1
|
Henrich TJ, Bosch RJ, Godfrey C, Mar H, Nair A, Keefer M, Fichtenbaum C, Moisi D, Clagett B, Buck AM, Deitchman AN, Aweeka F, Li JZ, Kuritzkes DR, Lederman MM, Hsue PY, Deeks SG. Sirolimus reduces T cell cycling, immune checkpoint marker expression, and HIV-1 DNA in people with HIV. Cell Rep Med 2024; 5:101745. [PMID: 39321793 PMCID: PMC11513808 DOI: 10.1016/j.xcrm.2024.101745] [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: 11/12/2023] [Revised: 04/10/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024]
Abstract
Key HIV cure strategies involve reversing immune dysfunction and limiting the proliferation of infected T cells. We evaluate the safety of sirolimus, a mammalian target of rapamycin (mTOR) inhibitor, in people with HIV (PWH) and study the impact of sirolimus on HIV-1 reservoir size and HIV-1-specific immunity in a single-arm study of 20 weeks of treatment in PWH on antiretroviral therapy (ART). Sirolimus treatment does not impact HIV-1-specific CD8 T cell responses but leads to a significant decrease in CD4+ T cell-associated HIV-1 DNA levels at 20 weeks of therapy in the primary efficacy population (n = 16; 31% decline, p = 0.008). This decline persists for at least 12 weeks following cessation of the study drug. Sirolimus treatment also leads to a significant reduction in CD4+ T cell cycling and PD-1 expression on CD8+ lymphocytes. These data suggest that homeostatic proliferation of infected cells, an important mechanism for HIV persistence, is an intriguing therapeutic target.
Collapse
Affiliation(s)
- Timothy J Henrich
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA.
| | - Ronald J Bosch
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Catherine Godfrey
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Hanna Mar
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Apsara Nair
- Frontier Science and Technology Research Foundation, Amherst, NY 14226, USA
| | - Michael Keefer
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Carl Fichtenbaum
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniela Moisi
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian Clagett
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Amanda M Buck
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA; San Francisco State University, San Francisco, CA 94132, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacology, University of California San Francisco, San Francisco, CA 94110, USA
| | - Francesca Aweeka
- Department of Clinical Pharmacology, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael M Lederman
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Priscilla Y Hsue
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
2
|
Mainou E, Berendam SJ, Obregon-Perko V, Uffman EA, Phan CT, Shaw GM, Bar KJ, Kumar MR, Fray EJ, Siliciano JM, Siliciano RF, Silvestri G, Permar SR, Fouda GG, McCarthy J, Chahroudi A, Chan C, Conway JM. Comparative analysis of within-host dynamics of acute infection and viral rebound dynamics in postnatally SHIV-infected ART-treated infant rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595130. [PMID: 38826467 PMCID: PMC11142125 DOI: 10.1101/2024.05.21.595130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Viral dynamics of acute HIV infection and HIV rebound following suspension of antiretroviral therapy may be qualitatively similar but must differ given, for one, development of adaptive immune responses. Understanding the differences of acute HIV infection and viral rebound dynamics in pediatric populations may provide insights into the mechanisms of viral control with potential implications for vaccine design and the development of effective targeted therapeutics for infants and children. Mathematical models have been a crucial tool to elucidate the complex processes driving viral infections within the host. Traditionally, acute HIV infection has been modeled with a standard model of viral dynamics initially developed to explore viral decay during treatment, while viral rebound has necessitated extensions of that standard model to incorporate explicit immune responses. Previous efforts to fit these models to viral load data have underscored differences between the two infection stages, such as increased viral clearance rate and increased death rate of infected cells during rebound. However, these findings have been predicated on viral load measurements from disparate adult individuals. In this study, we aim to bridge this gap, in infants, by comparing the dynamics of acute infection and viral rebound within the same individuals by leveraging an infant nonhuman primate Simian/Human Immunodeficiency Virus (SHIV) infection model. Ten infant Rhesus macaques (RMs) orally challenged with SHIV.C.CH505 375H dCT and given ART at 8 weeks post-infection. These infants were then monitored for up to 60 months post-infection with serial viral load and immune measurements. We use the HIV standard viral dynamics model fitted to viral load measurements in a nonlinear mixed effects framework. We find that the primary difference between acute infection and rebound is the increased death rate of infected cells during rebound. We use these findings to generate hypotheses on the effects of adaptive immune responses. We leverage these findings to formulate hypotheses to elucidate the observed results and provide arguments to support the notion that delayed viral rebound is characterized by a stronger CD8+ T cell response.
Collapse
Affiliation(s)
- Ellie Mainou
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | | | | | - Emilie A Uffman
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Caroline T Phan
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - George M Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katharine J Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mithra R Kumar
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily J Fray
- Department of Biochemistry and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janet M Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | | - Janice McCarthy
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Jessica M Conway
- Department of Mathematics, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
3
|
Mazzuti L, Turriziani O, Mezzaroma I. The Many Faces of Immune Activation in HIV-1 Infection: A Multifactorial Interconnection. Biomedicines 2023; 11:biomedicines11010159. [PMID: 36672667 PMCID: PMC9856151 DOI: 10.3390/biomedicines11010159] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
Chronic immune activation has a significant role in HIV-1 disease pathogenesis and CD4+ T-cell depletion. The causes of chronic inflammation and immune activation are incompletely understood, but they are likely multifactorial in nature, involving both direct and indirect stimuli. Possible explanations include microbial translocation, coinfection, and continued presence of competent replicating virus. In fact, long-term viral suppression treatments are unable to normalize elevated markers of systemic immune activation. Furthermore, high levels of pro-inflammatory cytokines increase susceptibility to premature aging of the immune system. The phenomenon of "inflammaging" has begun to be evident in the last decades, as a consequence of increased life expectancy due to the introduction of cART. Quality of life and survival have improved substantially; however, PLWH are predisposed to chronic inflammatory conditions leading to age-associated diseases, such as inflammatory bowel disease, neurocognitive disorders, cardiovascular diseases, metabolic syndrome, bone abnormalities, and non-HIV-associated cancers. Several approaches have been studied in numerous uncontrolled and/or randomized clinical trials with the aim of reducing immune activation/inflammatory status in PLWH, none of which have achieved consistent results.
Collapse
Affiliation(s)
- Laura Mazzuti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Ombretta Turriziani
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Ivano Mezzaroma
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
4
|
Mu W, Rezek V, Martin H, Carrillo MA, Tomer S, Hamid P, Lizarraga MA, Tibbe TD, Yang OO, Jamieson BD, Kitchen SG, Zhen A. Autophagy inducer rapamycin treatment reduces IFN-I-mediated Inflammation and improves anti-HIV-1 T cell response in vivo. JCI Insight 2022; 7:e159136. [PMID: 36509289 PMCID: PMC9746825 DOI: 10.1172/jci.insight.159136] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
A hallmark of HIV-1 infection is chronic inflammation, even in patients treated with antiretroviral therapy (ART). Chronic inflammation drives HIV-1 pathogenesis, leading to loss of CD4+ T cells and exhaustion of antiviral immunity. Therefore, strategies to safely reduce systematic inflammation are needed to halt disease progression and restore defective immune responses. Autophagy is a cellular mechanism for disposal of damaged organelles and elimination of intracellular pathogens. Autophagy is pivotal for energy homeostasis and plays critical roles in regulating immunity. However, how it regulates inflammation and antiviral T cell responses during HIV infection is unclear. Here, we demonstrate that autophagy is directly linked to IFN-I signaling, which is a key driver of immune activation and T cell exhaustion during chronic HIV infection. Impairment of autophagy leads to spontaneous IFN-I signaling, and autophagy induction reduces IFN-I signaling in monocytic cells. Importantly, in HIV-1-infected humanized mice, autophagy inducer rapamycin treatment significantly reduced persistent IFN-I-mediated inflammation and improved antiviral T cell responses. Cotreatment of rapamycin with ART led to significantly reduced viral rebound after ART withdrawal. Taken together, our data suggest that therapeutically targeting autophagy is a promising approach to treat persistent inflammation and improve immune control of HIV replication.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Valerie Rezek
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Heather Martin
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mayra A. Carrillo
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Shallu Tomer
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Philip Hamid
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Miguel A. Lizarraga
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Tristan D. Tibbe
- Statistic Core, Department of Medicine at UCLA, Los Angeles, California, USA
| | - Otto O. Yang
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Division of Infectious Disease and
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | | | - Scott G. Kitchen
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Anjie Zhen
- Division of Hematology/Oncology, Department of Medicine and
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
5
|
Varco-Merth BD, Brantley W, Marenco A, Duell DD, Fachko DN, Richardson B, Busman-Sahay K, Shao D, Flores W, Engelman K, Fukazawa Y, Wong SW, Skalsky RL, Smedley J, Axthelm MK, Lifson JD, Estes JD, Edlefsen PT, Picker L, Cameron CM, Henrich TJ, Okoye AA. Rapamycin limits CD4+ T cell proliferation in simian immunodeficiency virus-infected rhesus macaques on antiretroviral therapy. J Clin Invest 2022; 132:156063. [PMID: 35316218 PMCID: PMC9106346 DOI: 10.1172/jci156063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/16/2022] [Indexed: 11/28/2022] Open
Abstract
Proliferation of latently infected CD4+ T cells with replication-competent proviruses is an important mechanism contributing to HIV persistence during antiretroviral therapy (ART). One approach to targeting this latent cell expansion is to inhibit mTOR, a regulatory kinase involved with cell growth, metabolism, and proliferation. Here, we determined the effects of chronic mTOR inhibition with rapamycin with or without T cell activation in SIV-infected rhesus macaques (RMs) on ART. Rapamycin perturbed the expression of multiple genes and signaling pathways important for cellular proliferation and substantially decreased the frequency of proliferating CD4+ memory T cells (TM cells) in blood and tissues. However, levels of cell-associated SIV DNA and SIV RNA were not markedly different between rapamycin-treated RMs and controls during ART. T cell activation with an anti-CD3LALA antibody induced increases in SIV RNA in plasma of RMs on rapamycin, consistent with SIV production. However, upon ART cessation, both rapamycin and CD3LALA–treated and control-treated RMs rebounded in less than 12 days, with no difference in the time to viral rebound or post-ART viral load set points. These results indicate that, while rapamycin can decrease the proliferation of CD4+ TM cells, chronic mTOR inhibition alone or in combination with T cell activation was not sufficient to disrupt the stability of the SIV reservoir.
Collapse
Affiliation(s)
- Benjamin D Varco-Merth
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - William Brantley
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Alejandra Marenco
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Derick D Duell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Devin N Fachko
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Brian Richardson
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, United States of America
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Danica Shao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, United States of America
| | - Walter Flores
- MassBiologics, University of Massachusetts Medical School, Boston, United States of America
| | - Kathleen Engelman
- MassBiologics, University of Massachusetts Medical School, Boston, United States of America
| | - Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Scott W Wong
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Rebecca L Skalsky
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, United States of America
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, United States of America
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, United States of America
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, United States of America
| | - Louis Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| | - Cheryl Ma Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, United States of America
| | - Timothy J Henrich
- Department of Medicine, UCSF, San Francisco, United States of America
| | - Afam A Okoye
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, United States of America
| |
Collapse
|
6
|
HIV-Related Immune Activation and Inflammation: Current Understanding and Strategies. J Immunol Res 2021; 2021:7316456. [PMID: 34631899 PMCID: PMC8494587 DOI: 10.1155/2021/7316456] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
Although antiretroviral therapy effectively controls human immunodeficiency virus (HIV) replication, a residual chronic immune activation/inflammation persists throughout the disease. This aberrant immune activation and inflammation are considered an accelerator of non-AIDS-related events and one of the driving forces of CD4+ T cell depletion. Unfortunately, HIV-associated immune activation is driven by various factors, while the mechanism of excessive inflammation has not been formally clarified. To date, several clinical interventions or treatment candidates undergoing clinical trials have been proposed to combat this systemic immune activation/inflammation. However, these strategies revealed limited results, or their nonspecific anti-inflammatory properties are similar to previous interventions. Here, we reviewed recent learnings of immune activation and persisting inflammation associated with HIV infection, as well as the current directions to overcome it. Of note, a more profound understanding of the specific mechanisms for aberrant inflammation is still imperative for identifying an effective clinical intervention strategy.
Collapse
|
7
|
Guerrini GP, Berretta M, Guaraldi G, Magistri P, Esposito G, Ballarin R, Serra V, Di Sandro S, Di Benedetto F. Liver Transplantation for HCC in HIV-Infected Patients: Long-Term Single-Center Experience. Cancers (Basel) 2021; 13:cancers13184727. [PMID: 34572954 PMCID: PMC8471924 DOI: 10.3390/cancers13184727] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND HIV-infected patients now have long life expectation since the introduction of the highly active antiretroviral therapy (HAART). Liver diseases, especially cirrhosis and hepatocellular carcinoma (HCC), currently represent a leading cause of death in this setting of patients. AIM To address the results of liver transplantation (LT) for HCC in HIV-infected patients. METHODS All patients with and without HIV infection who underwent LT for HCC (n = 420) between 2001 and 2021 in our center were analyzed with the intent of comparing graft and patient survival. Cox regression analysis was used to determine prognostic survival factors and logistic regression to determine the predictor factors of post-LT recurrence. RESULTS Among 1010 LT, 32 were HIV-infected recipients. With an average follow-up of 62 ± 51 months, 5-year overall survival in LT recipients with and without HIV-infection was 71.6% and 69.9%, respectively (p = ns), whereas 5-year graft survival in HIV-infected and HIV-non infected was 68.3% and 68.2%, respectively (p = ns). The independent predictive factor of survival in the study group was: HCV infection (HR 1.83, p = 0.024). There were no significant differences in the pathological characteristics of HCC between the two groups. The logistic regression analysis of the study population demonstrated that microvascular invasion (HR 5.18, p< 0.001), HCC diameter (HR 1.16, p = 0.028), and number of HCC nodules (HR 1.26, p = 0.003) were predictors of recurrence post-LT. CONCLUSION Our study shows that HIV patients undergoing LT for HCC have comparable results in terms of post-LT survival. Excellent results can be achieved for HIV-infected patients with HCC, as long as a strategy of close surveillance and precise treatment of the tumor is adopted while on the waiting list.
Collapse
Affiliation(s)
- Gian Piero Guerrini
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Massimiliano Berretta
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Giovanni Guaraldi
- Infectious Diseases Unit, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy;
| | - Paolo Magistri
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Giuseppe Esposito
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Roberto Ballarin
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Valentina Serra
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Stefano Di Sandro
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
| | - Fabrizio Di Benedetto
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41125 Modena, Italy; (G.P.G.); (P.M.); (G.E.); (R.B.); (V.S.); (S.D.S.)
- Correspondence:
| |
Collapse
|
8
|
Chen F, Shi Q, Pei F, Vogt A, Porritt RA, Garcia G, Gomez AC, Cheng MH, Schurdak ME, Liu B, Chan SY, Arumugaswami V, Stern AM, Taylor DL, Arditi M, Bahar I. A systems-level study reveals host-targeted repurposable drugs against SARS-CoV-2 infection. Mol Syst Biol 2021; 17:e10239. [PMID: 34339582 PMCID: PMC8328275 DOI: 10.15252/msb.202110239] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism of SARS-CoV-2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, we identified a set of repurposable and investigational drugs as potential therapeutics against COVID-19. These were deduced from the gene expression signature of SARS-CoV-2-infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral-host interactome. We also identified immuno-modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID-19 patients, based on the transcriptome of ACE2-overexpressing A549 cells. Experiments with Vero-E6 cells infected by SARS-CoV-2, as well as independent syncytia formation assays for probing ACE2/SARS-CoV-2 spike protein-mediated cell fusion using HEK293T and Calu-3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero-E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These novel findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID-19.
Collapse
Affiliation(s)
- Fangyuan Chen
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Qingya Shi
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Fen Pei
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Andreas Vogt
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Rebecca A Porritt
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Gustavo Garcia
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Angela C Gomez
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Mary Hongying Cheng
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Mark E Schurdak
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Bing Liu
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Stephen Y Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine InstituteUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of CardiologyDepartment of MedicineUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Andrew M Stern
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - D Lansing Taylor
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Moshe Arditi
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Ivet Bahar
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| |
Collapse
|
9
|
Campbell GR, Spector SA. Induction of Autophagy to Achieve a Human Immunodeficiency Virus Type 1 Cure. Cells 2021; 10:cells10071798. [PMID: 34359967 PMCID: PMC8307643 DOI: 10.3390/cells10071798] [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] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Effective antiretroviral therapy has led to significant human immunodeficiency virus type 1 (HIV-1) suppression and improvement in immune function. However, the persistence of integrated proviral DNA in latently infected reservoir cells, which drive viral rebound post-interruption of antiretroviral therapy, remains the major roadblock to a cure. Therefore, the targeted elimination or permanent silencing of this latently infected reservoir is a major focus of HIV-1 research. The most studied approach in the development of a cure is the activation of HIV-1 expression to expose latently infected cells for immune clearance while inducing HIV-1 cytotoxicity—the “kick and kill” approach. However, the complex and highly heterogeneous nature of the latent reservoir, combined with the failure of clinical trials to reduce the reservoir size casts doubt on the feasibility of this approach. This concern that total elimination of HIV-1 from the body may not be possible has led to increased emphasis on a “functional cure” where the virus remains but is unable to reactivate which presents the challenge of permanently silencing transcription of HIV-1 for prolonged drug-free remission—a “block and lock” approach. In this review, we discuss the interaction of HIV-1 and autophagy, and the exploitation of autophagy to kill selectively HIV-1 latently infected cells as part of a cure strategy. The cure strategy proposed has the advantage of significantly decreasing the size of the HIV-1 reservoir that can contribute to a functional cure and when optimised has the potential to eradicate completely HIV-1.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA;
- Correspondence: ; Tel.: +1-858-534-7477
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA;
- Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
10
|
Henrich TJ, Schreiner C, Cameron C, Hogan LE, Richardson B, Rutishauser RL, Deitchman AN, Chu S, Rogers R, Thanh C, Gibson EA, Zarinsefat A, Bakkour S, Aweeka F, Busch MP, Liegler T, Baker C, Milush J, Deeks SG, Stock PG. Everolimus, an mTORC1/2 inhibitor, in ART-suppressed individuals who received solid organ transplantation: A prospective study. Am J Transplant 2021; 21:1765-1779. [PMID: 32780519 PMCID: PMC9177122 DOI: 10.1111/ajt.16244] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/30/2020] [Accepted: 07/19/2020] [Indexed: 01/25/2023]
Abstract
Pharmacologic inhibition of the mammalian target of rapamycin (mTOR) in the setting of renal transplantation has previously been associated with lower human immunodeficiency virus 1 (HIV-1) DNA burden, and in vitro studies suggest that mTOR inhibition may lead to HIV transcriptional silencing. Because prospective clinical trials are lacking, we conducted an open-label, single-arm study to determine the impact of the broad mTOR inhibitor, everolimus, on residual HIV burden, transcriptional gene expression profiles, and immune responses in HIV-infected adult solid organ transplant (SOT) recipients on antiretroviral therapy. Whereas everolimus therapy did not have an overall effect on cell-associated HIV-1 DNA and RNA levels in the entire cohort, participants who maintained everolimus time-averaged trough levels >5 ng/mL during the first 2 months of therapy had significantly lower RNA levels up to 6 months after the cessation of study drug. Time-averaged everolimus trough levels significantly correlated with greater inhibition of mTOR gene pathway transcriptional activity. Everolimus treatment also led to decreased PD-1 expression on certain T cell subsets. These data support the rationale for further study of the effects of mTOR inhibition on HIV transcriptional silencing in non-SOT populations, either alone or in combination with other strategies. Trial Registration: ClinicalTrials.gov NCT02429869.
Collapse
Affiliation(s)
- Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Corinna Schreiner
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA,Institute of Biochemistry and Molecular Biology, Ulm University, Germany
| | - Cheryl Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Louise E. Hogan
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Brian Richardson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Rachel L. Rutishauser
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amelia N. Deitchman
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| | - Simon Chu
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Rodney Rogers
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Erica A. Gibson
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Arya Zarinsefat
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Francesca Aweeka
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| | | | - Teri Liegler
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Christopher Baker
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Jeffrey Milush
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Peter G. Stock
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
11
|
Cloherty APM, van Teijlingen NH, Eisden TJTHD, van Hamme JL, Rader AG, Geijtenbeek TBH, Schreurs RRCE, Ribeiro CMS. Autophagy-enhancing drugs limit mucosal HIV-1 acquisition and suppress viral replication ex vivo. Sci Rep 2021; 11:4767. [PMID: 33637808 PMCID: PMC7910550 DOI: 10.1038/s41598-021-84081-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 02/05/2021] [Indexed: 01/31/2023] Open
Abstract
Current direct-acting antiviral therapies are highly effective in suppressing HIV-1 replication. However, mucosal inflammation undermines prophylactic treatment efficacy, and HIV-1 persists in long-lived tissue-derived dendritic cells (DCs) and CD4+ T cells of treated patients. Host-directed strategies are an emerging therapeutic approach to improve therapy outcomes in infectious diseases. Autophagy functions as an innate antiviral mechanism by degrading viruses in specialized vesicles. Here, we investigated the impact of pharmaceutically enhancing autophagy on HIV-1 acquisition and viral replication. To this end, we developed a human tissue infection model permitting concurrent analysis of HIV-1 cellular targets ex vivo. Prophylactic treatment with autophagy-enhancing drugs carbamazepine and everolimus promoted HIV-1 restriction in skin-derived CD11c+ DCs and CD4+ T cells. Everolimus also decreased HIV-1 susceptibility to lab-adapted and transmitted/founder HIV-1 strains, and in vaginal Langerhans cells. Notably, we observed cell-specific effects of therapeutic treatment. Therapeutic rapamycin treatment suppressed HIV-1 replication in tissue-derived CD11c+ DCs, while all selected drugs limited viral replication in CD4+ T cells. Strikingly, both prophylactic and therapeutic treatment with everolimus or rapamycin reduced intestinal HIV-1 productive infection. Our findings highlight host autophagy pathways as an emerging target for HIV-1 therapies, and underscore the relevancy of repurposing clinically-approved autophagy drugs to suppress mucosal HIV-1 replication.
Collapse
Affiliation(s)
- Alexandra P M Cloherty
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Nienke H van Teijlingen
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Tracy-Jane T H D Eisden
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - John L van Hamme
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Anusca G Rader
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Teunis B H Geijtenbeek
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Renée R C E Schreurs
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands
| | - Carla M S Ribeiro
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Pediatric HIV: the Potential of Immune Therapeutics to Achieve Viral Remission and Functional Cure. Curr HIV/AIDS Rep 2020; 17:237-248. [PMID: 32356090 DOI: 10.1007/s11904-020-00495-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW In the absence of antiretroviral therapy (ART), more than 50% of perinatally HIV-infected children die by 2 years of age. Early ART from infancy is therefore a global recommendation and significantly improves immune health, child survival, and disease outcome. However, even early treatment does not prevent or eradicate the latent reservoir necessitating life-long ART. Adherence to life-long ART is challenging for children and longstanding ART during chronic HIV infection led to higher risks of non-AIDS co-morbidities and virologic failure in infected children. Thus, HIV-infected children are an important population for consideration for immune-based interventions to achieve ART-free remission and functional cure. This review summarizes how the uniqueness of the early life immune system can be harnessed for the development of ART-free remission and functional cure, which means complete virus control in absence of ART. In addition, recent advances in therapeutics in the HIV cure field and their potential for the treatment of pediatric HIV infections are discussed. RECENT FINDINGS Preclinical studies and clinical trials demonstrated that immune-based interventions target HIV replication, limit size of virus reservoir, maintain virus suppression, and delay time to virus rebound. However, these studies have been performed so far only in carefully selected HIV-infected adults, highlighting the need to evaluate the efficacy of immune-based therapeutics in HIV-infected children and to design interventions tailored to the early life maturing immune system. Immune-based therapeutics alone or in combination with ART should be actively explored as potential strategies to achieve viral remission and functional cure in HIV-infected pediatric populations.
Collapse
|
13
|
Jones NP. Immunosuppression in the Management of Presumed Non-infective Uveitis; Are We Sure What We are Treating? Notes on the Antimicrobial Properties of the Systemic Immunosuppressants. Ocul Immunol Inflamm 2020; 28:994-1003. [PMID: 31418624 DOI: 10.1080/09273948.2019.1643030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To describe the antimicrobial effects of immunosuppressants used for presumed autoimmune uveitis, and to discuss the potential importance of these effects in the context of increasing knowledge of the human microbiomes and their influence on inflammation. METHODS Literature review. REVIEW OF EVIDENCE All immunosuppressants have intrinsic antimicrobial effects; these vary considerably between drugs, and include antibacterial, antiviral and antifungal action. Immunosuppression is known to affect the composition of the gut microbiome, and alterations in microbiome composition are known to affect inflammations including uveitis. CONCLUSIONS Oral immunosuppressants are assumed to act on presumed autoimmune uveitis by downregulation of, or other interference with, an aberrant immune response. However, their antimicrobial properties are usually forgotten, and in the context of increasing knowledge of the involvement of microbes in the initiation of, and also potentially the perpetuation of, tissue inflammation, these effects may prove to be a fundamental part of their action.
Collapse
Affiliation(s)
- Nicholas P Jones
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust , Manchester, UK.,Medical Academic Health Science Centre, University of Manchester , Manchester, UK
| |
Collapse
|
14
|
Talotta R, Atzeni F, Laska MJ. Retroviruses in the pathogenesis of systemic lupus erythematosus: Are they potential therapeutic targets? Autoimmunity 2020; 53:177-191. [PMID: 32321325 DOI: 10.1080/08916934.2020.1755962] [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: 02/08/2023]
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is characterised by the hyper-activation of immunologic pathways related to the antiviral response. Exogenous and endogenous retroviruses, by integrating their DNA templates in the host cell genome, may epigenetically control the transcription of genes involved in the immune response. Furthermore, their nucleic acids or neo-synthesized proteins could stimulate the sensor molecules placed upstream the inflammatory cascade. Exogenous retroviruses, like human immunodeficiency virus, have been associated to SLE-like manifestations or to a fair SLE diagnosis. In addition, there is some evidence confirming a pathogenic role of human endogenous retroviruses in SLE. In line with these data, the use of antiretroviral agents could represent an attractive opportunity in the future therapeutic algorithms of this disease, but studies are still missing.
Collapse
Affiliation(s)
- Rossella Talotta
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital "Gaetano Martino", Messina, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital "Gaetano Martino", Messina, Italy
| | | |
Collapse
|
15
|
Alfano G, Fontana F, Guaraldi G, Cappelli G, Mussini C. Successful treatment of BK virus associated-nephropathy in a human immunodeficiency virus-positive kidney transplant recipient. Int J STD AIDS 2020; 31:387-391. [PMID: 32075537 DOI: 10.1177/0956462419900853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BK virus (BKV) is an opportunistic pathogen in those with impaired immunity. Viral replication is generally asymptomatic but is able to induce cytopathic alterations in renal cells. If BKV infection is left untreated, it leads to BKV-associated nephropathy (BKVAN) and graft loss. There is scarce experience in the management of BKV infection in kidney transplant recipients living with HIV. We report the successful treatment of BKVAN in an HIV-positive kidney transplant recipient who experienced BKV replication in the immediate post-transplantation period. A change in therapy from calcineurin inhibitor to sirolimus, steroid withdrawal and a short course of an immunomodulatory agent (leflunomide) controlled BKV viremia in the absence of drug side-effects or impairment of graft function.
Collapse
Affiliation(s)
- Gaetano Alfano
- Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy.,Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Fontana
- Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy
| | - Giovanni Guaraldi
- Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Gianni Cappelli
- Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy.,Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| |
Collapse
|
16
|
Alfano G, Fontana F, Mori G, Vicari E, Dolci G, Franceschini E, Guaraldi G, Mussini C, Cappelli G. Antiviral activity of sirolimus in an HIV-positive kidney transplant recipient. Int J STD AIDS 2019; 30:919-922. [PMID: 31280689 DOI: 10.1177/0956462419839520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Sirolimus (SIR) is a potent immunosuppressive agent with multiple proprieties. We report beneficial antiviral effects of SIR in an HIV-positive kidney transplant recipient who experienced low-level HIV-1 replication. The immunosuppressive agent was well tolerated by the patient, and no side effects were reported during follow-up. Despite immunosuppressive monotherapy, SIR ensured stable graft function.
Collapse
Affiliation(s)
- Gaetano Alfano
- 1 Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy.,2 Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Fontana
- 1 Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy.,2 Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giacomo Mori
- 1 Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy
| | - Emanuela Vicari
- 2 Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Dolci
- 3 Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Erica Franceschini
- 3 Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Giovanni Guaraldi
- 3 Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Cristina Mussini
- 3 Clinic of Infectious Diseases, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Gianni Cappelli
- 1 Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Modena, Italy.,2 Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
17
|
The Role of mTOR Inhibitors in the Management of Viral Infections: A Review of Current Literature. Transplantation 2018; 102:S50-S59. [PMID: 29369973 DOI: 10.1097/tp.0000000000001777] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Viruses are the leading cause of infections after solid organ transplant. The antiviral properties of mammalian target of rapamycin inhibitors (mTORis) have been ascribed to a variety of mechanisms and historical data have supported their use over other immunosuppressants for a myriad of viruses. Herein, we summarize the most current data to highlight the role of mTORis in the management of viral infections after solid organ transplant. The mTORis play a clear role in the management of cytomegalovirus, and have data supporting their potential use for BK virus and human herpesvirus 8-related Kaposi sarcoma. No data definitively supports mTORis for use in Epstein-Barr virus-mediated posttransplant lymphoproliferative disorder or hepatitis C virus viral replication. Although theoretically an advantageous therapy for hepatitis C virus-related liver allograft fibrosis and human immunodeficiency virus, mTORi use specifically for these indications is less attractive with modern treatments currently available. Data surrounding mTORi efficacy in preventing rejection, and their toxicity profile must be balanced with their potential antiviral effects in combination with patient-specific factors.
Collapse
|
18
|
Pacheco LS, Garcia VD, Prá RLD, Cardoso BD, Rodrigues MF, Zanetti HK, Meinerz G, Neumann J, Gnatta D, Keitel E. Effect of conversion from calcineurin inhibitors to everolimus on hepatitis C viremia in adult kidney transplant recipients. ACTA ACUST UNITED AC 2018; 40:143-150. [PMID: 29771270 PMCID: PMC6533980 DOI: 10.1590/2175-8239-jbn-3860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/03/2017] [Indexed: 01/15/2023]
Abstract
Introduction: Currently, there is no specific immunosuppressive protocol for hepatitis C
(HCV)-positive renal transplants recipients. Thus, the aim of this study was
to evaluate the conversion effect to everolimus (EVR) on HCV in adult kidney
recipients. Method: This is an exploratory single-center, prospective, randomized, open label
controlled trial with renal allograft recipients with HCV-positive serology.
Participants were randomized for conversion to EVR or maintenance of
calcineurin inhibitors. Results: Thirty patients were randomized and 28 were followed-up for 12 months
(conversion group, Group 1 =15 and control group, Group 2 =13). RT-PCR HCV
levels reported in log values were comparable in both groups and among
patients in the same group. The statistical analysis showed no interaction
effect between time and group (p value G*M= 0.852), overtime intra-groups
(p-value M=0.889) and between group (p-value G=0.286). Group 1 showed a
higher incidence of dyslipidemia (p=0.03) and proteinuria events (p=0.01),
while no difference was observed in the incidence of anemia (p=0.17), new
onset of post-transplant diabetes mellitus (p=1.00) or urinary tract
infection (p=0.60). The mean eGFR was similar in both groups. Conclusion: Our study did not show viral load decrease after conversion to EVR with
maintenance of antiproliferative therapy.
Collapse
Affiliation(s)
- Larissa Sgaria Pacheco
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Valter Duro Garcia
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Ronivan Luis Dal Prá
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Bruna Doleys Cardoso
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Mariana Ferras Rodrigues
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Helen Kris Zanetti
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Gisele Meinerz
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Jorge Neumann
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Diego Gnatta
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| | - Elizete Keitel
- Santa Casa de Misericórdia de Porto Alegre, Departamento de Nefrologia e Transplante de Rim, Porto Alegre, RS, Brasil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Patologia, Porto Alegre, RS, Brasil
| |
Collapse
|
19
|
Dyall J, Gross R, Kindrachuk J, Johnson RF, Olinger GG, Hensley LE, Frieman MB, Jahrling PB. Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies. Drugs 2017; 77:1935-1966. [PMID: 29143192 PMCID: PMC5733787 DOI: 10.1007/s40265-017-0830-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
No specific antivirals are currently available for two emerging infectious diseases, Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS). A literature search was performed covering pathogenesis, clinical features and therapeutics, clinically developed drugs for repurposing and novel drug targets. This review presents current knowledge on the epidemiology, pathogenesis and clinical features of the SARS and MERS coronaviruses. The rationale for and outcomes with treatments used for SARS and MERS is discussed. The main focus of the review is on drug development and the potential that drugs approved for other indications provide for repurposing. The drugs we discuss belong to a wide range of different drug classes, such as cancer therapeutics, antipsychotics, and antimalarials. In addition to their activity against MERS and SARS coronaviruses, many of these approved drugs have broad-spectrum potential and have already been in clinical use for treating other viral infections. A wealth of knowledge is available for these drugs. However, the information in this review is not meant to guide clinical decisions, and any therapeutic described here should only be used in context of a clinical trial. Potential targets for novel antivirals and antibodies are discussed as well as lessons learned from treatment development for other RNA viruses. The article concludes with a discussion of the gaps in our knowledge and areas for future research on emerging coronaviruses.
Collapse
Affiliation(s)
- Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA.
| | - Robin Gross
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Jason Kindrachuk
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MN, Canada
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | | | - Lisa E Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Matthew B Frieman
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Peter B Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
20
|
Sokoya T, Steel HC, Nieuwoudt M, Rossouw TM. HIV as a Cause of Immune Activation and Immunosenescence. Mediators Inflamm 2017; 2017:6825493. [PMID: 29209103 PMCID: PMC5676471 DOI: 10.1155/2017/6825493] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 12/20/2022] Open
Abstract
Systemic immune activation has emerged as an essential component of the immunopathogenesis of HIV. It not only leads to faster disease progression, but also to accelerated decline of overall immune competence. HIV-associated immune activation is characterized by an increase in proinflammatory mediators, dysfunctional T regulatory cells, and a pattern of T-cell-senescent phenotypes similar to those seen in the elderly. These changes predispose HIV-infected persons to comorbid conditions that have been linked to immunosenescence and inflamm-ageing, such as atherosclerosis and cardiovascular disease, neurodegeneration, and cancer. In the antiretroviral treatment era, development of such non-AIDS-defining, age-related comorbidities is a major cause of morbidity and mortality. Treatment strategies aimed at curtailing persistent immune activation and inflammation may help prevent the development of these conditions. At present, the most effective strategy appears to be early antiretroviral treatment initiation. No other treatment interventions have been found effective in large-scale clinical trials, and no adjunctive treatment is currently recommended in international HIV treatment guidelines. This article reviews the role of systemic immune activation in the immunopathogenesis of HIV infection, its causes and the clinical implications linked to immunosenescence in adults, and the therapeutic interventions that have been investigated.
Collapse
Affiliation(s)
- T. Sokoya
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria 0001, South Africa
| | - H. C. Steel
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria 0001, South Africa
| | - M. Nieuwoudt
- South African Department of Science and Technology (DST)/National Research Foundation (NRF) Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch 7600, South Africa
| | - T. M. Rossouw
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria 0001, South Africa
| |
Collapse
|
21
|
Cinti A, Le Sage V, Milev MP, Valiente-Echeverría F, Crossie C, Miron MJ, Panté N, Olivier M, Mouland AJ. HIV-1 enhances mTORC1 activity and repositions lysosomes to the periphery by co-opting Rag GTPases. Sci Rep 2017; 7:5515. [PMID: 28710431 PMCID: PMC5511174 DOI: 10.1038/s41598-017-05410-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/30/2017] [Indexed: 12/20/2022] Open
Abstract
HIV-1 co-opts several host machinery to generate a permissive environment for viral replication and transmission. In this work we reveal how HIV-1 impacts the host translation and intracellular vesicular trafficking machineries for protein synthesis and to impede the physiological late endosome/lysosome (LEL) trafficking in stressful conditions. First, HIV-1 enhances the activity of the master regulator of protein synthesis, the mammalian target of rapamycin (mTOR). Second, the virus commandeers mTOR-associated late endosome/lysosome (LEL) trafficking and counteracts metabolic and environmental stress-induced intracellular repositioning of LEL. We then show that the small Rag GTPases, RagA and RagB, are required for the HIV-1-mediated LEL repositioning that is likely mediated by interactions between the Rags and the viral proteins, Gag and Vif. siRNA-mediated depletion of RagA and RagB leads to a loss in mTOR association to LEL and to a blockade of viral particle assembly and release at the plasma membrane with a marked concomitant reduction in virus production. These results show that HIV-1 co-opts fundamental mechanisms that regulate LEL motility and positioning and support the notion that LEL positioning is critical for HIV-1 replication.
Collapse
Affiliation(s)
- Alessandro Cinti
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada.,Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada
| | - Valerie Le Sage
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada
| | - Miroslav P Milev
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada.,Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada
| | - Fernando Valiente-Echeverría
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada.,Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada.,Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia, 834100, Santiago, Chile
| | - Christina Crossie
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada.,Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada
| | - Marie-Joelle Miron
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada
| | - Nelly Panté
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Martin Olivier
- Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, H3A 2B4, Canada
| | - Andrew J Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, H3T 1E2, Canada. .,Department of Medicine and the Division of Experimental Medicine, McGill University, Montréal, Québec, H3A 0G4, Canada. .,Department of Microbiology and Immunology, McGill University, Montréal, Québec, H3A 2B4, Canada.
| |
Collapse
|
22
|
Nardacci R, Ciccosanti F, Marsella C, Ippolito G, Piacentini M, Fimia GM. Role of autophagy in HIV infection and pathogenesis. J Intern Med 2017; 281:422-432. [PMID: 28139864 DOI: 10.1111/joim.12596] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The aim of autophagy is to re-establish homeostasis in response to a variety of stress conditions. By forming double-membrane vesicles, autophagy engulfs damaged or superfluous cytoplasmic material and recycles degradation products for new synthesis or energy production. Of note, the same mechanism is used to capture pathogens and has important implications in both innate and adaptive immunity. To establish a chronic infection, pathogens have therefore evolved multiple mechanisms to evade autophagy-mediated degradation. HIV infection represents one of the best characterized systems in which autophagy is disarmed by a virus using multiple strategies to prevent the sequestration and degradation of its proteins and to establish a chronic infection. HIV alters autophagy at various stages of the process in both infected and bystander cells. In particular, the HIV proteins TAT, NEF and ENV are involved in this regulation by either blocking or stimulating autophagy through direct interaction with autophagy proteins and/or modulation of the mTOR pathway. Although the roles of autophagy during HIV infection are multiple and vary amongst the different cell types, several lines of evidence point to a potential beneficial effect of stimulating autophagy-mediated lysosomal degradation to potentiate the immune response to HIV. Characterization of the molecular mechanisms regulating selective autophagy is expected to be valuable for developing new drugs able to specifically enhance the anti-HIV response.
Collapse
Affiliation(s)
- R Nardacci
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy
| | - F Ciccosanti
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy
| | - C Marsella
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy
| | - G Ippolito
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy
| | - M Piacentini
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy.,Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - G M Fimia
- National Institute for Infectious Diseases 'L. Spallanzani', IRCCS, Rome, Italy.,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| |
Collapse
|
23
|
Ripa M, Chiappetta S, Tambussi G. Immunosenescence and hurdles in the clinical management of older HIV-patients. Virulence 2017; 8:508-528. [PMID: 28276994 DOI: 10.1080/21505594.2017.1292197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
People living with HIV (PLWH) who are treated with effective highly active antiretroviral therapy (HAART) have a similar life expectancy to the general population. Moreover, an increasing proportion of new HIV diagnoses are made in people older than 50 y. The number of older HIV-infected patients is thus constantly growing and it is expected that by 2030 around 70% of PLWH will be more than 50 y old. On the other hand, HIV infection itself is responsible for accelerated immunosenescence, a progressive decline of immune system function in both the adaptive and the innate arm, which impairs the ability of an individual to respond to infections and to give rise to long-term immunity; furthermore, older patients tend to have a worse immunological response to HAART. In this review we focus on the pathogenesis of HIV-induced immunosenescence and on the clinical management of older HIV-infected patients.
Collapse
Affiliation(s)
- Marco Ripa
- a Department of Infectious and Tropical Diseases , Ospedale San Raffaele , Milan , Italy
| | - Stefania Chiappetta
- a Department of Infectious and Tropical Diseases , Ospedale San Raffaele , Milan , Italy
| | - Giuseppe Tambussi
- a Department of Infectious and Tropical Diseases , Ospedale San Raffaele , Milan , Italy
| |
Collapse
|
24
|
Li Y, Shen C, Zhu B, Shi F, Eisen HN, Chen J. Persistent Antigen and Prolonged AKT-mTORC1 Activation Underlie Memory CD8 T Cell Impairment in the Absence of CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:1591-8. [PMID: 26163589 DOI: 10.4049/jimmunol.1500451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/17/2015] [Indexed: 01/11/2023]
Abstract
Recall responses by memory CD8 T cells are impaired in the absence of CD4 T cells. Although several mechanisms have been proposed, the molecular basis is still largely unknown. Using a local influenza virus infection in the respiratory tract and the lung of CD4(-/-) mice, we show that memory CD8 T cell impairment is limited to the lungs and the lung-draining lymph nodes, where viral Ags are unusually persistent and abundant in these mice. Persistent Ag exposure results in prolonged activation of the AKT-mTORC1 pathway in Ag-specific CD8 T cells, favoring their development into effector memory T cells at the expense of central memory T cells, and inhibition of mTORC1 by rapamycin largely corrects the impairment by promoting central memory T cell development. The findings suggest that the prolonged AKT-mTORC1 activation driven by persistent Ag is a critical mechanism underlying the impaired memory CD8 T cell development and responses in the absence of CD4 T cells.
Collapse
Affiliation(s)
- Yingzhong Li
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Chase Shen
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Bingdong Zhu
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Feng Shi
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Herman N Eisen
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jianzhu Chen
- Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
25
|
Abstract
Autophagy, a lysosomal degradative pathway that maintains cellular homeostasis, has emerged as an innate immune defense against pathogens. The role of autophagy in the deregulated HIV-infected central nervous system (CNS) is unclear. We have found that HIV-1-induced neuro-glial (neurons and astrocytes) damage involves modulation of the autophagy pathway. Neuro-glial stress induced by HIV-1 led to biochemical and morphological dysfunctions. X4 HIV-1 produced neuro-glial toxicity coupled with suppression of autophagy, while R5 HIV-1-induced toxicity was restricted to neurons. Rapamycin, a specific mTOR inhibitor (autophagy inducer) relieved the blockage of the autophagy pathway caused by HIV-1 and resulted in neuro-glial protection. Further understanding of the regulation of autophagy by cytokines and chemokines or other signaling events may lead to recognition of therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajeev Mehla
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ashok Chauhan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
26
|
Soliman A, Fathy A, Khashab S, Shaheen N, Soliman M. Sirolimus conversion may suppress viral replication in hepatitis C virus-positive renal transplant candidates. EXP CLIN TRANSPLANT 2014; 11:408-11. [PMID: 24128134 DOI: 10.6002/ect.2013.0017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Hepatitis C virus in renal transplant recipients is an independent risk factor for sickness and death. It has been shown that one might limit hepatitis C virus progression in liver transplant recipients with sirolimus-based immunosuppression. The mammalian target of rapamycin is an influential molecule for the anti-hepatitis C virus action of interferon. We report our experience with sirolimus conversion in hepatitis C virus-positive patients with chronic allograft nephropathy regarding hepatic and hematologic effects that might affect its future use. MATERIALS AND METHODS Twenty-five patients who had received renal transplants with anti-hepatitis C virus-positive and normal liver function were enrolled. Ten patients had allograft dysfunction because of cyclosporine nephrotoxicity. Sirolimus was initiated at 2 mg/d and adjusted to 6 to 8 ng/mL. Cyclosporine was gradually tapered and then stopped; 15 patients were used as a control group. Sirolimus-related hepatitis was defined as a rise in liver transferases or alkaline phosphatase or bilirubin over twice the upper limit of normal. Viral replication was defined as elevated liver enzymes and increasing viral load and/or biopsy-proven hepatitis C virus active hepatitis. RESULTS After conversion, there was a reduction of hemoglobin and hematocrit. In 1 patient, the immunosuppressive regimen was changed back to cyclosporine owing to anemia and hepatotoxicity leading to prompt return of hematocrit and liver enzymes to their original values. One of 10 antihepatitis C virus-positive patients (10.0%) developed sirolimus-associated hepatotoxicity, compared with 2 patients in the control group (13%). Sirolimus patients showed a significant decrease in the HCV PCR levels from 700 000 to 400 000 IU/mL; P < .001, compared to 680 000 to 660 000 IU/mL in cyclosporine patients; P = NS, with comparable levels of transaminases CONCLUSIONS Our data suggest that sirolimus has the potential to suppress viral replication in hepatitis C virus-positive renal transplant candidates.
Collapse
Affiliation(s)
- Amin Soliman
- Department of Nephrology, Cairo University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
27
|
Baccarani U, Righi E, Adani GL, Lorenzin D, Pasqualucci A, Bassetti M, Risaliti A. Pros and cons of liver transplantation in human immunodeficiency virus infected recipients. World J Gastroenterol 2014; 20:5353-5362. [PMID: 24833865 PMCID: PMC4017050 DOI: 10.3748/wjg.v20.i18.5353] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/05/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Before the introduction of combined highly active antiretroviral therapy, a positive human immunodeficiency virus (HIV) serological status represented an absolute contraindication for solid organ transplant (SOT). The advent of highly effective combined antiretroviral therapy in 1996 largely contributed to the increased demand for SOT in HIV-positive individuals due to increased patients’ life expectancy associated with the increasing prevalence of end-stage liver disease (ESLD). Nowadays, liver failure represents a frequent cause of mortality in the HIV-infected population mainly due to coinfection with hepatitis viruses sharing the same way of transmission. Thus, liver transplantation (LT) represents a reasonable approach in HIV patients with stable infection and ESLD. Available data presently supports with good evidence the practice of LT in the HIV-positive population. Thus, the issue is no longer “whether it is correct to transplant HIV-infected patients”, but “who are the patients who can be safely transplanted” and “when is the best time to perform LT”. Indeed, the benefits of LT in HIV-infected patients, especially in terms of mid- and long-term patient and graft survivals, are strictly related to the patients’ selection and to the correct timing for transplantation, especially when hepatitis C virus coinfection is present. Aim of this article is to review the pros and cons of LT in the cohort of HIV infected recipients.
Collapse
|
28
|
Passaes CP, Sáez-Cirión A. HIV cure research: advances and prospects. Virology 2014; 454-455:340-52. [PMID: 24636252 DOI: 10.1016/j.virol.2014.02.021] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/16/2022]
Abstract
Thirty years after the identification of HIV, a cure for HIV infection is still to be achieved. Advances of combined antiretroviral therapy (cART) in recent years have transformed HIV infection into a chronic disease when treatment is available. However, in spite of the favorable outcomes provided by the newer therapies, cART is not curative and patients are at risk of developing HIV-associated disorders. Moreover, universal access to antiretroviral treatment is restricted by financial obstacles. This review discusses the most recent strategies that have been developed in the search for an HIV cure and to improve life quality of people living with HIV.
Collapse
Affiliation(s)
- Caroline P Passaes
- Unité de Régulation des Infections Rétrovirales, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France; CEA, Division of Immuno-Virology, iMETI/DSV, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Asier Sáez-Cirión
- Unité de Régulation des Infections Rétrovirales, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
29
|
Congly SE, Doucette KE, Coffin CS. Outcomes and management of viral hepatitis and human immunodeficiency virus co-infection in liver transplantation. World J Gastroenterol 2014; 20:414-424. [PMID: 24574710 PMCID: PMC3923016 DOI: 10.3748/wjg.v20.i2.414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 10/22/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Liver transplantation for human immunodeficiency virus (HIV) positive patients with viral hepatitis co-infection is increasingly offered in many North American and European liver transplant centers. Prior studies have demonstrated acceptable post-transplant outcomes and no increased risk of HIV complications in patients co-infected with hepatitis B virus (HBV). However, liver transplantation in HIV positive patients with hepatitis C virus (HCV) has poorer outcomes overall, requiring careful selection of candidates. This review aims to summarize the published literature on outcomes after transplant in HIV patients with HBV or HCV related end-stage liver disease and recommendations for management. In particular the pre-transplant factors impacting outcomes in HCV/HIV co-infected candidates and importance of multidisciplinary management will be discussed.
Collapse
|
30
|
Takatsuki M, Soyama A, Eguchi S. Liver transplantation for HIV/hepatitis C virus co-infected patients. Hepatol Res 2014; 44:17-21. [PMID: 23607831 DOI: 10.1111/hepr.12132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 04/07/2013] [Accepted: 04/07/2013] [Indexed: 12/26/2022]
Abstract
Since the introduction of antiretroviral therapy (ART) in the mid-1990s, AIDS-related death has been dramatically reduced, and hepatitis-C-virus (HCV)-related liver failure or hepatocellular carcinoma has currently become the leading cause of death in HIV/HCV co-infected patients. Liver transplantation may be one of the treatments of choices in such cases, but the indications for transplantation, perioperative management including both HIV and HCV treatments, immunosuppression and the prevention/treatment of infectious complications are all still topics of debate. With the improved understanding of the viral behaviors of both HIV and HCV and the development of novel strategies, especially to avoid drug interactions between ART and immunosuppression, liver transplantation has become a realistic treatment for HIV/HCV co-infected patients.
Collapse
Affiliation(s)
- Mitsuhisa Takatsuki
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | |
Collapse
|
31
|
Stosor V. Organ Transplantation in HIV Patients: Current Status and New Directions. Curr Infect Dis Rep 2013; 15:526-35. [PMID: 24142801 DOI: 10.1007/s11908-013-0381-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combination antiretroviral therapy has resulted in longer life expectancies in persons living with HIV; however, end organ disease and death from organ failure have become growing issues for this population. With effective therapies for viral suppression, HIV is no longer considered an absolute contraindication to organ transplantation. Over the past decade, studies of transplantation in patients with HIV have had encouraging results such that patients with organ failure are pursuing transplantation. This review focuses on the current status of organ transplantation for HIV-infected persons.
Collapse
Affiliation(s)
- Valentina Stosor
- Divisions of Infectious Diseases and Organ Transplantation and Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, 645 North Michigan Avenue, Suite 900, Chicago, IL, 60611, USA,
| |
Collapse
|
32
|
Eguchi S, Takatsuki M, Kuroki T. Liver transplantation for patients with human immunodeficiency virus and hepatitis C virus co-infection: update in 2013. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2013; 21:263-8. [PMID: 24027085 DOI: 10.1002/jhbp.31] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Because of the progress of anti-retroviral therapy (ART) for human immunodeficiency virus (HIV), mortality due to opportunistic infection resulting in AIDS has been remarkably reduced. However, meanwhile, half of those patients have died of end-stage liver cirrhosis due to hepatitis C virus (HCV) with liver cirrhosis and early occurrence of hepatocellular carcinoma. Recently, in 2013, non-cirrhotic portal hypertension due to ART drugs or still unknown mechanisms have become problematic with early progression of the disease in this patient population. Liver transplantation (LT) could be one treatment of choice in such cases, but the indications for LT perioperative management, including both HIV and HCV treatments and immunosuppression, are still challenging. In this review, we update the literature on HIV/HCV co-infection and LT as well as recent effort for modifying allocation system for those patients.
Collapse
Affiliation(s)
- Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | | | | |
Collapse
|
33
|
Abstract
Systemic chronic immune activation is considered today as the driving force of CD4(+) T-cell depletion and acquired immunodeficiency syndrome (AIDS). A residual chronic immune activation persists even in HIV-infected patients in which viral replication is successfully inhibited by anti-retroviral therapy, with the extent of this residual immune activation being associated with CD4(+) T-cell loss. Unfortunately, the causal link between chronic immune activation and CD4(+) T-cell loss has not been formally established. This article provides first a brief historical overview on how the perception of the causative role of immune activation has changed over the years and lists the different kinds of immune activation characteristic of human immunodeficiency virus (HIV) infection. The mechanisms proposed to explain the chronic immune activation are multiple and are enumerated here, as well as the mechanisms proposed on how chronic immune activation could lead to AIDS. In addition, we summarize the lessons learned from natural hosts that know how to 'show AIDS the door', and discuss how these studies informed the design of novel immune modulatory interventions that are currently being tested. Finally, we review the current approaches aimed at targeting chronic immune activation and evoke future perspectives.
Collapse
Affiliation(s)
- Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30329, USA.
| | | |
Collapse
|
34
|
Di Benedetto F, Tarantino G, Ercolani G, Baccarani U, Montalti R, De Ruvo N, Berretta M, Adani GL, Zanello M, Tavio M, Cautero N, Tirelli U, Pinna AD, Gerunda GE, Guaraldi G. Multicenter italian experience in liver transplantation for hepatocellular carcinoma in HIV-infected patients. Oncologist 2013; 18:592-9. [PMID: 23666950 DOI: 10.1634/theoncologist.2012-0255] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The aim of our work is to assess the clinical outcomes of liver transplantation (LT) for hepatocellular carcinoma (HCC) in HIV-coinfected patients. This is a multicenter study involving three Italian transplant centers in northern Italy: University of Modena, University of Bologna, and University of Udine. PATIENTS AND METHODS We compared 30 HIV-positive patients affected by HCC who underwent LT with 125 HIV-uninfected patients who received the same treatment from September 2004 to June 2009. At listing, there were no differences between HIV-infected and -uninfected patients regarding HCC features. Patients outside the University of California, San Francisco criteria (UCSF) were considered eligible for LT if a down-staging program permitted a reduction of tumor burden. RESULTS HIV-infected patients were younger, they were more frequently anti-HCV positive, and a higher number of HIV-infected patients presented a coinfection HBV-HCV. Pre-LT treatments (liver resection and or locoregional treatments) were similar between the two groups. Histological characteristics of the tumor were similar in patients with and without HIV infection. No differences were observed in terms of overall survival and HCC recurrence rates. CONCLUSION LT for HCC is a feasible procedure and the presence of HIV does not particularly affect the post-LT outcome.
Collapse
Affiliation(s)
- Fabrizio Di Benedetto
- Liver and Multivisceral Transplant Center, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rapamycin-induced modulation of miRNA expression is associated with amelioration of HIV-associated nephropathy (HIVAN). Exp Cell Res 2013; 319:2073-2080. [PMID: 23611955 DOI: 10.1016/j.yexcr.2013.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/12/2013] [Accepted: 04/14/2013] [Indexed: 02/06/2023]
Abstract
Recent studies suggested that miRNAs are involved in the development of the pathogenesis of HIV-associated nephropathy (HIVAN). Rapamycin, a widely used mTOR inhibitor, has been demonstrated to slow down the progression of HIVAN. However, the role of miRNA in the regulation of these processes has not been investigated so far. In the current study, we have used a microarray-based approach in combination with real-time PCR to profile the miRNA expression patterns in rapamycin-treated HIVAN mice (Tg26). Our results demonstrated that 19 miRNAs belonging to 13 different families expressed differentially in renal tissues of rapamycin-receiving Tg26 mice when compared to Tg26 mice-receiving saline only. The patterns of miRNAs expression in rapamycin-receiving Tg26 mice took a reverse turn. These miRNAs were classified into 8 functional categories. In in vitro studies, we examined the expression of specific miRNAs in HIV-1 transduced human podocytes (HIV/HPs). HIV/HPs displayed attenuation of expression of miR-99a, -100a, -199a and miR-200, whereas, rapamycin inhibited this effect of HIV. These findings suggest that rapamycin-mediated up-regulation of specific miRNAs could contribute to amelioration of renal lesions in HIVAN mice.
Collapse
|
36
|
Brennan DC, Aguado JM, Potena L, Jardine AG, Legendre C, Säemann MD, Mueller NJ, Merville P, Emery V, Nashan B. Effect of maintenance immunosuppressive drugs on virus pathobiology: evidence and potential mechanisms. Rev Med Virol 2012; 23:97-125. [PMID: 23165654 DOI: 10.1002/rmv.1733] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 09/07/2012] [Accepted: 09/20/2012] [Indexed: 12/11/2022]
Abstract
Recent evidence suggesting a potential anti-CMV effect of mTORis is of great interest to the transplant community. However, the concept of an immunosuppressant with antiviral properties is not new, with many accounts of the antiviral properties of several agents over the years. Despite these reports, to date, there has been little effort to collate the evidence into a fuller picture. This manuscript was developed to gather the evidence of antiviral activity of the agents that comprise a typical immunosuppressive regimen against viruses that commonly reactivate following transplant (HHV1 and 2, VZV, EBV, CMV and HHV6, 7, and 8, HCV, HBV, BKV, HIV, HPV, and parvovirus). Appropriate immunosuppressive regimens posttransplant that avoid acute rejection while reducing risk of viral reactivation are also reviewed. The existing literature was disparate in nature, although indicating a possible stimulatory effect of tacrolimus on BKV, potentiation of viral reactivation by steroids, and a potential advantage of mammalian target of rapamycin (mTOR) inhibition in several viral infections, including BKV, HPV, and several herpesviruses.
Collapse
|
37
|
Joo DJ, Kim SH, Huh KH, Kim YS. Verruca Eradication Following Conversion to Sirolimus in a Renal Transplant Recipient after Longstanding Cyclosporine A Treatment. KOREAN JOURNAL OF TRANSPLANTATION 2012. [DOI: 10.4285/jkstn.2012.26.3.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Dong Jin Joo
- Department of Surgery, Yonsei University Health System, Seoul, Korea
- Research Institute for Transplantation, Yonsei University Health System, Seoul, Korea
| | - Sung Hoon Kim
- Department of Surgery, Yonsei University Health System, Seoul, Korea
| | - Kyu Ha Huh
- Department of Surgery, Yonsei University Health System, Seoul, Korea
- Research Institute for Transplantation, Yonsei University Health System, Seoul, Korea
| | - Yu Seun Kim
- Department of Surgery, Yonsei University Health System, Seoul, Korea
- Research Institute for Transplantation, Yonsei University Health System, Seoul, Korea
| |
Collapse
|
38
|
Rai P, Plagov A, Kumar D, Pathak S, Ayasolla KR, Chawla AK, Mathieson PW, Saleem MA, Husain M, Malhotra A, Singhal PC. Rapamycin-induced modulation of HIV gene transcription attenuates progression of HIVAN. Exp Mol Pathol 2012; 94:255-61. [PMID: 23010541 DOI: 10.1016/j.yexmp.2012.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/15/2012] [Indexed: 11/15/2022]
Abstract
HIV-associated nephropathy (HIVAN) is the manifestation of HIV gene expression by kidney cells in the presence of specific host factors. Recently, rapamycin (sirolimus) has been demonstrated to modulate the progression of HIVAN. We hypothesized that rapamycin would modulate the progression of HIVAN by attenuating HIV gene expression. To test our hypothesis, three weeks old Tg26 mice (n=6) were administered either vehicle or rapamycin (5 mg/kg, every other day, intraperitoneal) for eight weeks. At the end of the experimental period, the kidneys were harvested. In in vitro studies, human podocytes were transduced with either HIV-1 (NL4-3) or empty vector (EV), followed by treatment with either vehicle or rapamycin. Total RNA and proteins were extracted from renal tissues/cellular lysates and HIV gene transcription/translation was measured by real time PCR and Western blotting studies. Renal histological slides were graded for glomerular sclerosis and tubular dilatation with microcyst formation. Rapamycin attenuated both glomerular and tubular lesions in Tg26 mice. Rapamycin decreased transcription of HIV genes both in renal tissues as well as in HIV-1 transduced podocytes. Our data strongly indicate that HIV-1 long terminal repeat-mediated transcriptional activity was targeted by rapamycin. Rapamycin enhanced podocyte NF-κB and CREB activities but then it decreased AP-1 binding activity. Since expression of HIV genes by kidney cells has been demonstrated to be the key factor in the development HIVAN, it appears that rapamycin-induced altered transcription of HIV genes might have partly contributed to its disease modulating effects.
Collapse
Affiliation(s)
- Partab Rai
- Department of Medicine, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sun Y, Chen X, Zhao J, Zou X, Li G, Li X, Shen B, Sun S. Combined use of rapamycin and leflunomide in prevention of acute cardiac allografts rejection in rats. Transpl Immunol 2012; 27:19-24. [DOI: 10.1016/j.trim.2012.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/31/2012] [Accepted: 04/08/2012] [Indexed: 01/11/2023]
|
40
|
Yadav A, Kumar D, Salhan D, Rattanavich R, Maheshwari S, Adabala M, Ding G, Singhal PC. Sirolimus modulates HIVAN phenotype through inhibition of epithelial mesenchymal transition. Exp Mol Pathol 2012; 93:173-81. [PMID: 22579465 DOI: 10.1016/j.yexmp.2012.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 04/26/2012] [Accepted: 04/26/2012] [Indexed: 10/28/2022]
Abstract
HIV-associated nephropathy (HIVAN) is characterized by proliferative phenotype in the form of collapsing glomerulopathy and microcystic dilatation of tubules. Recently, epithelial mesenchymal transition (EMT) of renal cells has been demonstrated to contribute to the pathogenesis of proliferative HIVAN phenotype. We hypothesized that sirolimus would modulate HIVAN phenotype by attenuating renal cell EMT. In the present study, we evaluated the effect of sirolimus on the development of renal cell EMT as well as on display of HIVAN phenotype in a mouse model of HIVAN (Tg26). Tg26 mice receiving normal saline (TgNS) showed enhanced proliferation of both glomerular and tubular cells when compared to control mice-receiving normal saline (CNS); on the other hand, Tg26 mice receiving sirolimus (TgS) showed attenuated renal cell proliferation when compared with TgNS. TgNS also showed increased number of α-SMA-, vimentin-, and FSP1-positive cells (glomerular as well as tubular) when compared with CNS; however, TgS showed reduced number of SMA, vimentin, and FSP1+ve renal cells when compared to TgNS. Interestingly, sirolimus preserved renal epithelial cell expression of E-cadherin in TgS. Since sirolimus attenuated renal cell ZEB expression (a repressor of E-cadherin transcription), it appears that sirolimus may be attenuating renal cell EMT by preserving epithelial cell E-cadherin expression.
Collapse
Affiliation(s)
- Anju Yadav
- Immunology Center, Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY 11030, United States
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Successful transplant outcomes require optimal patient selection and timing. This review will update clinicians with current status and challenges in liver transplantation. Currently, the major limitation facing liver transplant centers is the shortage of organs. The limited availability of organs has led to long waiting periods for liver transplantation and consequently many patients become seriously ill or die while on the waiting list. RECENT FINDINGS This has major implications in the selection of patients, as well as the timing of transplant, for optimal use of these scarce organs. Indications and contraindications have changed slightly over the years and will be reviewed in this article. SUMMARY Timing for transplantation has changed more dramatically in the recent years because major changes to organ allocation systems have been undertaken to provide clinicians with a better way to prioritize patients for liver transplant.
Collapse
|
42
|
Abstract
In recent years, the technology of constructing chimeric mice with humanized immune systems has markedly improved. Multiple lineages of human immune cells develop in immunodeficient mice that have been transplanted with human hematopoietic stem cells. More importantly, these mice mount functional humoral and cellular immune responses upon immunization or microbial infection. Human immunodeficiency virus type I (HIV-1) can establish an infection in humanized mice, resulting in CD4(+) T-cell depletion and an accompanying nonspecific immune activation, which mimics the immunopathology in HIV-1-infected human patients. This makes humanized mice an optimal model for studying the mechanisms of HIV-1 immunopathogenesis and for developing novel immune-based therapies.
Collapse
|
43
|
Rehman S, Husain M, Yadav A, Kasinath BS, Malhotra A, Singhal PC. HIV-1 promotes renal tubular epithelial cell protein synthesis: role of mTOR pathway. PLoS One 2012; 7:e30071. [PMID: 22253885 PMCID: PMC3253808 DOI: 10.1371/journal.pone.0030071] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 12/13/2011] [Indexed: 01/01/2023] Open
Abstract
Tubular cell HIV-infection has been reported to manifest in the form of cellular hypertrophy and apoptosis. In the present study, we evaluated the role of mammalian target of rapamycin (mTOR) pathway in the HIV induction of tubular cell protein synthesis. Mouse proximal tubular epithelial cells (MPTECs) were transduced with either gag/pol-deleted NL4-3 (HIV/MPTEC) or empty vector (Vector/MPTEC). HIV/MPTEC showed enhanced DNA synthesis when compared with Vector/MPTECs by BRDU labeling studies. HIV/MPTECs also showed enhanced production of β-laminin and fibronection in addition to increased protein content per cell. In in vivo studies, renal cortical sections from HIV transgenic mice and HIVAN patients showed enhanced tubular cell phosphorylation of mTOR. Analysis of mTOR revealed increased expression of phospho (p)-mTOR in HIV/MPTECs when compared to vector/MPTECs. Further downstream analysis of mTOR pathway revealed enhanced phosphorylation of p70S6 kinase and associated diminished phosphorylation of eEF2 (eukaryotic translation elongation factor 2) in HIV/MPTECs; moreover, HIV/MPTECs displayed enhanced phosphorylation of eIF4B (eukaryotic translation initiation factor 4B) and 4EBP-1 (eukaryotic 4E binding protein). To confirm our hypothesis, we evaluated the effect of rapamycin on HIV-induced tubular cell downstream signaling. Rapamycin not only attenuated phosphorylation of p70S6 kinase and associated down stream signaling in HIV/MPTECs but also inhibited HIV-1 induced tubular cell protein synthesis. These findings suggest that mTOR pathway is activated in HIV-induced enhanced tubular cell protein synthesis and contributes to tubular cell hypertrophy.
Collapse
Affiliation(s)
- Shabina Rehman
- Department of Medicine, North Shore LIJ Health System, New York, New York, United States of America
| | - Mohammad Husain
- Department of Medicine, North Shore LIJ Health System, New York, New York, United States of America
| | - Anju Yadav
- Department of Medicine, North Shore LIJ Health System, New York, New York, United States of America
| | - Balakuntalam S. Kasinath
- Department of Medicine, Texas Health Science Center, San Antonio, Texas, United States of America
| | - Ashwani Malhotra
- Department of Medicine, North Shore LIJ Health System, New York, New York, United States of America
| | - Pravin C. Singhal
- Department of Medicine, North Shore LIJ Health System, New York, New York, United States of America
| |
Collapse
|
44
|
Veroux M, Tallarita T, Corona D, D’Assoro A, Gurrieri C, Veroux P. Sirolimus in solid organ transplantation: current therapies and new frontiers. Immunotherapy 2011; 3:1487-97. [DOI: 10.2217/imt.11.143] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
45
|
|
46
|
Campbell GR, Spector SA. Hormonally active vitamin D3 (1alpha,25-dihydroxycholecalciferol) triggers autophagy in human macrophages that inhibits HIV-1 infection. J Biol Chem 2011; 286:18890-902. [PMID: 21454634 PMCID: PMC3099705 DOI: 10.1074/jbc.m110.206110] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/29/2011] [Indexed: 01/08/2023] Open
Abstract
Autophagy is a self-digestion pathway essential for maintaining cellular homeostasis and cell survival and for degrading intracellular pathogens. Human immunodeficiency virus-1 (HIV-1) may utilize autophagy for replication as the autophagy-related protein-7 (ATG-7), microtubule-associated protein 1 light chain 3, ATG-12, and ATG-16L2 are required for productive HIV-1 infection; however, the effects of autophagy induction on HIV-1 infection are unknown. HIV-1-infected individuals have lower levels of 1α,25-dihydroxycholecalciferol, the hormonally active form of vitamin D, than uninfected individuals. with the lowest concentrations found in persons with AIDS. Using human macrophages and RNA interference for ATG-5 and Beclin-1 and chemical inhibition of phosphatidylinositol 3-kinase, we have found that physiologically relevant concentrations of 1α,25-dihydroxycholecalciferol induce autophagy in human macrophages through a phosphatidylinositol 3-kinase-, ATG-5-, and Beclin-1-dependent mechanism that significantly inhibits HIV-1 replication in a dose-dependent manner. We also show that the inhibition of basal autophagy inhibits HIV-1 replication. Furthermore, although 1α,25-dihydroxycholecalciferol induces the secretion of human cathelicidin, at the concentrations produced in vitro, cathelicidin does not trigger autophagy. Our findings support an important role for autophagy during HIV-1 infection and provide new insights into novel approaches to prevent and treat HIV-1 infection and related opportunistic infections.
Collapse
Affiliation(s)
- Grant R. Campbell
- From the Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California 92093 and
| | - Stephen A. Spector
- From the Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California 92093 and
- Rady Children's Hospital, San Diego, California 92123
| |
Collapse
|
47
|
Nicoletti F, Fagone P, Meroni P, McCubrey J, Bendtzen K. mTOR as a multifunctional therapeutic target in HIV infection. Drug Discov Today 2011; 16:715-21. [PMID: 21624501 DOI: 10.1016/j.drudis.2011.05.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/03/2011] [Accepted: 05/11/2011] [Indexed: 12/31/2022]
Abstract
Patients undergoing long-term highly active antiretroviral therapy treatment are probably at a higher risk of various HIV-related complications. Hyperactivation of The mammalian target of rapamycin (mTOR) has been found to contribute to dysregulated apoptosis and autophagy which determine CD4(+)-T-cell loss, impaired function of innate immunity and development of neurocognitive disorders. Dysregulated mTOR activation has also been shown to play a key part in the development of nephropathy and in the pathogenesis of HIV-associated malignancies. These studies strongly support a multifunctional key role for mTOR in the pathogenesis of HIV-related disorders and suggest that specific mTOR inhibitors could represent a novel approach for the prevention and treatment of these pathologies.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Bio-Medical Sciences, School of Medicine, University of Catania, Italy.
| | | | | | | | | |
Collapse
|
48
|
Di Benedetto F, Tarantino G, De Ruvo N, Cautero N, Montalti R, Guerrini GP, Ballarin R, Spaggiari M, Smerieri N, Serra V, Rompianesi G, D'Amico G, Mimmo A, Iemmolo RM, Codeluppi M, Cocchi S, Guaraldi G, Gerunda GE. University of Modena experience in HIV-positive patients undergoing liver transplantation. Transplant Proc 2011; 43:1114-8. [PMID: 21620066 DOI: 10.1016/j.transproceed.2011.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Highly effective antiretroviral therapy in the last decade has increased the survival rates of HIV-positive patients, yielding a greater number of HIV patients suffering from liver-related disease. Liver transplantation (LT) is the only curative treatment for end-stage liver disease (ESLD) associated or not with hepatocellular carcinoma (HCC). PATIENTS AND METHODS From June 2003 to September 2010, 23 patients underwent cadaveric donor LT for ESLD at our institution. Inclusion criteria followed the Italian Protocol for LT in HIV-positive patients. Immunosuppressive regimens were based on cyclosporine or tacrolimus, eventually switched to Rapamycin. RESULTS The median CD4 T-cell count was 275/mmc (range=119-924). All patients were affected by ESLD, which was associated with HCC in 14 cases. Ten patients were within the Milan criteria and four patients exceeded them but were within the San Francisco criteria. Conversion from calcineurin inhibitors (CNI) to rapamycin occurred in ten cases. Hepatitis C virus (HCV) recurrence occurred in 13/21 HCV-positive patients. Acute cellular rejection occurred in eight patients with one developing chronic cellular rejection. Overall patient and graft survivals at 80 months were 50% and 45% respectively. DISCUSSION LT in HIV-positive patients is a feasible procedure, even if in our experience was burdened by a greater incidence of complications including HCV recurrence and infection compared with HIV-negative patients.
Collapse
Affiliation(s)
- F Di Benedetto
- Liver and Multivisceral Transplant Center, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW To provide the nontransplant clinician with a basic understanding of the liver transplant process. RECENT FINDINGS Since its inception, the technique of liver transplantation and patient management has evolved considerably. We present an up-to-date overview of the evaluation of the transplant recipient and the listing and timing of transplant. We conclude with a brief summary of long-term complications, which should be considered when caring for the posttransplant patient. SUMMARY Liver transplantation is the only definitive treatment option for patients dying of liver failure. The growing population of patients with liver disease means that more transplants will be performed. As these patients now live longer lives, it is crucial that clinicians have a basic understanding of the process and outcomes.
Collapse
|
50
|
d'Ettorre G, Paiardini M, Ceccarelli G, Silvestri G, Vullo V. HIV-associated immune activation: from bench to bedside. AIDS Res Hum Retroviruses 2011; 27:355-64. [PMID: 21309730 DOI: 10.1089/aid.2010.0342] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
HIV infection is associated with a state of chronic, generalized immune activation that has been shown in many studies to be a key predictor of progression to AIDS. Consistent with this model, nonpathogenic SIV infections of natural hosts, such as the sooty mangabeys, are characterized by low levels of immune activation during the chronic phase of infection. The molecular, cellular, and pathophysiological mechanisms underlying the HIV-associated immune activation are complex and still poorly understood. There is, however, growing consensus that both viral and host factors contribute to this phenotype, with emphasis on the role played by the mucosal immune dysfunction (and consequent microbial translocation) as well as the pattern of in vivo-infected CD4(+) T cells. The observation that antiretroviral therapy (ART)-induced suppression of HIV replication does not fully resolve immune activation provided the rationale for a number of exploratory studies of potential immune modulatory treatments to be used in HIV-infected individuals in addition to standard ART. This review provides an update on the causes and consequences of the HIV-associated immune activation, and a summary of the immune modulatory approaches that are currently under clinical investigation.
Collapse
Affiliation(s)
- Gabriella d'Ettorre
- Department of Hygiene, Public Health and Infectious Diseases, “Sapienza” University of Rome, Rome, Italy
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Giancarlo Ceccarelli
- Department of Hygiene, Public Health and Infectious Diseases, “Sapienza” University of Rome, Rome, Italy
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Vincenzo Vullo
- Department of Hygiene, Public Health and Infectious Diseases, “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|