There remains considerable uncertainty about the physiological role of DHEA and DHEAS. Moreover, the origin of the trophic drives that determine the waxing and waning of DHEA synthesis are poorly understood. These gaps in knowledge arise in some measure from the difficulty of understanding mechanistic determinants from observations made opportunistically in humans and primates, and have stimulated a search for other suitable species that exhibit adrenarche- and adrenopause-like changes of adrenal function. DHEA and DHEAS are clearly neuroactive steroids with actions at several neurotransmitter receptors; indeed, DHEA is now known to be also synthesized by many parts of the brain, and this capacity undergoes ontogenic changes, but whether this is dependent or independent of the changes in adrenal synthesis is unknown. In this chapter we review key contributions to this field over the last 50+ years, and speculate on the importance of DHEA for the brain, both during development and for maturation and aging of cerebral function and behavior.
Collapse
Affiliation(s)
- Tracey Quinn
- The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, VIC, Australia
| | - Ronda Greaves
- School of Health & Biomedical Sciences, RMIT University-Bundoora Campus, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
| | - Emilio Badoer
- School of Health & Biomedical Sciences, RMIT University-Bundoora Campus, Melbourne, VIC, Australia
| | - David Walker
- School of Health & Biomedical Sciences, RMIT University-Bundoora Campus, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Power C. Neurologic disease in feline immunodeficiency virus infection: disease mechanisms and therapeutic interventions for NeuroAIDS. J Neurovirol 2017; 24:220-228. [PMID: 29247305 DOI: 10.1007/s13365-017-0593-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022]
Abstract
Feline immunodeficiency virus (FIV) is a lentivirus that causes immunosuppression through virus-mediated CD4+ T cell depletion in feline species. FIV infection is complicated by virus-induced disease in the nervous system. FIV enters the brain soon after primary infection and is detected as FIV-encoded RNA, DNA, and proteins in microglia, macrophages, and astrocytes. FIV infection activates neuroinflammatory pathways including cytokines, chemokines, proteases, and ROS with accompanying neuronal injury and loss. Neurobehavioral deficits during FIV infection are manifested as impaired motor and cognitive functions. Several treatment strategies have emerged from studies of FIV neuropathogenesis including the therapeutic benefits of antiretroviral therapies, other protease inhibitors, anti-inflammatory, and neurotrophic compounds. Recently, insulin's antiviral, anti-inflammatory, and neuroprotective effects were investigated in models of lentivirus brain infection. Insulin suppressed HIV-1 replication in human microglia as well as FIV replication of lymphocytes. Insulin treatment diminished cytokine and chemokine activation in HIV-infected microglia while also protecting neurons from HIV-1 Vpr protein-mediated neurotoxicity. Intranasal (IN) insulin delivery for 6 weeks suppressed FIV expression in the brains of treated cats. IN insulin also reduced neuroinflammation and protected neurons in the hippocampus, striatum, and neocortex of FIV-infected animals. These morphological and molecular effects of IN insulin were confirmed by neurobehavioral studies that showed IN insulin-treated FIV-infected animals displayed improved motor and cognitive performance compared to sham-treated FIV-infected animals. Thus, FIV infection of the nervous system provides a valuable comparative in vivo model for discovering and evaluating disease mechanisms as well as developing therapeutic strategies for NeuroAIDS in humans.
Collapse
Affiliation(s)
- Christopher Power
- Department of Medicine (Neurology) and the Neuroscience and Mental Health Institute, University of Alberta, HMRC 6-11, Edmonton, AB, Canada.
| |
Collapse
|
17
|
Tuem KB, Atey TM. Neuroactive Steroids: Receptor Interactions and Responses. Front Neurol 2017; 8:442. [PMID: 28894435 PMCID: PMC5581316 DOI: 10.3389/fneur.2017.00442] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022] Open
Abstract
Neuroactive steroids (NASs) are naturally occurring steroids, which are synthesized centrally as de novo from cholesterol and are classified as pregnane, androstane, and sulfated neurosteroids (NSs). NASs modulate many processes via interacting with gamma-aminobutyric acid (GABA), N-methyl-d-aspartate, serotonin, voltage-gated calcium channels, voltage-dependent anion channels, α-adrenoreceptors, X-receptors of the liver, transient receptor potential channels, microtubule-associated protein 2, neurotrophin nerve growth factor, and σ1 receptors. Among these, NSs (especially allopregnanolone) have high potency and extensive GABA-A receptors and hence demonstrate anticonvulsant, anesthetic, central cytoprotectant, and baroreflex inhibitory effects. NSs are also involved in mood and learning via serotonin and anti-nociceptive activity via T-type voltage-gated Ca2+ channels. Moreover, they are modulators of mitochondrial function, synaptic plasticity, or regulators of apoptosis, which have a role in neuroprotective via voltage-dependent anion channels receptors. For proper functioning, NASs need to be in their normal level, whereas excess and deficiency may lead to abnormalities. When they are below the normal, NSs could have a part in development of depression, neuro-inflammation, multiple sclerosis, experimental autoimmune encephalitis, epilepsy, and schizophrenia. On the other hand, stress and attention deficit disorder could occur during excessive level. Overall, NASs are very important molecules with major neuropsychiatric activity.
Collapse
Affiliation(s)
- Kald Beshir Tuem
- Department of Pharmacology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Tesfay Mehari Atey
- Clinical Pharmacy Unit, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
18
|
Boghozian R, McKenzie BA, Saito LB, Mehta N, Branton WG, Lu J, Baker GB, Noorbakhsh F, Power C. Suppressed oligodendrocyte steroidogenesis in multiple sclerosis: Implications for regulation of neuroinflammation. Glia 2017; 65:1590-1606. [PMID: 28707358 DOI: 10.1002/glia.23179] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Neurosteroids are reported to exert anti-inflammatory effects in several neurological disorders. We investigated the expression and actions of the neurosteroid, dehydroepiandrosterone (DHEA), and its more stable 3β-sulphated ester, DHEA-S, in MS and associated experimental models. CNS tissues from patients with MS and animals with experimental autoimmune encephalomyelitis (EAE) displayed reduced DHEA concentrations, accompanied by diminished expression of the DHEA-synthesizing enzyme CYP17A1 in oligodendrocytes (ODCs), in association with increased expression of inflammatory genes including interferon (IFN)-γ and interleukin (IL)-1β. CYP17A1 was expressed variably in different human neural cell types but IFN-γ exposure selectively reduced CYP17A1 detection in ODCs. DHEA-S treatment reduced IL-1β and -6 release from activated human myeloid cells with minimal effect on lymphocyte viability. Animals with EAE receiving DHEA-S treatment showed reduced Il1b and Ifng transcript levels in spinal cord compared to vehicle-treated animals with EAE. DHEA-S treatment also preserved myelin basic protein immunoreactivity and reduced axonal loss in animals with EAE, relative to vehicle-treated EAE animals. Neurobehavioral deficits were reduced in DHEA-S-treated EAE animals compared with vehicle-treated animals with EAE. Thus, CYP17A1 expression in ODCs and its product DHEA were downregulated in the CNS during inflammatory demyelination while DHEA-S provision suppressed neuroinflammation, demyelination, and axonal injury that was evident as improved neurobehavioral performance. These findings indicate that DHEA production is an immunoregulatory pathway within the CNS and its restoration represents a novel treatment approach for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Roobina Boghozian
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada.,Department of Medical Microbiology & Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Brienne A McKenzie
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - Leina B Saito
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - Ninad Mehta
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - William G Branton
- Department of, Medicine, University of Alberta Edmonton, Alberta, Canada
| | - JianQiang Lu
- Department of Laboratory Medicine & Pathology, University of Alberta Edmonton, Alberta, Canada
| | - Glen B Baker
- Depatment of Psychiatry, University of Alberta Edmonton, Alberta, Canada
| | - Farshid Noorbakhsh
- Department of Medical Microbiology & Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada.,Department of, Medicine, University of Alberta Edmonton, Alberta, Canada.,Depatment of Psychiatry, University of Alberta Edmonton, Alberta, Canada
| |
Collapse
|
19
|
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2017; 38:1126-1158. [PMID: 28697282 DOI: 10.1002/med.21458] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Neurodegeneration is a complex process, which leads to progressive brain damage due to loss of neurons. Despite exhaustive research, the cause of neuronal loss in various degenerative disorders is not entirely understood. Neuroprotective steroids constitute an important line of attack, which could play a major role against the common mechanisms associated with various neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Natural endogenous steroids induce the neuroprotection by protecting the nerve cells from neuronal injury through multiple mechanisms, therefore the structural modifications of the endogenous steroids could be helpful in the generation of new therapeutically useful neuroprotective agents. The review article will keep the readers apprised of the detailed description of natural as well as synthetic neuroprotective steroids from the medicinal chemistry point of view, which would be helpful in drug discovery efforts aimed toward neurodegenerative diseases.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
20
|
HIV-1 Tat Primes and Activates Microglial NLRP3 Inflammasome-Mediated Neuroinflammation. J Neurosci 2017; 37:3599-3609. [PMID: 28270571 DOI: 10.1523/jneurosci.3045-16.2017] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/30/2017] [Accepted: 02/24/2017] [Indexed: 11/21/2022] Open
Abstract
Neuroinflammation associated with HIV-1 infection is a problem affecting ∼50% of HIV-infected individuals. NLR family pyrin domain containing 3 (NLRP3) inflammasome has been implicated in HIV-induced microglial activation, but the mechanism(s) remain unclear. Because HIV-1 Transactivator of Transcription (Tat) protein continues to be present despite antiretroviral therapy and activates NF-kB, we hypothesized that Tat could prime the NLRP3 inflammasome. We found a dose- and time-dependent induction of NLRP3 expression in microglia exposed to Tat compared with control. Tat exposure also time-dependently increased the mature caspase-1 and IL-1β levels and enhanced the IL-1β secretion. These in vitro findings were validated in archival brain tissues from Simian Immunodeficiency Virus (SIV)-infected and uninfected rhesus macaques. Further validation of NLRP3 priming in vivo involved administration of lipopolysaccharide (LPS) to HIV transgenic (Tg) rats followed by assessment of IL-1β mRNA expression and inflammasome activation (ASC oligomers and mature IL-1β). Intriguingly, LPS potentiated upregulation of IL-1β mRNA and inflammasome activation in HIV-Tg rats compared with the wild-type controls. Interestingly, we found an inverse relationship in the expression of NLRP3 and its negative regulator, miR-223, suggesting a miR-223-mediated mechanism for Tat-induced NLRP3 priming. Furthermore, blockade of NLRP3 resulted in decreased IL-1β secretion. Collectively, these findings suggest a novel role of Tat in priming and activating the NLRP3 inflammasome. Therefore, NLRP3 can be envisioned as a therapeutic target for ameliorating Tat-mediated neuroinflammation.SIGNIFICANCE STATEMENT Despite successful suppression of viremia with increased longevity in the era of combined antiretroviral therapy, chronic inflammation with underlying neurocognitive impairment continues to afflict almost 50% of infected individuals. Viral, bacterial, and cellular products have all been implicated in promoting the chronic inflammation found in these individuals. Understanding the molecular mechanism(s) by which viral proteins such as HIV-1 Transactivator of Transcription (Tat) protein can activate microglia is thus of paramount importance. Herein, we demonstrate a novel role of Tat in priming and activating NLR family pyrin domain containing 3 (NLRP3) inflammasomes in microglial cells and in HIV-Tg rats administered lipopolysaccharide. Targeting NLRP3 inflammasome pathway mediators could thus be developed as therapeutic interventions to alleviate or prevent neuroinflammation and subsequent cognitive impairment in HIV-positive patients.
Collapse
|
21
|
Feline Immunodeficiency Virus Neuropathogenesis: A Model for HIV-Induced CNS Inflammation and Neurodegeneration. Vet Sci 2017; 4:vetsci4010014. [PMID: 29056673 PMCID: PMC5606611 DOI: 10.3390/vetsci4010014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
Feline Immunodeficiency virus (FIV), similar to its human analog human immunodeficiency virus (HIV), enters the central nervous system (CNS) soon after infection and establishes a protected viral reservoir. The ensuing inflammation and damage give rise to varying degrees of cognitive decline collectively known as HIV-associated neurocognitive disorders (HAND). Because of the similarities to HIV infection and disease, FIV has provided a useful model for both in vitro and in vivo studies of CNS infection, inflammation and pathology. This mini review summarizes insights gained from studies of early infection, immune cell trafficking, inflammation and the mechanisms of neuropathogenesis. Advances in our understanding of these processes have contributed to the development of therapeutic interventions designed to protect neurons and regulate inflammatory activity.
Collapse
|
22
|
5α-reduced progestogens ameliorate mood-related behavioral pathology, neurotoxicity, and microgliosis associated with exposure to HIV-1 Tat. Brain Behav Immun 2016; 55:202-214. [PMID: 26774528 PMCID: PMC4899138 DOI: 10.1016/j.bbi.2016.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 02/05/2023] Open
Abstract
Human immunodeficiency virus (HIV) is associated with motor and mood disorders, likely influenced by reactive microgliosis and subsequent neural damage. We have recapitulated aspects of this pathology in mice that conditionally express the neurotoxic HIV-1 regulatory protein, trans-activator of transcription (Tat). Progestogens may attenuate Tat-related behavioral impairments and reduce neurotoxicity in vitro, perhaps via progesterone's 5α-reductase-dependent metabolism to the neuroprotective steroid, allopregnanolone. To test this, ovariectomized female mice that conditionally expressed (or did not express) central HIV-1 Tat were administered vehicle or progesterone (4mg/kg), with or without pretreatment of a 5α-reductase inhibitor (finasteride, 50mg/kg). Tat induction significantly increased anxiety-like behavior in an open field, elevated plus maze and a marble burying task concomitant with elevated protein oxidation in striatum. Progesterone administration attenuated anxiety-like effects in the open field and elevated plus maze, but not in conjunction with finasteride pretreatment. Progesterone also attenuated Tat-promoted protein oxidation in striatum, independent of finasteride pretreatment. Concurrent experiments in vitro revealed Tat (50nM)-mediated reductions in neuronal cell survival over 60h, as well as increased neuronal and microglial intracellular calcium, as assessed via fura-2 AM fluorescence. Co-treatment with allopregnanolone (100nM) attenuated neuronal death in time-lapse imaging and blocked the Tat-induced exacerbation of intracellular calcium in neurons and microglia. Lastly, neuronal-glial co-cultures were labeled for Iba-1 to reveal that Tat increased microglial numbers in vitro and co-treatment with allopregnanolone attenuated this effect. Together, these data support the notion that 5α-reduced pregnane steroids exert protection over the neurotoxic effects of HIV-1 Tat.
Collapse
|
23
|
Abstract
Objective: The neurotoxic actions of the HIV protease inhibitors, amprenavir (APV) and lopinavir (LPV) were investigated. Design: With combination antiretroviral therapy (cART), HIV-infected persons exhibit neurocognitive impairments, raising the possibility that cART might exert adverse central nervous system (CNS) effects. We examined the effects of LPV and APV using in-vitro and in-vivo assays of CNS function. Methods: Gene expression, cell viability and amino-acid levels were measured in human astrocytes, following exposure to APV or LPV. Neurobehavioral performance, amino-acid levels and neuropathology were examined in HIV-1 Vpr transgenic mice after treatment with APV or LPV. Results: Excitatory amino-acid transporter-2 (EAAT2) expression was reduced in astrocytes treated with LPV or APV, especially LPV (P < 0.05), which was accompanied by reduced intracellular l-glutamate levels in LPV-treated cells (P < 0.05). Treatment of astrocytes with APV or LPV reduced the expression of proliferating cell nuclear antigen (PCNA) and Ki-67 (P < 0.05) although cell survival was unaffected. Exposure of LPV to astrocytes augmented glutamate-evoked transient rises in [Cai] (P < 0.05). Vpr mice treated with LPV showed lower concentrations of l-glutamate, l-aspartate and l-serine in cortex compared with vehicle-treated mice (P < 0.05). Total errors in T-maze assessment were increased in LPV and APV-treated animals (P < 0.05). EAAT2 expression was reduced in the brains of protease inhibitor-treated animals, which was associated with gliosis (P < 0.05). Conclusion: These results indicated that contemporary protease inhibitors disrupt astrocyte functions at therapeutic concentrations with enhanced sensitivity to glutamate, which can lead to neurobehavioral impairments. ART neurotoxicity should be considered in future therapeutic regimens for HIV/AIDS.
Collapse
|
24
|
Höllig A, Thiel M, Stoffel-Wagner B, Coburn M, Clusmann H. Neuroprotective properties of dehydroepiandrosterone-sulfate and its relationship to interleukin 6 after aneurysmal subarachnoid hemorrhage: a prospective cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:231. [PMID: 25993987 PMCID: PMC4462180 DOI: 10.1186/s13054-015-0954-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/15/2015] [Indexed: 11/10/2022]
Abstract
INTRODUCTION The established neuroprotective property of the sex steroid precursor dehydroepiandrosterone-sulfate (DHEAS) has not yet been investigated in the context of aneurysmal subarachnoid hemorrhage (aSAH). The influence of DHEAS on inflammatory response resulting in modulation of interleukin 6 (IL-6) synthesis has been shown. Here, we evaluate DHEAS serum levels after aSAH (day 0-14) and levels of IL-6 related to functional outcome at discharge and at six months. METHODS A complete data set (DHEAS and IL-6 serum levels for days 0, 1, 4, 7, 10 and 14 after aSAH) and outcome assessment at discharge according to modified Rankin Scale score (mRS) was available for 53 patients of the initially screened cohort (n = 109). Outcome assessment six months after aSAH was obtained from 41 patients. Logarithmized levels of DHEAS and IL-6 were related to dichotomized functional outcome either assessed at discharge or at six months. A mixed between-within subjects ANOVA was applied for statistical analysis (SPSS 21.0). RESULTS DHEAS and IL-6 levels across time were related to functional outcome. Regarding outcome assessment at discharge and at six months after aSAH, DHEAS levels (transformed to square root for statistical purposes) were considerably higher in patients with favorable outcome (mRS 0-2) (p = .001; p = .020). Inversely, in patients with favorable outcome either at discharge or six months after aSAH, lower IL-6 levels (logarithmized for statistical purposes) were observed across time (both p < .001). CONCLUSION We provide new evidence that DHEAS is associated with protective properties resulting in improvement of functional outcome after aSAH, possibly by influencing the inflammatory response after aSAH shown in the decreasing IL-6 serum levels. But the results for outcome six months after SAH are limited due to a high drop-out rate.
Collapse
Affiliation(s)
- Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Pauwelsstr. 30, D-52074, Aachen, Germany. .,Department of Anesthesiology, RWTH Aachen University, Aachen, Germany.
| | - Miriam Thiel
- Department of Pediatrics, GFO Kliniken Bonn Zweigstelle St. Marien-Hospital, Bonn, Germany.
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany.
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University, Aachen, Germany.
| | - Hans Clusmann
- Department of Neurosurgery, RWTH Aachen University, Pauwelsstr. 30, D-52074, Aachen, Germany.
| |
Collapse
|
25
|
Chrousos GP, Zapanti ED. Hypothalamic-pituitary-adrenal axis in HIV infection and disease. Endocrinol Metab Clin North Am 2014; 43:791-806. [PMID: 25169568 DOI: 10.1016/j.ecl.2014.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HIV infection induces hypothalamic-pituitary-adrenal (HPA) axis derangements. Partial glucocorticoid resistance has been observed in a subset of AIDS patients, possibly owing to HIV-induced altered cytokine secretion and action. Because glucocorticoids have immunomodulatory effects, the severity of the HPA axis disorder could play a central role in disease progression. The characteristic phenotype of AIDS patients (visceral obesity, lipodystrophy) may be owing to effects of HIV proteins on the HPA axis, including changes in glucocorticoid and insulin sensitivity of target tissues, as well as altered cytokine production and interaction with the HPA axis, genetic causes, comorbidities, and, possibly, use of antiretroviral agents.
Collapse
Affiliation(s)
- George P Chrousos
- First Department of Pediatrics, "Agia Sofia" Children's Hospital, University of Athens, Thivon and Papadiamantopoulou, Athens 11527, Greece
| | - Evangelia D Zapanti
- First Endocrine Department and Diabetes Center, Alexandra Hospital, 80 Vassilisis Sofias Avenue, Athens 11528, Greece.
| |
Collapse
|
26
|
Ashraf T, Jiang W, Hoque MT, Henderson J, Wu C, Bendayan R. Role of anti-inflammatory compounds in human immunodeficiency virus-1 glycoprotein120-mediated brain inflammation. J Neuroinflammation 2014; 11:91. [PMID: 24884548 PMCID: PMC4046047 DOI: 10.1186/1742-2094-11-91] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 04/15/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Neuroinflammation is a common immune response associated with brain human immunodeficiency virus-1 (HIV-1) infection. Identifying therapeutic compounds that exhibit better brain permeability and can target signaling pathways involved in inflammation may benefit treatment of HIV-associated neurological complications. The objective of this study was to implement an in vivo model of brain inflammation by intracerebroventricular administration of the HIV-1 viral coat protein gp120 in rats and to examine anti-inflammatory properties of HIV adjuvant therapies such as minocycline, chloroquine and simvastatin. METHODS Male Wistar rats were administered a single dose of gp120ADA (500 ng) daily for seven consecutive days, intracerebroventricularly, with or without prior intraperitoneal administration of minocycline, chloroquine or simvastatin. Maraviroc, a CCR5 antagonist, was administered intracerebroventricularly prior to gp120 administration for seven days as control. Real-time qPCR was used to assess gene expression of inflammatory markers in the frontal cortex, hippocampus and striatum. Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) secretion in cerebrospinal fluid (CSF) was measured applying ELISA. Protein expression of mitogen-activated protein kinases (MAPKs) (extracellular signal-related kinase 1/2 (ERK1/2), c-Jun N-terminal kinases (JNKs) and P38 kinases (P38Ks)) was detected using immunoblot analysis. Student's t-test and ANOVA were applied to determine statistical significance. RESULTS In gp120ADA-injected rats, mRNA transcripts of interleukin-1β (IL-1β) and inducible nitric oxide synthase (iNOS) were significantly elevated in the frontal cortex, striatum and hippocampus compared to saline or heat-inactivated gp120-injected controls. In CSF, a significant increase in TNF-α and IL-1β was detected. Maraviroc reduced upregulation of these markers suggesting that the interaction of R5-tropic gp120 to CCR5 chemokine receptor is critical for induction of an inflammatory response. Minocycline, chloroquine or simvastatin attenuated upregulation of IL-1β and iNOS transcripts in different brain regions. In CSF, minocycline suppressed TNF-α and IL-1β secretion, whereas chloroquine attenuated IL-1β secretion. In gp120-injected animals, activation of ERK1/2 and JNKs was observed in the hippocampus and ERK1/2 activation was significantly reduced by the anti-inflammatory agents. CONCLUSIONS Our data demonstrate that anti-inflammatory compounds can completely or partially reverse gp120-associated brain inflammation through an interaction with MAPK signaling pathways and suggest their potential role in contributing towards the prevention and treatment of HIV-associated neurological complications.
Collapse
Affiliation(s)
- Tamima Ashraf
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| | - Wenlei Jiang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| | - Md Tozammel Hoque
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| | - Jeffrey Henderson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| | - Chiping Wu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Room 1001, Toronto, ON M5S 3 M2, Canada
| |
Collapse
|
27
|
Walsh JG, Reinke SN, Mamik MK, McKenzie BA, Maingat F, Branton WG, Broadhurst DI, Power C. Rapid inflammasome activation in microglia contributes to brain disease in HIV/AIDS. Retrovirology 2014; 11:35. [PMID: 24886384 PMCID: PMC4038111 DOI: 10.1186/1742-4690-11-35] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 04/24/2014] [Indexed: 01/18/2023] Open
Abstract
Background Human immunodeficiency virus type 1(HIV-1) infects and activates innate immune cells in the brain resulting in inflammation and neuronal death with accompanying neurological deficits. Induction of inflammasomes causes cleavage and release of IL-1β and IL-18, representing pathogenic processes that underlie inflammatory diseases although their contribution HIV-associated brain disease is unknown. Results Investigation of inflammasome-associated genes revealed that IL-1β, IL-18 and caspase-1 were induced in brains of HIV-infected persons and detected in brain microglial cells. HIV-1 infection induced pro-IL-1β in human microglia at 4 hr post-infection with peak IL-1β release at 24 hr, which was accompanied by intracellular ASC translocation and caspase-1 activation. HIV-dependent release of IL-1β from a human macrophage cell line, THP-1, was inhibited by NLRP3 deficiency and high extracellular [K+]. Exposure of microglia to HIV-1 gp120 caused IL-1β production and similarly, HIV-1 envelope pseudotyped viral particles induced IL-1β release, unlike VSV-G pseudotyped particles. Infection of cultured feline macrophages by the related lentivirus, feline immunodeficiency virus (FIV), also resulted in the prompt induction of IL-1β. In vivo FIV infection activated multiple inflammasome-associated genes in microglia, which was accompanied by neuronal loss in cerebral cortex and neurological deficits. Multivariate analyses of data from FIV-infected and uninfected animals disclosed that IL-1β, NLRP3 and caspase-1 expression in cerebral cortex represented key molecular determinants of neurological deficits. Conclusions NLRP3 inflammasome activation was an early and integral aspect of lentivirus infection of microglia, which was associated with lentivirus-induced brain disease. Inflammasome activation in the brain might represent a potential target for therapeutic interventions in HIV/AIDS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher Power
- Department of Medicine (Neurology), Heritage Medical Research Centre 6-11, University of Alberta, Edmonton T6G 2S2, Canada.
| |
Collapse
|
28
|
Baker JV, Brummel-Ziedins K, Neuhaus J, Duprez D, Cummins N, Dalmau D, DeHovitz J, Lehmann C, Sullivan A, Woolley I, Kuller L, Neaton JD, Tracy RP. HIV replication alters the composition of extrinsic pathway coagulation factors and increases thrombin generation. J Am Heart Assoc 2013; 2:e000264. [PMID: 23896681 PMCID: PMC3828789 DOI: 10.1161/jaha.113.000264] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background HIV infection leads to activation of coagulation, which may increase the risk for atherosclerosis and venous thromboembolic disease. We hypothesized that HIV replication increases coagulation potentially through alterations in extrinsic pathway factors. Methods and Results Extrinsic pathway factors were measured among a subset of HIV participants from the Strategies for Management of Anti‐Retroviral Therapy (SMART) trial. Thrombin generation was estimated using validated computational modeling based on factor composition. We characterized the effect of antiretroviral therapy (ART) treatment versus the untreated state (HIV replication) via 3 separate analyses: (1) a cross‐sectional comparison of those on and off ART (n=717); (2) a randomized comparison of deferring versus starting ART (n=217); and (3) a randomized comparison of stopping versus continuing ART (n=500). Compared with viral suppression, HIV replication consistently showed short‐term increases in some procoagulants (eg, 15% to 23% higher FVIII; P<0.001) and decreases in key anticoagulants (eg, 5% to 9% lower antithrombin [AT] and 6% to 10% lower protein C; P<0.01). The net effect of HIV replication was to increase coagulation potential (eg, 24% to 48% greater thrombin generation from computational models; P<0.01 for all). The pattern of changes from HIV replication was reversed with ART treatment and consistent across all 3 independent comparisons. Conclusions HIV replication leads to complex changes in extrinsic pathway factors, with the net effect of increasing coagulation potential to a degree that may be clinically relevant. The key influence of changes in FVIII and AT suggests that HIV‐related coagulation abnormalities may involve changes in hepatocyte function in the context of systemic inflammation. Clinical Trial Registration URL: ClinicalTrials.gov. Unique identifier: NCT00027352.
Collapse
|
29
|
Niranjan R. Molecular Basis of Etiological Implications in Alzheimer’s Disease: Focus on Neuroinflammation. Mol Neurobiol 2013; 48:412-28. [DOI: 10.1007/s12035-013-8428-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/06/2013] [Indexed: 12/31/2022]
|