1
|
Annadurai N, Kanmogne GD. Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse. Cells 2024; 13:1415. [PMID: 39272987 PMCID: PMC11393916 DOI: 10.3390/cells13171415] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Blood-brain barrier (BBB) injury and dysfunction following infection with the human immunodeficiency virus (HIV) enables viral entry into the brain, infection of resident brain cells, neuronal injury and subsequent neurodegeneration leading to HIV-associated neurocognitive disorders (HAND). Although combination antiretroviral therapy has significantly reduced the incidence and prevalence of acquired immunodeficiency syndrome and increased the life expectancy of people living with HIV, the prevalence of HAND remains high. With aging of people living with HIV associated with increased comorbidities, the prevalence of HIV-related central nervous system (CNS) complications is expected to remain high. Considering the principal role of the brain endothelium in HIV infection of the CNS and HAND, the purpose of this manuscript is to review the current literature on the pathobiology of the brain endothelium structural and functional dysregulation in HIV infection, including in the presence of HIV-1 and viral proteins (gp120, Tat, Nef, and Vpr). We summarize evidence from human and animal studies, in vitro studies, and associated mechanisms. We further summarize evidence of synergy or lack thereof between commonly abused substances (cocaine, methamphetamine, alcohol, tobacco, opioids, and cannabinoids) and HIV- or viral protein-induced BBB injury and dysfunction.
Collapse
Affiliation(s)
| | - Georgette D. Kanmogne
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA;
| |
Collapse
|
2
|
Chasseigneaux S, Cochois-Guégan V, Lecorgne L, Lochus M, Nicolic S, Blugeon C, Jourdren L, Gomez-Zepeda D, Tenzer S, Sanquer S, Nivet-Antoine V, Menet MC, Laplanche JL, Declèves X, Cisternino S, Saubaméa B. Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation. Fluids Barriers CNS 2024; 21:33. [PMID: 38589879 PMCID: PMC11003008 DOI: 10.1186/s12987-024-00526-8] [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: 10/28/2023] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters. METHODS We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion. RESULTS Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting. CONCLUSIONS Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.
Collapse
Affiliation(s)
- Stéphanie Chasseigneaux
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Véronique Cochois-Guégan
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Lucas Lecorgne
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Murielle Lochus
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Sophie Nicolic
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Corinne Blugeon
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Laurent Jourdren
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - David Gomez-Zepeda
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | | | - Valérie Nivet-Antoine
- AP-HP Biochimie générale, Hôpital Necker Enfants Malades, Université Paris Cité, Inserm, Innovations Thérapeutiques en Hémostase, Paris, France
| | - Marie-Claude Menet
- Institut de Chimie Physique, CNRS UMR8000, Université Paris-Saclay, 91400, Orsay, France
| | - Jean-Louis Laplanche
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Xavier Declèves
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Salvatore Cisternino
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Bruno Saubaméa
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
3
|
Gonçalves SMC, Galdino LV, Lima MC, da Silva Moura JA, Viana DCF, da Rosa MM, Ferreira LFGR, Hernandes MZ, Pereira MC, de Melo Rêgo MJB, da Rocha Pitta I, de Oliveira França R, da Rocha Pitta MG, da Rocha Pitta MG. Evaluation of Thiazolidine Derivatives with Potential Anti-ZIKV Activity. Curr Top Med Chem 2024; 24:2224-2237. [PMID: 39136505 DOI: 10.2174/0115680266315388240801053401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE In this study, we have synthesized 19 Thiazolidine (TZD) derivatives to investigate their potential anti-ZIKV effects. METHODS Nineteen thiazolidine derivatives were synthesized and evaluated for their cytotoxicity and antiviral activity against the ZIKA virus. RESULTS Among them, six demonstrated remarkable selectivity against the ZIKV virus, exhibiting IC50 values of <5μM, and the other compounds did not demonstrate selectivity for the virus. Interestingly, several derivatives effectively suppressed the replication of ZIKV RNA copies, with derivatives significantly reducing ZIKV mRNA levels at 24 hours post-infection (hpi). Notably, two derivatives (ZKC-4 and -9) stood out by demonstrating a protective effect against ZIKV cell entry. Informed by computational analysis of binding affinity and intermolecular interactions within the NS5 domain's N-7 and O'2 positions, ZKC-4 and FT-39 displayed the highest predicted affinities. Intriguingly, ZKC-4 and ZKC-9 derivatives exhibited the most favorable predicted binding affinities for the ZIKV-E binding site. CONCLUSION The significance of TZDs as potent antiviral agents is underscored by these findings, suggesting that exploring TZD derivatives holds promise for advancing antiviral therapeutic strategies.
Collapse
Affiliation(s)
| | - Lília Vieira Galdino
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Morganna Costa Lima
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation / FIOCRUZ, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - José Arion da Silva Moura
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Douglas Carvalho Francisco Viana
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Michelle Melgarejo da Rosa
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | | | - Marcelo Zaldini Hernandes
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Artur de Sá, Recife-PE, 50740-521, Brazil
| | - Michelly Cristiny Pereira
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | | | - Ivan da Rocha Pitta
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Rafael de Oliveira França
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation / FIOCRUZ, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Marina Galdino da Rocha Pitta
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Maira Galdino da Rocha Pitta
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| |
Collapse
|
4
|
Chen Q, Wu Y, Yu Y, Wei J, Huang W. Rho-kinase inhibitor hydroxyfasudil protects against HIV-1 Tat-induced dysfunction of tight junction and neprilysin/Aβ transfer receptor expression in mouse brain microvessels. Mol Cell Biochem 2021; 476:2159-2170. [PMID: 33548010 PMCID: PMC8057965 DOI: 10.1007/s11010-021-04056-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 01/11/2021] [Indexed: 11/28/2022]
Abstract
HIV-1 transactivator protein (Tat) induces tight junction (TJ) dysfunction and amyloid-beta (Aβ) clearance dysfunction, contributing to the development and progression of HIV-1-associated neurocognitive disorder (HAND). The Rho/ROCK signaling pathway has protective effects on neurodegenerative disease. However, the underlying mechanisms of whether Rho/ROCK protects against HIV-1 Tat-caused dysfunction of TJ and neprilysin (NEP)/Aβ transfer receptor expression have not been elucidated. C57BL/6 mice were administered sterile saline (i.p., 100 μL) or Rho-kinase inhibitor hydroxyfasudil (HF) (i.p., 10 mg/kg) or HIV-1 Tat (i.v., 100 μg/kg) or HF 30 min before being exposed to HIV-1 Tat once a day for seven consecutive days. Evans Blue (EB) leakage was detected via spectrophotometer and brain slides in mouse brains. The protein and mRNA levels of zonula occludens-1 (ZO-1), occludin, NEP, receptor for advanced glycation end products (RAGE), and low-density lipoprotein receptor-related protein 1 (LRP1) in mouse brain microvessels were, respectively, analyzed by Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) analyses. Exposure of the mice to HIV-1 Tat increased the amount of EB leakage, EB fluorescence intensity, blood–brain barrier (BBB) permeability, as well as the RAGE protein and mRNA levels, and decreased the protein and mRNA levels of ZO-1, occludin, NEP, and LRP1 in mouse brain microvessels. However, these effects were weakened by Rho-kinase inhibitor HF. Taken together, these results provide information that the Rho/ROCK signaling pathway is involved in HIV-1 Tat-induced dysfunction of TJ and NEP/Aβ transfer receptor expression in the C57BL/6 mouse brain. These findings shed some light on potentiality of inhibiting Rho/Rock signaling pathway in handling HAND.
Collapse
Affiliation(s)
- Qiangtang Chen
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.,Department of Neurology, The First People's Hospital of Qinzhou, Qinzhou, 535099, Guangxi, China
| | - Yu Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yachun Yu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Junxiang Wei
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Wen Huang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Aouizerat BE, Byun E, Pullinger CR, Gay C, Lerdal A, Lee KA. Sleep disruption and duration are associated with variants in genes involved in energy homeostasis in adults with HIV/AIDS. Sleep Med 2020; 82:84-95. [PMID: 33906044 DOI: 10.1016/j.sleep.2020.08.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 07/21/2020] [Accepted: 08/25/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To determine whether selected genes and plasma markers involved in energy homeostasis are associated with sleep disruption or duration in adults with HIV/AIDS. METHODS A sample of 289 adults with HIV/AIDS wore a wrist actigraph for 72 h to estimate total sleep time (TST) and wake after sleep onset (WASO). Twenty-three single nucleotide polymorphisms (SNP) spanning 5 energy homeostasis genes (adiponectin [ADIPOQ], ghrelin [GHRL], leptin [LEP], peroxisome proliferator-activated receptor-alpha [PPARA], and -gamma [PPARG]) were genotyped using a custom array. Plasma markers of energy homeostasis (adiponectin, ghrelin, leptin) were measured by commercial multiplex assay. RESULTS After adjusting for demographic and clinical characteristics (race/ethnicity, gender, CD4 cell count, waist circumference, medications), both WASO and TST were associated with SNPs in ADIPOQ (rs182052), LEP (rs10244329, rs3828942), PPARA (rs135551, rs4253655), and PPARG (rs709151). Additional SNPs in ADIPOQ were associated with WASO (rs1501299, rs3821799, rs6773957) and TST (rs2241766). TST was also associated with SNPs in GHRL (rs26802), LEP (rs11760956), PPARA (rs135547, rs8138102, rs4253776), and PPARG (rs12490265, rs796313). Many covariate-adjusted associations involved a significant interaction with markers of HIV (viral load, years since diagnosis). Among plasma markers, higher adiponectin was associated with less WASO, higher ghrelin and glucose levels with shorter TST, and higher leptin with longer TST. CONCLUSIONS Replication of SNPs in all five genes and three plasma markers of energy homeostasis were associated with objective sleep measures. HIV disease influenced many of the associations. Findings strengthen evidence for associations between energy homeostasis genetics and poor sleep, and provide direction for pharmacological intervention research.
Collapse
Affiliation(s)
- Bradley E Aouizerat
- Bluestone Center for Clinical Research, New York University, NY, USA; Department of Oral and Maxillofacial Surgery, New York University, NY, USA.
| | - Eeeseung Byun
- Department of Biobehavioral Nursing and Health Informatics, University of Washington, Seattle, WA, USA
| | - Clive R Pullinger
- Cardiovascular Research Institute, University of California at San Francisco, San Francisco, CA, USA; Department of Physiological Nursing, University of California at San Francisco, San Francisco, CA, USA
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California at San Francisco, San Francisco, CA, USA; Department of Patient Safety and Research, Lovisenberg Diakonale Hospital, Oslo, Norway
| | - Anners Lerdal
- Department of Patient Safety and Research, Lovisenberg Diakonale Hospital, Oslo, Norway; Department of Interdisciplinary Health Sciences, Institute of Health and Society, Faculty of Medicine, University of Oslo, Norway
| | - Kathryn A Lee
- Department of Family Health Care Nursing, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Gorska AM, Eugenin EA. The Glutamate System as a Crucial Regulator of CNS Toxicity and Survival of HIV Reservoirs. Front Cell Infect Microbiol 2020; 10:261. [PMID: 32670889 PMCID: PMC7326772 DOI: 10.3389/fcimb.2020.00261] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamate (Glu) is the most abundant excitatory neurotransmitter in the central nervous system (CNS). HIV-1 and viral proteins compromise glutamate synaptic transmission, resulting in poor cell-to-cell signaling and bystander toxicity. In this study, we identified that myeloid HIV-1-brain reservoirs survive in Glu and glutamine (Gln) as a major source of energy. Thus, we found a link between synaptic compromise, metabolomics of viral reservoirs, and viral persistence. In the current manuscript we will discuss all these interactions and the potential to achieve eradication and cure using this unique metabolic profile.
Collapse
Affiliation(s)
- Anna Maria Gorska
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
7
|
Chen X, Chen H, Zhang Z, Fu Y, Han X, Zhang Y, Xu J, Ding H, Cui H, Dong T, Shang H, Jiang Y. Elevated CD54 Expression Renders CD4+ T Cells Susceptible to Natural Killer Cell-Mediated Killing. J Infect Dis 2020; 220:1892-1903. [PMID: 31433832 DOI: 10.1093/infdis/jiz413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells are an important type of effector cell in the innate immune response, and also have a role in regulation of the adaptive immune response. Several studies have indicated that NK cells may influence CD4+ T cells during HIV infection. METHODS In total, 51 HIV-infected individuals and 15 healthy controls participated in this study. We performed the flow cytometry assays and real-time PCR for the phenotypic analysis and the functional assays of NK cell-mediated deletion of CD4+ T cells, phosphorylation of nuclear factor-κB (NF-κB/p65) and the intervention of metformin. RESULTS Here we detected high CD54 expression on CD4+ T cells in HIV-infected individuals, and demonstrate that upregulated CD54 is associated with disease progression in individuals infected with HIV. We also show that CD54 expression leads to the deletion of CD4+ T cells by NK cells in vitro, and that this is modulated by NF-κB/p65 signaling. Further, we demonstrate that metformin can suppress CD54 expression on CD4+ T cells by inhibiting NF-κB/p65 phosphorylation. CONCLUSIONS Our data suggest that further studies to evaluate the potential role of metformin as adjunctive therapy to reconstitute immune function in HIV-infected individuals are warranted.
Collapse
Affiliation(s)
- Xi Chen
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Huihui Chen
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Clinical Laboratory, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Zining Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yajing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiaoxu Han
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yue Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Junjie Xu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Haibo Ding
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Hualu Cui
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Tao Dong
- MRC Human Immunology Unit, Radcliffe Department of Medicine, Oxford University, United Kingdom
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yongjun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
8
|
Yu X, Shang H, Jiang Y. ICAM-1 in HIV infection and underlying mechanisms. Cytokine 2019; 125:154830. [PMID: 31491723 DOI: 10.1016/j.cyto.2019.154830] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/29/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a glycoprotein that participates in inflammatory and immune responses. Both cell surface and soluble ICAM-1 are significantly increased during human immunodeficiency virus (HIV) infection, and ICAM-1 has important functions in promoting inflammatory responses and enhancing HIV infectivity; however, a comprehensive summary these roles has yet to be elaborated. In this review we describe the general biological characteristics of ICAM-1, its association with HIV disease progression and promotion of HIV production, mechanisms inducing upregulation of ICAM-1, and possible intervention strategies, representing important insights in the context of HIV treatment.
Collapse
Affiliation(s)
- Xiaowen Yu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China.
| | - Yongjun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China.
| |
Collapse
|
9
|
Hijmans JG, Stockelman K, Levy M, Brewster LM, Bammert TD, Greiner JJ, Connick E, DeSouza CA. Effects of HIV-1 gp120 and TAT-derived microvesicles on endothelial cell function. J Appl Physiol (1985) 2019; 126:1242-1249. [PMID: 30789287 DOI: 10.1152/japplphysiol.01048.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The aims of this study were twofold. The first was to determine if human immunodeficiency virus (HIV)-1 glycoprotein (gp) 120 and transactivator of transcription (Tat) stimulate the release of endothelial microvesicles (EMVs). The second was to determine whether viral protein-induced EMVs are deleterious to endothelial cell function (inducing endothelial cell inflammation, oxidative stress, senescence and increasing apoptotic susceptibility). Human aortic endothelial cells (HAECs) were treated with recombinant HIV-1 proteins Bal gp120 (R5), Lav gp120 (X4), or Tat. EMVs released in response to each viral protein were isolated and quantified. Fresh HAECs were treated with EMVs generated under control conditions and from each of the viral protein conditions for 24 h. EMV release was higher (P < 0.05) in HAECs treated with R5 (141 ± 21 MV/µl), X4 (132 ± 20 MV/µl), and Tat (130 ± 20 MV/µl) compared with control (61 ± 13 MV/µl). Viral protein EMVs induced significantly higher endothelial cell release of proinflammatory cytokines and expression of cell adhesion molecules than control. Reactive oxygen species production was more pronounced (P < 0.05) in the R5-, X4- and Tat-EMV-treated cells. In addition, viral protein-stimulated EMVs significantly augmented endothelial cell senescence and apoptotic susceptibility. Concomitant with these functional changes, viral protein-stimulated EMVs disrupted cell expression of micro-RNAs 34a, 126, 146a, 181b, 221, and miR-Let-7a (P < 0.05). These results demonstrate that HIV-1 gp120 and Tat stimulate microvesicle release from endothelial cells, and these microvesicles confer pathological effects on endothelial cells by inducing inflammation, oxidative stress, and senescence as well as enhancing susceptibility to apoptosis. Viral protein-generated EMVs may contribute to the increased risk of vascular disease in patients with HIV-1. NEW & NOTEWORTHY Human immunodeficiency virus (HIV)-1-related proteins glycoprotein (gp) 120 and transactivator of transcription (Tat)-mediated endothelial damage and dysfunction are poorly understood. Endothelial microvesicles (EMVs) serve as indicators and potent mediators of endothelial dysfunction. In the present study we determined if HIV-1 R5- and X4-tropic gp120 and Tat stimulate EMV release in vitro and if viral protein-induced EMVs are deleterious to endothelial cell function. gp120 and Tat induced a marked increase in EMV release. Viral protein-induced EMVs significantly increased endothelial cell inflammation, oxidative stress, senescence, and apoptotic susceptibility in vitro. gp120- and Tat-derived EMVs promote a proinflammatory, pro-oxidative, prosenescent, and proapoptotic endothelial phenotype and may contribute to the endothelial damage and dysfunction associated with gp120 and Tat.
Collapse
Affiliation(s)
- Jamie G Hijmans
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Kelly Stockelman
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Ma'ayan Levy
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - L Madden Brewster
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Tyler D Bammert
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| | - Elizabeth Connick
- Division of Infectious Disease, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado , Boulder, Colorado
| |
Collapse
|
10
|
Song J, Wang Y, Liu C, Huang Y, He L, Cai X, Lu J, Liu Y, Wang D. Cordyceps militaris fruit body extract ameliorates membranous glomerulonephritis by attenuating oxidative stress and renal inflammation via the NF-κB pathway. Food Funct 2016; 7:2006-15. [PMID: 27008597 DOI: 10.1039/c5fo01017a] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Membranous glomerulonephritis (MGN) is a common pathogenesis of nephritic syndrome in adult patients. Nuclear factor kappa B (NF-κB) serves as the main transcription factor for the inflammatory response mediated nephropathy. Cordyceps militaris, containing various pharmacological components, has been used as a kind of crude drug and folk tonic food for improving immunity and reducing inflammation. The current study aims to investigate the renoprotective activity of Cordyceps militaris aqueous extract (CM) in the cationic bovine serum albumin (C-BSA)-induced rat model of membranous glomerulonephritis. Significant renal dysfunction was observed in MGN rats; comparatively, 4-week CM administration strongly decreased the levels of 24 h urine protein, total cholesterol, triglyceride, blood urea nitrogen and serum creatinine, and increased the levels of serum albumin and total serum protein. Strikingly, recovery of the kidney histological architecture was noted in CM-treated MGN rats. A significant improvement in the glutathione peroxidase and superoxide dismutase levels, and a reduced malondialdehyde concentration were observed in the serum and kidney of CM-treated rats. Altered levels of inflammatory cytokines including interleukins, monocyte chemoattractant protein-1, intercellular adhesion molecule 1, vascular adhesion molecule 1, tumor necrosis factor-α, 6-keto-prostaglandin F1α, and nuclear transcriptional factor subunit NF-κB p65 reverted to normal levels upon treatment with CM. The present data suggest that CM protects rats against membranous glomerulonephritis via the normalization of NF-κB activity, thereby inhibiting oxidative damage and reducing inflammatory cytokine levels, which further provide experimental evidence in support of the clinical use of CM as an effective renoprotective agent.
Collapse
Affiliation(s)
- Jingjing Song
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Yingwu Wang
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Chungang Liu
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Yan Huang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110015, China
| | - Liying He
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110015, China
| | - Xueying Cai
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Jiahui Lu
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Yan Liu
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|