1
|
Passaes C, Desjardins D, Chapel A, Monceaux V, Lemaitre J, Mélard A, Perdomo-Celis F, Planchais C, Gourvès M, Dimant N, David A, Dereuddre-Bosquet N, Barrail-Tran A, Gouget H, Guillaume C, Relouzat F, Lambotte O, Guedj J, Müller-Trutwin M, Mouquet H, Rouzioux C, Avettand-Fenoël V, Le Grand R, Sáez-Cirión A. Early antiretroviral therapy favors post-treatment SIV control associated with the expansion of enhanced memory CD8 + T-cells. Nat Commun 2024; 15:178. [PMID: 38212337 PMCID: PMC10784587 DOI: 10.1038/s41467-023-44389-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
HIV remission can be achieved in some people, called post-treatment HIV controllers, after antiretroviral treatment discontinuation. Treatment initiation close to the time of infection was suggested to favor post-treatment control, but the circumstances and mechanisms leading to this outcome remain unclear. Here we evaluate the impact of early (week 4) vs. late (week 24 post-infection) treatment initiation in SIVmac251-infected male cynomolgus macaques receiving 2 years of therapy before analytical treatment interruption. We show that early treatment strongly promotes post-treatment control, which is not related to a lower frequency of infected cells at treatment interruption. Rather, early treatment favors the development of long-term memory CD8+ T cells with enhanced proliferative and SIV suppressive capacity that are able to mediate a robust secondary-like response upon viral rebound. Our model allows us to formally demonstrate a link between treatment initiation during primary infection and the promotion of post-treatment control and provides results that may guide the development of new immunotherapies for HIV remission.
Collapse
Affiliation(s)
- Caroline Passaes
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France.
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France.
| | - Delphine Desjardins
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Anaïs Chapel
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Valérie Monceaux
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Julien Lemaitre
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Adeline Mélard
- Université Paris Cité; INSERM, U1016; CNRS, UMR8104, Paris, France
| | - Federico Perdomo-Celis
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Cyril Planchais
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Maël Gourvès
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
| | - Nastasia Dimant
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Annie David
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Aurélie Barrail-Tran
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, Service de Pharmacie, Le Kremlin Bicêtre, France
| | - Hélène Gouget
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Céline Guillaume
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Olivier Lambotte
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
- Université Paris-Saclay, AP-HP. Hôpital Bicêtre, Clinical Immunology Department, 94270, Le Kremlin Bicêtre, France
| | - Jérémie Guedj
- Université Paris Cité, IAME, INSERM, F-75018, Paris, France
| | - Michaela Müller-Trutwin
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Christine Rouzioux
- Université Paris Cité/APHP Hôpital Necker - Enfants Malades, Paris, France
| | - Véronique Avettand-Fenoël
- Université Paris Cité; INSERM, U1016; CNRS, UMR8104, Paris, France
- APHP Hôpital Cochin, Service de Virologie, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France.
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France.
| |
Collapse
|
2
|
Wu HL, Busman-Sahay K, Weber WC, Waytashek CM, Boyle CD, Bateman KB, Reed JS, Hwang JM, Shriver-Munsch C, Swanson T, Northrup M, Armantrout K, Price H, Robertson-LeVay M, Uttke S, Kumar MR, Fray EJ, Taylor-Brill S, Bondoc S, Agnor R, Junell SL, Legasse AW, Moats C, Bochart RM, Sciurba J, Bimber BN, Sullivan MN, Dozier B, MacAllister RP, Hobbs TR, Martin LD, Panoskaltsis-Mortari A, Colgin LMA, Siliciano RF, Siliciano JD, Estes JD, Smedley JV, Axthelm MK, Meyers G, Maziarz RT, Burwitz BJ, Stanton JJ, Sacha JB. Allogeneic immunity clears latent virus following allogeneic stem cell transplantation in SIV-infected ART-suppressed macaques. Immunity 2023; 56:1649-1663.e5. [PMID: 37236188 PMCID: PMC10524637 DOI: 10.1016/j.immuni.2023.04.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/30/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) from donors lacking C-C chemokine receptor 5 (CCR5Δ32/Δ32) can cure HIV, yet mechanisms remain speculative. To define how alloHSCT mediates HIV cure, we performed MHC-matched alloHSCT in SIV+, anti-retroviral therapy (ART)-suppressed Mauritian cynomolgus macaques (MCMs) and demonstrated that allogeneic immunity was the major driver of reservoir clearance, occurring first in peripheral blood, then peripheral lymph nodes, and finally in mesenteric lymph nodes draining the gastrointestinal tract. While allogeneic immunity could extirpate the latent viral reservoir and did so in two alloHSCT-recipient MCMs that remained aviremic >2.5 years after stopping ART, in other cases, it was insufficient without protection of engrafting cells afforded by CCR5-deficiency, as CCR5-tropic virus spread to donor CD4+ T cells despite full ART suppression. These data demonstrate the individual contributions of allogeneic immunity and CCR5 deficiency to HIV cure and support defining targets of alloimmunity for curative strategies independent of HSCT.
Collapse
Affiliation(s)
- Helen L Wu
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Kathleen Busman-Sahay
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Whitney C Weber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Courtney M Waytashek
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Carla D Boyle
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Katherine B Bateman
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Jason S Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Joseph M Hwang
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Christine Shriver-Munsch
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Tonya Swanson
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Mina Northrup
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Kimberly Armantrout
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Heidi Price
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Mitch Robertson-LeVay
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Samantha Uttke
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Mithra R Kumar
- Department of Medicine and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Emily J Fray
- Department of Medicine and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Sol Taylor-Brill
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Stephen Bondoc
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Rebecca Agnor
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephanie L Junell
- Division of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alfred W Legasse
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Rachele M Bochart
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Joseph Sciurba
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Benjamin N Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Michelle N Sullivan
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Brandy Dozier
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Rhonda P MacAllister
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Theodore R Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Lauren D Martin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55454, USA
| | - Lois M A Colgin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Robert F Siliciano
- Department of Medicine and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Janet D Siliciano
- Department of Medicine and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jacob D Estes
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Jeremy V Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Gabrielle Meyers
- Division of Blood and Marrow Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Richard T Maziarz
- Division of Blood and Marrow Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Benjamin J Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Jeffrey J Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA
| | - Jonah B Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97007, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97007, USA.
| |
Collapse
|
3
|
Sawaswong V, Chanchaem P, Kemthong T, Warit S, Chaiprasert A, Malaivijitnond S, Payungporn S. Alteration of gut microbiota in wild-borne long-tailed macaques after 1-year being housed in hygienic captivity. Sci Rep 2023; 13:5842. [PMID: 37037869 PMCID: PMC10085984 DOI: 10.1038/s41598-023-33163-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023] Open
Abstract
The wild-born long-tailed macaques (Macaca fascicularis) were recently recruited and used as breeders for the National Primate Research Center of Thailand, Chulalongkorn University (NPRCT-CU), and changes in their in-depth gut microbiota profiles were investigated. The Oxford Nanopore Technology (ONT) was used to explore full-length 16S rDNA sequences of gut microbiota in animals once captured in their natural habitat and 1-year following translocation and housing in a hygienic environment at NPRCT-CU. Our findings show that the gut microbiota of macaques after 1 year of hygienic housing and programmed diets feeding was altered and reshaped. The prevalent gut bacteria such as Prevotella copri and Faecalibacterium prausnitzii were enriched after translocation, causing the lower alpha diversity. The correlation analysis revealed that Prevotella copri, Phascolarctobacterium succinatutens, and Prevotella stercorea, showed a positive correlation with each other. Significantly enriched pathways in the macaques after translocation included biosynthesis of essential amino acids, fatty acids, polyamine and butanoate. The effects of microbiota change could help macaques to harvest the energy from programmed diets and adapt their gut metabolism. The novel probiotics and microbiota engineering approach could be further developed based on the current findings and should be helpful for captive animal health care management.
Collapse
Affiliation(s)
- Vorthon Sawaswong
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Patumwan, Bangkok, 10330, Thailand
- Nucleic Acid Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Prangwalai Chanchaem
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Patumwan, Bangkok, 10330, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand, Chulalongkorn University, Saraburi, 18110, Thailand
| | - Saradee Warit
- Industrial Tuberculosis Team, Industrial Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Angkana Chaiprasert
- Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand, Chulalongkorn University, Saraburi, 18110, Thailand
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sunchai Payungporn
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Patumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
4
|
Warne RK, Moloney GK, Chaber AL. Is biomedical research demand driving a monkey business? One Health 2023. [DOI: 10.1016/j.onehlt.2023.100520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
5
|
Jiang C, Pan X, Luo J, Liu X, Zhang L, Liu Y, Lei G, Hu G, Li J. Alterations in Microbiota and Metabolites Related to Spontaneous Diabetes and Pre-Diabetes in Rhesus Macaques. Genes (Basel) 2022; 13:genes13091513. [PMID: 36140683 PMCID: PMC9498908 DOI: 10.3390/genes13091513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Spontaneous type 2 diabetes mellitus (T2DM) macaques are valuable resources for our understanding the pathological mechanism of T2DM. Based on one month’s fasting blood glucose survey, we identified seven spontaneous T2DM macaques and five impaired glucose regulation (IGR) macaques from 1408 captive individuals. FPG, HbA1c, FPI and IR values were significant higher in T2DM and IGR than in controls. 16S rRNA sequencing of fecal microbes showed the significantly greater abundance of Oribacterium, bacteria inhibiting the production of secondary bile acids, and Phascolarctobacterium, bacteria producing short-chain fatty acids was significantly lower in T2DM macaques. In addition, several opportunistic pathogens, such as Mogibacterium and Kocuria were significantly more abundant in both T2DM and IGR macaques. Fecal metabolites analysis based on UHPLC-MS identified 50 differential metabolites (DMs) between T2DM and controls, and 26 DMs between IGR and controls. The DMs were significantly enriched in the bile acids metabolism, fatty acids metabolism and amino acids metabolism pathways. Combining results from physiochemical parameters, microbiota and metabolomics, we demonstrate that the imbalance of gut microbial community leading to the dysfunction of glucose, bile acids, fatty acids and amino acids metabolism may contribute to the hyperglycaemia in macaques, and suggest several microbes and metabolites are potential biomarkers for T2DM and IGR macaques.
Collapse
Affiliation(s)
- Cong Jiang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Xuan Pan
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Jinxia Luo
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Xu Liu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Lin Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Yun Liu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Guanglun Lei
- SCU-SGHB Joint Laboratory on Non-Human Primates Research, Sichuan Green-House Biotech Co., Ltd., Meishan 620000, China
| | - Gang Hu
- SCU-SGHB Joint Laboratory on Non-Human Primates Research, Sichuan Green-House Biotech Co., Ltd., Meishan 620000, China
| | - Jing Li
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
- Correspondence:
| |
Collapse
|
6
|
Allogeneic MHC-matched T-cell receptor α/β-depleted bone marrow transplants in SHIV-infected, ART-suppressed Mauritian cynomolgus macaques. Sci Rep 2022; 12:12345. [PMID: 35853970 PMCID: PMC9296477 DOI: 10.1038/s41598-022-16306-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/07/2022] [Indexed: 11/08/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplants (allo-HSCTs) dramatically reduce HIV reservoirs in antiretroviral therapy (ART) suppressed individuals. However, the mechanism(s) responsible for these post-transplant viral reservoir declines are not fully understood. Therefore, we modeled allo-HSCT in ART-suppressed simian-human immunodeficiency virus (SHIV)-infected Mauritian cynomolgus macaques (MCMs) to illuminate factors contributing to transplant-induced viral reservoir decay. Thus, we infected four MCMs with CCR5-tropic SHIV162P3 and started them on ART 6-16 weeks post-infection (p.i.), maintaining continuous ART during myeloablative conditioning. To prevent graft-versus-host disease (GvHD), we transplanted allogeneic MHC-matched α/β T cell-depleted bone marrow cells and prophylactically treated the MCMs with cyclophosphamide and tacrolimus. The transplants produced ~ 85% whole blood donor chimerism without causing high-grade GvHD. Consequently, three MCMs had undetectable SHIV DNA in their blood post-transplant. However, SHIV-harboring cells persisted in various tissues, with detectable viral DNA in lymph nodes and tissues between 38 and 62 days post-transplant. Further, removing one MCM from ART at 63 days post-transplant resulted in SHIV rapidly rebounding within 7 days of treatment withdrawal. In conclusion, transplanting SHIV-infected MCMs with allogeneic MHC-matched α/β T cell-depleted bone marrow cells prevented high-grade GvHD and decreased SHIV-harboring cells in the blood post-transplant but did not eliminate viral reservoirs in tissues.
Collapse
|
7
|
Kashem MA, Li L, Yuan XY, Plummer FA, Luo M. Toll-like Interleukin -1 Receptor Regulator (TILRR) Protein, a Major Modulator of Inflammation, is Expressed in Normal Human and Macaque Tissues and PBMCs. J Inflamm Res 2022; 15:2925-2937. [PMID: 35592073 PMCID: PMC9113122 DOI: 10.2147/jir.s357866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose TILRR is a modulator of genes in the NF-κB inflammation pathway. It regulates inflammation-responsive genes, the secretion of inflammatory mediators, and the migration of immune cells. Because inflammation drives the pathogenesis of many infectious and inflammatory diseases, it is important to know the expression of TILRR protein in tissues and cells. This study examined TILRR protein expression in healthy adult human and macaques’ tissues and PBMCs (peripheral blood mononuclear cells). Methods and Results Tissues (trachea, lungs, stomach, small intestine [ileum], cecum, colon, rectum, vagina, cervix, uterus, and penis) and PBMCs from humans and macaques were lysed in RIPA (radioimmunoprecipitation assay) lysis buffer. The TILRR protein was examined by fluorescent Western blot analysis. The relative fluorescence units (rfu) of TILRR protein expression were quantified by Image Studio software (LI-COR). The results showed that adult healthy female (n=1) rectal and cervicovaginal tissues expressed a higher level of TILRR protein than the other tissues (trachea, lungs, stomach, small intestine [ileum], cecum, colon, uterus, and penis) examined. Like humans, the lungs, colon, and rectal tissues of healthy adult female cynomolgus monkeys (Macaca fascicularis) (n=2) expressed the TILRR protein. In addition, PBMCs of healthy adult women (n=4), adult female cynomolgus monkeys (Macaca fascicularis) (n=4), and adult male and female rhesus monkeys (Macaca mulatta) (n=4) showed a similar expression level of TILRR protein (p= 0.2858). TILRR protein was not detected in most of the human cell lines examined, except in Jurkat cells. Conclusion Our study for the first time showed that TILRR protein is expressed in healthy adult human and monkey tissues and PBMCs. The TILRR protein in these tissues and PBMCs may play a role in the inflammatory response of these tissues and cells in response to infectious pathogens.
Collapse
Affiliation(s)
- Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- JC Wilt Infectious Diseases Research Center, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Microbiology and Veterinary Public Health, Chittagong Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Lin Li
- JC Wilt Infectious Diseases Research Center, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Xin-Yong Yuan
- JC Wilt Infectious Diseases Research Center, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- JC Wilt Infectious Diseases Research Center, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Correspondence: Ma Luo, JC Wilt Infectious Diseases Research Center, National Microbiology Laboratory, 745 Logan Avenue, Winnipeg, MB, R3E 3L5, Canada, Tel +1 204-789-5072, Fax +1 204-789-2018, Email
| |
Collapse
|
8
|
Lu Y, Zhang MX, Pang W, Song TZ, Zheng HY, Tian RR, Zheng YT. Transcription Factor ZNF683 Inhibits SIV/HIV Replication through Regulating IFNγ Secretion of CD8+ T Cells. Viruses 2022; 14:v14040719. [PMID: 35458449 PMCID: PMC9030044 DOI: 10.3390/v14040719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary microbial invasion frequently occurs during AIDS progression in HIV patients. Inflammatory cytokines and other immunoregulatory factors play important roles in this process. We previously established an AIDS model of SIVmac239 infection in northern pig-tailed macaques (NPMs), which were divided into rapid progressor (RP) and slow progressor (SP) groups according to their AIDS progression rates. In this study, we performed 16S rDNA and transcriptome sequencing of the lungs to reveal the molecular mechanism underlying the difference in progression rate between the RPs and SPs. We found that microbial invasion in the RP group was distinct from that in the SP group, showing marker flora of the Family XI, Enterococcus and Ezakiella, and more Lactobacilli. Through pulmonary transcriptome analysis, we found that the transcription factor ZNF683 had higher expression in the SP group than in the RP group. In subsequent functional experiments, we found that ZNF683 increased the proliferation and IFNγ secretion ability of CD8+ T cells, thus decreasing SIV or HIV replication, which may be related to AIDS progression in SIVmac239-infected NPMs. This study helps elucidate the various complexities of disease progression in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Ying Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Ming-Xu Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Tian-Zhang Song
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Hong-Yi Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Ren-Rong Tian
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
- Correspondence: ; Tel.: +86-871-65295684
| |
Collapse
|
9
|
Optimal Maturation of the SIV-Specific CD8 + T Cell Response after Primary Infection Is Associated with Natural Control of SIV: ANRS SIC Study. Cell Rep 2021; 32:108174. [PMID: 32966788 DOI: 10.1016/j.celrep.2020.108174] [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] [Received: 12/27/2019] [Revised: 07/10/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
Highly efficient CD8+ T cells are associated with natural HIV control, but it has remained unclear how these cells are generated and maintained. We have used a macaque model of spontaneous SIVmac251 control to monitor the development of efficient CD8+ T cell responses. Our results show that SIV-specific CD8+ T cells emerge during primary infection in all animals. The ability of CD8+ T cells to suppress SIV is suboptimal in the acute phase but increases progressively in controller macaques before the establishment of sustained low-level viremia. Controller macaques develop optimal memory-like SIV-specific CD8+ T cells early after infection. In contrast, a persistently skewed differentiation phenotype characterizes memory SIV-specific CD8+ T cells in non-controller macaques. Accordingly, the phenotype of SIV-specific CD8+ T cells defined early after infection appears to favor the development of protective immunity in controllers, whereas SIV-specific CD8+ T cells in non-controllers fail to gain antiviral potency, feasibly as a consequence of early defects imprinted in the memory pool.
Collapse
|
10
|
Mao GP, Niu MH, Cui YH, Tang RL, Chen W, Liu B, He Z. Characterization, isolation, and culture of spermatogonial stem cells in Macaca fascicularis. Asian J Androl 2021; 23:240-248. [PMID: 33533740 PMCID: PMC8152426 DOI: 10.4103/aja.aja_95_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/02/2020] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) have great applications in both reproductive and regenerative medicine. Primates including monkeys are very similar to humans with regard to physiology and pathology. Nevertheless, little is known about the isolation, the characteristics, and the culture of primate SSCs. This study was designed to identify, isolate, and culture monkey SSCs. Immunocytochemistry was used to identify markers for monkey SSCs. Glial cell line-derived neurotrophic factor family receptor alpha-1 (GFRA1)-enriched spermatogonia were isolated from monkeys, namely Macaca fascicularis (M. fascicularis), by two-step enzymatic digestion and magnetic-activated cell sorting, and they were cultured on precoated plates in the conditioned medium. Reverse transcription-polymerase chain reaction (RT-PCR), immunocytochemistry, and RNA sequencing were used to compare phenotype and transcriptomes in GFRA1-enriched spermatogonia between 0 day and 14 days of culture, and xenotransplantation was performed to evaluate the function of GFRA1-enriched spermatogonia. SSCs shared some phenotypes with rodent and human SSCs. GFRA1-enriched spermatogonia with high purity and viability were isolated from M. fascicularis testes. The freshly isolated cells expressed numerous markers for rodent SSCs, and they were cultured for 14 days. The expression of numerous SSC markers was maintained during the cultivation of GFRA1-enriched spermatogonia. RNA sequencing reflected a 97.3% similarity in global gene profiles between 0 day and 14 days of culture. The xenotransplantation assay indicated that the GFRA1-enriched spermatogonia formed colonies and proliferated in vivo in the recipient c-KitW/W (W) mutant mice. Collectively, GFRA1-enriched spermatogonia are monkey SSCs phenotypically both in vitro and in vivo. This study suggests that monkey might provide an alternative to human SSCs for basic research and application in human diseases.
Collapse
Affiliation(s)
- Guo-Ping Mao
- Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Ming-Hui Niu
- Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ying-Hong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Rui-Ling Tang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Wei Chen
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Bang Liu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
11
|
Li H, Omange RW, Liang B, Toledo N, Hai Y, Liu LR, Schalk D, Crecente-Campo J, Dacoba TG, Lambe AB, Lim SY, Li L, Kashem MA, Wan Y, Correia-Pinto JF, Seaman MS, Liu XQ, Balshaw RF, Li Q, Schultz-Darken N, Alonso MJ, Plummer FA, Whitney JB, Luo M. Vaccine targeting SIVmac251 protease cleavage sites protects macaques against vaginal infection. J Clin Invest 2021; 130:6429-6442. [PMID: 32853182 DOI: 10.1172/jci138728] [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] [Received: 04/08/2020] [Accepted: 08/20/2020] [Indexed: 01/03/2023] Open
Abstract
After over 3 decades of research, an effective anti-HIV vaccine remains elusive. The recently halted HVTN702 clinical trial not only further stresses the challenge to develop an effective HIV vaccine but also emphasizes that unconventional and novel vaccine strategies are urgently needed. Here, we report that a vaccine focusing the immune response on the sequences surrounding the 12 viral protease cleavage sites (PCSs) provided greater than 80% protection to Mauritian cynomolgus macaques against repeated intravaginal SIVmac251 challenges. The PCS-specific T cell responses correlated with vaccine efficacy. The PCS vaccine did not induce immune activation or inflammation known to be associated with increased susceptibility to HIV infection. Machine learning analyses revealed that the immune microenvironment generated by the PCS vaccine was predictive of vaccine efficacy. Our study demonstrates, for the first time to our knowledge, that a vaccine which targets only viral maturation, but lacks full-length Env and Gag immunogens, can prevent intravaginal infection in a stringent macaque/SIV challenge model. Targeting HIV maturation thus offers a potentially novel approach to developing an effective HIV vaccine.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert W Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nikki Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yan Hai
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lewis R Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dane Schalk
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | - So-Yon Lim
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lin Li
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yanmin Wan
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jorge F Correia-Pinto
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiao Qing Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert F Balshaw
- Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Nancy Schultz-Darken
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Maria J Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| |
Collapse
|
12
|
Lin Z, Huang Y, Jiang H, Zhang D, Yang Y, Geng X, Li B. Functional differences and similarities in activated peripheral blood mononuclear cells by lipopolysaccharide or phytohemagglutinin stimulation between human and cynomolgus monkeys. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:257. [PMID: 33708884 PMCID: PMC7940909 DOI: 10.21037/atm-20-4548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background The monkey is a primary species used in toxicological research. However, the failures of preclinical studies to predict a life-threatening “cytokine storm”, which, for instance, rapidly occurred in six healthy volunteers with the CD28 superagonist monoclonal antibody (mAb) TGN1412 in the first-in-human phase I clinical trial, have emphasized a need to clarify the differences between human and monkey immune systems. Methods In the present study, we analyzed and compared the lymphocyte proliferation, cytokine secretion, and gene expression profiles after phytohemagglutinin (PHA) and lipopolysaccharide (LPS) stimulation of peripheral blood mononuclear cells (PBMCs) from three healthy humans and cynomolgus monkeys (Macaca fascicularis). Results The results derived from comparison with the corresponding control groups showed that PHA in humans induced a stronger proliferation and wider range of cytokine secretion, along with a greater number of differently expressed genes (DEGs), than when PHA was applied in cynomolgus monkeys. The significant upregulation of genes involved in the mitotic cell cycle, including cyclin B2, TOP2A, TYMS, and CEP55, was observed in human PBMCs with PHA stimulation, while only infrequent or slight upregulation occurred in cynomolgus monkey PBMCs, which may be one of the reasons for a stronger response to PHA in humans. In contrast to PHA, LPS in both species induced a similar proliferation ratio, cytokine profile, and DEG count, suggesting that human and cynomolgus monkeys have a similar response intensity for innate immune responses. Furthermore, 38 and 20 overlapped genes under PHA and LPS stimulation, respectively, were found in both species. These overlapped DEGs were associated with the same biological functions, including DNA replication, mitosis, immune response, chemotaxis, and inflammatory response. Thus, these results might reflect the highly conserved signatures of immune responses to PHA/LPS stimulation across the primates. Moreover, there were some differences in antigen processing and presentation, and the interferon gamma (INF-γ)–mediated signaling pathway in these species detected by gene expression profile study. Conclusions In conclusion, this is the first study to compare data on the responses of PBMCs to PHA and LPS in humans versus cynomolgus monkeys, and these findings may provide crucial insights into translating non-human primate (NHP) studies into human trials.
Collapse
Affiliation(s)
- Zhi Lin
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Ying Huang
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Hua Jiang
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Di Zhang
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Yanwei Yang
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Xingchao Geng
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| | - Bo Li
- National Institute for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing Key Lab for Preclinical Safety Evaluation of Drugs, Beijing, China
| |
Collapse
|
13
|
Sawaswong V, Fahsbender E, Altan E, Kemthong T, Deng X, Malaivijitnond S, Payungporn S, Delwart E. High Diversity and Novel Enteric Viruses in Fecal Viromes of Healthy Wild and Captive Thai Cynomolgus Macaques ( Macaca fascicularis). Viruses 2019; 11:E971. [PMID: 31652508 PMCID: PMC6832579 DOI: 10.3390/v11100971] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Cynomolgus macaques are common across South East Asian countries including Thailand. The National Primate Research Center of Thailand, Chulalongkorn University (NPRCT-CU) captures wild-borne cynomolgus macaque for research use. Limited information is available on the enteric viruses and possible zoonotic infections into or from cynomolgus macaques. We characterized and compare the fecal virome of two populations; healthy wild-originated captive cynomolgus macaques (n = 43) reared in NPRCT-CU and healthy wild cynomolgus macaques (n = 35). Over 90% of recognized viral sequence reads amplified from feces were from bacterial viruses. Viruses from seven families of mammalian viruses were also detected (Parvoviridae, Anelloviridae, Picornaviridae, Adenoviridae, Papillomaviridae, Herpesviridae, and Caliciviridae). The genomes of a member of a new picornavirus genus we named Mafapivirus, a primate chapparvovirus, and a circular Rep-encoding single-strand (CRESS) DNA virus were also characterized. Higher abundance of CRESS DNA viruses of unknown tropism and invertebrate-tropic ambidensovirus were detected in wild versus captive macaques likely reflecting dietary differences. Short term rearing in captivity did not have a pronounced effect on the diversity of mammalian viruses of wild cynomolgus macaques. This study is the first report of the fecal virome of cynomolgus macaques, non-human primates frequently used in biomedical research and vaccination studies.
Collapse
Affiliation(s)
- Vorthon Sawaswong
- Vitalant Research Institute, San Francisco, CA 94118, USA.
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Elizabeth Fahsbender
- Vitalant Research Institute, San Francisco, CA 94118, USA.
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 9413, USA.
| | - Eda Altan
- Vitalant Research Institute, San Francisco, CA 94118, USA.
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 9413, USA.
| | - Taratorn Kemthong
- National Primate Research Center-Chulalongkorn University, Saraburi 18110, Thailand.
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA 94118, USA.
| | | | - Sunchai Payungporn
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
- Center of Excellence in Systems Biology, Chulalongkorn University (CUSB), Bangkok 10330, Thailand.
| | - Eric Delwart
- Vitalant Research Institute, San Francisco, CA 94118, USA.
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 9413, USA.
| |
Collapse
|
14
|
Sii-Felice K, Castillo Padilla J, Relouzat F, Cheuzeville J, Tantawet S, Maouche L, Le Grand R, Leboulch P, Payen E. Enhanced Transduction of Macaca fascicularis Hematopoietic Cells with Chimeric Lentiviral Vectors. Hum Gene Ther 2019; 30:1306-1323. [DOI: 10.1089/hum.2018.179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Karine Sii-Felice
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Javier Castillo Padilla
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Francis Relouzat
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Joëlle Cheuzeville
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- bluebird bio France, Fontenay aux Roses, France
| | - Siriporn Tantawet
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Leïla Maouche
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- INSERM, Paris, France
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, UMR 1184, IDMIT Department, Institute of Biology François Jacob, INSERM, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Philippe Leboulch
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- Ramathibodi Hospital and Mahidol University, Bangkok, Thailand
- Harvard Medical School and Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston Massachusetts
| | - Emmanuel Payen
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- INSERM, Paris, France
| |
Collapse
|
15
|
Yu W, Hao X, Yang F, Ma J, Zhao Y, Li Y, Wang J, Xu H, Chen L, Liu Q, Duan S, Yang Y, Huang F, He Z. Hematological and biochemical parameters for Chinese rhesus macaque. PLoS One 2019; 14:e0222338. [PMID: 31527891 PMCID: PMC6748566 DOI: 10.1371/journal.pone.0222338] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022] Open
Abstract
Rhesus macaque is an important animal model in biomedical research, especially human disease, developmental, translational, and pre-clinical research. Blood physiological and biochemical parameters are important markers for physiology, pathology, and toxicology research. However, these parameters have not been systematically reported for Chinese rhesus macaques. To characterize the reference for these parameters, this study collected 1805 Chinese rhesus macaques living in Southwestern China. A total of 24 blood physiological indexes and 27 biochemical parameters were determined. Sex and age were found to affect these parameters. In conclusion, a comprehensive and systematic reference of hematological and biochemical parameters for Chinese rhesus macaque was established in this work on the basis of a large cohort. Such reference will benefit biomedical research employing rhesus macaques as animal models.
Collapse
Affiliation(s)
- Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Xianhui Hao
- Medical Faculty, Kunming University of Science and Technology, Kunming, PR China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Jin Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Hongjie Xu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Lixiong Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Quan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Suqin Duan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Yaping Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
| | - Fen Huang
- Medical Faculty, Kunming University of Science and Technology, Kunming, PR China
- * E-mail: (FH); (ZH)
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, PR China
- * E-mail: (FH); (ZH)
| |
Collapse
|
16
|
Shiina T, Blancher A. The Cynomolgus Macaque MHC Polymorphism in Experimental Medicine. Cells 2019; 8:E978. [PMID: 31455025 PMCID: PMC6770713 DOI: 10.3390/cells8090978] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Among the non-human primates used in experimental medicine, cynomolgus macaques (Macaca fascicularis hereafter referred to as Mafa) are increasingly selected for the ease with which they are maintained and bred in captivity. Macaques belong to Old World monkeys and are phylogenetically much closer to humans than rodents, which are still the most frequently used animal model. Our understanding of the Mafa genome has progressed rapidly in recent years and has greatly benefited from the latest technical advances in molecular genetics. Cynomolgus macaques are widespread in Southeast Asia and numerous studies have shown a distinct genetic differentiation of continental and island populations. The major histocompatibility complex of cynomolgus macaque (Mafa MHC) is organized in the same way as that of human, but it differs from the latter by its high degree of classical class I gene duplication. Human polymorphic MHC regions play a pivotal role in allograft transplantation and have been associated with more than 100 diseases and/or phenotypes. The Mafa MHC polymorphism similarly plays a crucial role in experimental allografts of organs and stem cells. Experimental results show that the Mafa MHC class I and II regions influence the ability to mount an immune response against infectious pathogens and vaccines. MHC also affects cynomolgus macaque reproduction and impacts on numerous biological parameters. This review describes the Mafa MHC polymorphism and the methods currently used to characterize it. We discuss some of the major areas of experimental medicine where an effect induced by MHC polymorphism has been demonstrated.
Collapse
Affiliation(s)
- Takashi Shiina
- Department of Molecular Life Sciences, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Antoine Blancher
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier (UPS), Toulouse 31000, France.
- Laboratoire d'immunologie, CHU de Toulouse, Institut Fédératif de Biologie, hôpital Purpan, 330 Avenue de Grande Bretagne, TSA40031, 31059 Toulouse CEDEX 9, France.
| |
Collapse
|
17
|
Nugeyre MT, Tchitchek N, Adapen C, Cannou C, Contreras V, Benjelloun F, Ravel J, Le Grand R, Marlin R, Menu E. Dynamics of Vaginal and Rectal Microbiota Over Several Menstrual Cycles in Female Cynomolgus Macaques. Front Cell Infect Microbiol 2019; 9:188. [PMID: 31249812 PMCID: PMC6582644 DOI: 10.3389/fcimb.2019.00188] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
The composition of the microbiota in cynomolgus macaques is only partially characterized, although this animal model is often used to study pathogenesis and preventive strategies against infections. We thus performed, for the first time, a longitudinal characterization of the vaginal and rectal microbiota of five cycling female cynomolgus macaques. Samples were collected weekly for 15 weeks and the V3/V4 regions of the16S rRNA gene sequenced. Sequences were analyzed with QIIME for OTU detection and taxonomic assignment. Progesterone levels were also determined to evaluate hormonal influence on bacteria relative abundance. The rectal and vaginal bacterial composition in cynomolgus macaques is polymicrobial and clearly distinct, with larger individual variability in the vagina. Rectal microbiota profiles were consistent between animals, whereas they were highly variable and animal-specific in the vagina. In the rectum, the most abundant taxa were Ruminococcaceae, Prevotella, and Clostridiales. In the vagina, the most abundant genera were Sneathia, Porphyromonas, Prevotella, and Fusobacterium. Lactobacillus were found at relative abundances higher than 1% in only one animal and were not predominant. Comparison of the vaginal cynomolgus macaque microbiota with that of humans showed similarity to community state type IV-A usually associated with dysbiosis. In the vagina, the relative abundance of 12 bacterial genera was found to be associated with progesterone levels. Our study provides a detailed characterization of the rectal and vaginal microbiota in female cynomolgus macaques and opens new perspectives of this animal model.
Collapse
Affiliation(s)
- Marie-Thérèse Nugeyre
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Nicolas Tchitchek
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Cindy Adapen
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Claude Cannou
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Vanessa Contreras
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Fahd Benjelloun
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Jacques Ravel
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Roger Le Grand
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Romain Marlin
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Elisabeth Menu
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
18
|
Cynomolgus macaque IL37 polymorphism and control of SIV infection. Sci Rep 2019; 9:7981. [PMID: 31138840 PMCID: PMC6538695 DOI: 10.1038/s41598-019-44235-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/27/2019] [Indexed: 01/17/2023] Open
Abstract
The association between gene polymorphisms and plasma virus load at the set point (SP-PVL) was investigated in Mauritian macaques inoculated with SIV. Among 44 macaques inoculated with 50 AID50, six individuals were selected: three with SP-PVL among the highest and three with SP-PVL among the lowest. The exons of 390 candidate genes of these six animals were sequenced. Twelve non-synonymous single nucleotide polymorphisms (NS-SNPs) lying in nine genes potentially associated with PVL were genotyped in 23 animals. Three NS-SNPs with probabilities of association with PVL less than 0.05 were genotyped in a total of 44 animals. One NS-SNP lying in exon 1 of the IL37 gene displayed a significant association (p = 3.33 × 10−4) and a strong odds ratio (19.52). Multiple linear regression modeling revealed three significant predictors of SP-PVL, including the IL37 exon 1 NS-SNP (p = 0.0004) and the MHC Class IB haplotypes M2 (p = 0.0007) and M6 (p = 0.0013). These three factors in conjunction explained 48% of the PVL variance (p = 4.8 × 10−6). The potential role of IL37 in the control of SIV infection is discussed.
Collapse
|
19
|
Current advances in HIV vaccine preclinical studies using Macaque models. Vaccine 2019; 37:3388-3399. [PMID: 31088747 DOI: 10.1016/j.vaccine.2019.04.094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/02/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
The macaque simian or simian/human immunodeficiency virus (SIV/SHIV) challenge model has been widely used to inform and guide human vaccine trials. Substantial advances have been made recently in the application of repeated-low-dose challenge (RLD) approach to assess SIV/SHIV vaccine efficacies (VE). Some candidate HIV vaccines have shown protective effects in preclinical studies using the macaque SIV/SHIV model but the model's true predictive value for screening potential HIV vaccine candidates needs to be evaluated further. Here, we review key parameters used in the RLD approach and discuss their relevance for evaluating VE to improve preclinical studies of candidate HIV vaccines.
Collapse
|
20
|
Multivariate profiling of African green monkey and rhesus macaque T lymphocytes. Sci Rep 2019; 9:4834. [PMID: 30886198 PMCID: PMC6423277 DOI: 10.1038/s41598-019-41209-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/27/2019] [Indexed: 12/22/2022] Open
Abstract
The complexity of immune responses limits the usefulness of univariate methods in answering complex immunology questions. To demonstrate the utility of a multivariate approach, we employ such approach to compare T cells of African green monkeys (AGMs) and rhesus macaques (RMs). Among the most prominent distinguishing features we found were lower CD3 and higher CD28 surface expression in AGMs compared to RMs. After in vitro stimulation, a larger proportion of AGM T cells secreted cytokines, especially those producing more than one cytokine (i.e. multifunctional cells). To find out whether multifunctional responses associate with protection in other species, we compared T cells of cynomolgus macaques (CMs) infected with wild-type Simian Immunodeficiency Virus (SIV) to those of CMs infected (vaccinated) with a replication-defective virus. Wild-type SIV infection in macaques leads to simian Acquired Immunodeficiency Syndrome (AIDS), which does not happen in animals previously vaccinated with a replication-defective virus. Interestingly, after in vitro stimulation, multifunctional cells were more abundant among T cells of vaccinated CMs. Our results propose T-cell multifunctionality as a potentially useful marker of immunity, although additional verification is needed. Finally, we hope our multivariate model and its associated validation methods will inform future studies in the field of immunology.
Collapse
|
21
|
Kurscheidt FA, Mesquita CSS, Damke GMZF, Damke E, Carvalho ARBDA, Suehiro TT, Teixeira JJV, da Silva VRS, Souza RP, Consolaro MEL. Persistence and clinical relevance of Zika virus in the male genital tract. Nat Rev Urol 2019; 16:211-230. [DOI: 10.1038/s41585-019-0149-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
22
|
Li H, Hai Y, Lim SY, Toledo N, Crecente-Campo J, Schalk D, Li L, Omange RW, Dacoba TG, Liu LR, Kashem MA, Wan Y, Liang B, Li Q, Rakasz E, Schultz-Darken N, Alonso MJ, Plummer FA, Whitney JB, Luo M. Mucosal antibody responses to vaccines targeting SIV protease cleavage sites or full-length Gag and Env proteins in Mauritian cynomolgus macaques. PLoS One 2018; 13:e0202997. [PMID: 30153293 PMCID: PMC6112674 DOI: 10.1371/journal.pone.0202997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023] Open
Abstract
HIV mutates rapidly and infects CD4+ T cells, especially when they are activated. A vaccine targeting conserved, essential viral elements while limiting CD4+ T cell activation could be effective. Learning from natural immunity observed in a group of highly HIV-1 exposed seronegative Kenyan female sex workers, we are testing a novel candidate HIV vaccine targeting the 12 viral protease cleavage sites (PCSs) (the PCS vaccine), in comparison with a vaccine targeting full-length Gag and Env (the Gag/Env vaccine) in a Mauritian cynomolgus macaque/SIV model. In this study we evaluated these vaccines for induction of mucosal antibodies to SIV immunogens at the female genital tract. Bio-Plex and Western blot analyses of cervicovaginal lavage samples showed that both the PCS and Gag/Env vaccines can elicit mucosal IgG antibody responses to SIV immunogens. Significantly higher increase of anti-PCS antibodies was induced by the PCS vaccine than by the Gag/Env vaccine (p<0.0001). The effect of the mucosal antibody responses in protection from repeated low dose pathogenic SIVmac251 challenges is being evaluated.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Yan Hai
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - So-Yon Lim
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Nikki Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Dane Schalk
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, WI, United States of America
| | - Lin Li
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Robert W Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Lewis R Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Yanmin Wan
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Eva Rakasz
- Immunology Services Unit, Wisconsin National Primate Research Center, Madison, WI, United States of America
| | - Nancy Schultz-Darken
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, WI, United States of America
| | - Maria J Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States of America
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| |
Collapse
|
23
|
de Manuel M, Shiina T, Suzuki S, Dereuddre-Bosquet N, Garchon HJ, Tanaka M, Congy-Jolivet N, Aarnink A, Le Grand R, Marques-Bonet T, Blancher A. Whole genome sequencing in the search for genes associated with the control of SIV infection in the Mauritian macaque model. Sci Rep 2018; 8:7131. [PMID: 29739964 PMCID: PMC5940699 DOI: 10.1038/s41598-018-25071-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/11/2018] [Indexed: 11/09/2022] Open
Abstract
In the Mauritian macaque experimentally inoculated with SIV, gene polymorphisms potentially associated with the plasma virus load at a set point, approximately 100 days post inoculation, were investigated. Among the 42 animals inoculated with 50 AID50 of the same strain of SIV, none of which received any preventive or curative treatment, nine individuals were selected: three with a plasma virus load (PVL) among the lowest, three with intermediate PVL values and three among the highest PVL values. The complete genomes of these nine animals were then analyzed. Initially, attention was focused on variants with a potential functional impact on protein encoding genes (non-synonymous SNPs (NS-SNPs) and splicing variants). Thus, 424 NS-SNPs possibly associated with PVL were detected. The 424 candidates SNPs were genotyped in these 42 SIV experimentally infected animals (including the nine animals subjected to whole genome sequencing). The genes containing variants most probably associated with PVL at a set time point are analyzed herein.
Collapse
Affiliation(s)
- Marc de Manuel
- Institute of Evolutionary Biology, UPF-CSIC, PRBB, Dr. Aiguader 88, 08003, Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, ICREA, Passeig de Lluís Companys, 23, 08010, Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation, CRG, Barcelona Institute of Science and Technology (BIST, Baldiri i Reixac 4, 08028, Barcelona, Spain
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Shingo Suzuki
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Nathalie Dereuddre-Bosquet
- CEA - Université Paris-Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, 92265, Fontenay-aux-Roses, France
| | - Henri-Jean Garchon
- Inserm U1173, Simone Veil School of Health Sciences, University of Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
- Genetics Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Masayuki Tanaka
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Nicolas Congy-Jolivet
- Laboratoire d'immunogénétique moléculaire (LIMT, EA 3034, Faculté de médecine Purpan, Université Toulouse 3 (Université Paul Sabatier, UPS), Toulouse, France
- Laboratoire d'immunologie, CHU de Toulouse, France
| | - Alice Aarnink
- Laboratoire d'immunogénétique moléculaire (LIMT, EA 3034, Faculté de médecine Purpan, Université Toulouse 3 (Université Paul Sabatier, UPS), Toulouse, France
| | - Roger Le Grand
- CEA - Université Paris-Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, 92265, Fontenay-aux-Roses, France
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology, UPF-CSIC, PRBB, Dr. Aiguader 88, 08003, Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, ICREA, Passeig de Lluís Companys, 23, 08010, Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation, CRG, Barcelona Institute of Science and Technology (BIST, Baldiri i Reixac 4, 08028, Barcelona, Spain
| | - Antoine Blancher
- Laboratoire d'immunogénétique moléculaire (LIMT, EA 3034, Faculté de médecine Purpan, Université Toulouse 3 (Université Paul Sabatier, UPS), Toulouse, France.
- Laboratoire d'immunologie, CHU de Toulouse, France.
| |
Collapse
|
24
|
Li H, Omange RW, Czarnecki C, Correia-Pinto JF, Crecente-Campo J, Richmond M, Li L, Schultz-Darken N, Alonso MJ, Whitney JB, Plummer FA, Luo M. Mauritian cynomolgus macaques with M3M4 MHC genotype control SIVmac251 infection. J Med Primatol 2018; 46:137-143. [PMID: 28748659 DOI: 10.1111/jmp.12300] [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] [Accepted: 06/28/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Understanding natural HIV control may lead to new preventative or therapeutic strategies. Several protective major histocompatibility complex (MHC) genotypes were found in humans and rhesus macaques. Here, we report a simian immunodeficiency virus (SIV) controller MHC genotype in Mauritian cynomolgus macaques (MCMs). METHODS Twelve MHC-genotyped MCMs were infected with SIVmac251 and monitored for viral loads and CD4+ T-cell counts. RESULTS Two macaques with M3M4 genotype exhibited the lowest peak viral loads (log plasma SIV RNA copies/mL), nearly 3 logs lower than those in most macaques with other MHC haplotype combinations, and set point viral loads below the level of detection limit by RT-qPCR (<2 log RNA copies/mL). They maintained healthy CD4+ T-cell counts of >500 cells/μL blood, while CD4 counts in the vast majority of other macaques were below this level. CONCLUSIONS The M3M4 MHC genotype may confer enhanced control of SIV replication in MCMs.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Robert W Omange
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Chris Czarnecki
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Jorge F Correia-Pinto
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Crecente-Campo
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Meika Richmond
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Lin Li
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | | | - Maria J Alonso
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Francis A Plummer
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Ma Luo
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| |
Collapse
|
25
|
Li H, Li L, Liu LR, Omange RW, Toledo N, Kashem MA, Hai Y, Liang B, Plummer FA, Luo M. Hypothetical endogenous SIV-like antigens in Mauritian cynomolgus macaques. Bioinformation 2018; 14:48-52. [PMID: 29618899 PMCID: PMC5879946 DOI: 10.6026/97320630014048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 01/24/2023] Open
Abstract
Simian immunodeficiency virus (SIV) infection of Mauritian cynomolgus macaques (MCMs) is an increasingly important nonhuman primate model for HIV vaccine research. We previously reported that in MCMs anti-SIV antibodies can be naturally developed without exogenous infection or vaccination, and that a vaccine targeting SIV protease cleavage sites (PCS) can cross-induce antibodies to non-PCS SIV antigens. We speculate that this is potentially caused by the existence of endogenous SIV-like antigens. External stimuli (such as environmental factors and vaccination) may induce expression of endogenous SIV-like antigens to elicit these antibodies. Database and mass spectrometry analyses were conducted to search for such antigens. We identified endogenous SIV-like DNA sequences in cynomolgus macaque genome and non-PCS peptide homologous to SIV Env protein in PBMCs of a PCS-vaccinated monkey. Our preliminary insights suggest that endogenous SIV-like antigens may be one of the possible reasons for the natural and cross-inducible SIV antibodies in MCMs.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Lin Li
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Lewis R Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Robert W Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Nikki Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Yan Hai
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 3N4, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| |
Collapse
|
26
|
Li H, Nykoluk M, Li L, Liu LR, Omange RW, Soule G, Schroeder LT, Toledo N, Kashem MA, Correia-Pinto JF, Liang B, Schultz-Darken N, Alonso MJ, Whitney JB, Plummer FA, Luo M. Natural and cross-inducible anti-SIV antibodies in Mauritian cynomolgus macaques. PLoS One 2017; 12:e0186079. [PMID: 28982126 PMCID: PMC5628977 DOI: 10.1371/journal.pone.0186079] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/25/2017] [Indexed: 01/16/2023] Open
Abstract
Cynomolgus macaques are an increasingly important nonhuman primate model for HIV vaccine research. SIV-free animals without pre-existing anti-SIV immune responses are generally needed to evaluate the effect of vaccine-induced immune responses against the vaccine epitopes. Here, in order to select such animals for vaccine studies, we screened 108 naïve female Mauritian cynomolgus macaques for natural (baseline) antibodies to SIV antigens using a Bio-Plex multiplex system. The antigens included twelve 20mer peptides overlapping the twelve SIV protease cleavage sites (-10/+10), respectively (PCS peptides), and three non-PCS Gag or Env peptides. Natural antibodies to SIV antigens were detected in subsets of monkeys. The antibody reactivity to SIV was further confirmed by Western blot using purified recombinant SIV Gag and Env proteins. As expected, the immunization of monkeys with PCS antigens elicited anti-PCS antibodies. However, unexpectedly, antibodies to non-PCS peptides were also induced, as shown by both Bio-Plex and Western blot analyses, while the non-PCS peptides do not share sequence homology with PCS peptides. The presence of natural and vaccine cross-inducible SIV antibodies in Mauritian cynomolgus macaques should be considered in animal selection, experimental design and result interpretation, for their best use in HIV vaccine research.
Collapse
Affiliation(s)
- Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mikaela Nykoluk
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Lin Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lewis R. Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert W. Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Geoff Soule
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Lukas T. Schroeder
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nikki Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jorge F. Correia-Pinto
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, La Coruña, Spain
| | - Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Maria J. Alonso
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, La Coruña, Spain
| | - James B. Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Francis A. Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- * E-mail: ,
| |
Collapse
|
27
|
Prall TM, Graham ME, Karl JA, Wiseman RW, Ericsen AJ, Raveendran M, Alan Harris R, Muzny DM, Gibbs RA, Rogers J, O'Connor DH. Improved full-length killer cell immunoglobulin-like receptor transcript discovery in Mauritian cynomolgus macaques. Immunogenetics 2017; 69:325-339. [PMID: 28343239 PMCID: PMC5856007 DOI: 10.1007/s00251-017-0977-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/08/2017] [Indexed: 12/25/2022]
Abstract
Killer cell immunoglobulin-like receptors (KIRs) modulate disease progression of pathogens including HIV, malaria, and hepatitis C. Cynomolgus and rhesus macaques are widely used as nonhuman primate models to study human pathogens, and so, considerable effort has been put into characterizing their KIR genetics. However, previous studies have relied on cDNA cloning and Sanger sequencing that lack the throughput of current sequencing platforms. In this study, we present a high throughput, full-length allele discovery method utilizing Pacific Biosciences circular consensus sequencing (CCS). We also describe a new approach to Macaque Exome Sequencing (MES) and the development of the Rhexome1.0, an adapted target capture reagent that includes macaque-specific capture probe sets. By using sequence reads generated by whole genome sequencing (WGS) and MES to inform primer design, we were able to increase the sensitivity of KIR allele discovery. We demonstrate this increased sensitivity by defining nine novel alleles within a cohort of Mauritian cynomolgus macaques (MCM), a geographically isolated population with restricted KIR genetics that was thought to be completely characterized. Finally, we describe an approach to genotyping KIRs directly from sequence reads generated using WGS/MES reads. The findings presented here expand our understanding of KIR genetics in MCM by associating new genes with all eight KIR haplotypes and demonstrating the existence of at least one KIR3DS gene associated with every haplotype.
Collapse
Affiliation(s)
- Trent M Prall
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA
| | - Michael E Graham
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Julie A Karl
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Roger W Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Adam J Ericsen
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA
| | | | - R Alan Harris
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeffrey Rogers
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA.
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, 53711, USA.
| |
Collapse
|
28
|
Tavano B, Tsipouri V, Hardy GAD, Royle CM, Keegan MR, Fuchs D, Patterson S, Almond N, Berry N, Ham C, Ferguson D, Boasso A. Immune Responses in the Central Nervous System Are Anatomically Segregated in a Non-Human Primate Model of Human Immunodeficiency Virus Infection. Front Immunol 2017; 8:361. [PMID: 28424694 PMCID: PMC5371826 DOI: 10.3389/fimmu.2017.00361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/14/2017] [Indexed: 12/05/2022] Open
Abstract
The human immunodeficiency virus (HIV) accesses the central nervous system (CNS) early during infection, leading to HIV-associated cognitive impairment and establishment of a viral reservoir. Here, we describe a dichotomy in inflammatory responses in different CNS regions in simian immunodeficiency virus (SIV)-infected macaques, a model for HIV infection. We found increased expression of inflammatory genes and perivascular leukocyte infiltration in the midbrain of SIV-infected macaques. Conversely, the frontal lobe showed downregulation of inflammatory genes associated with interferon-γ and interleukin-6 pathways, and absence of perivascular cuffing. These immunologic alterations were not accompanied by differences in SIV transcriptional activity within the tissue. Altered expression of genes associated with neurotoxicity was observed in both midbrain and frontal lobe. The segregation of inflammatory responses to specific regions of the CNS may both account for HIV-associated neurological symptoms and constitute a critical hurdle for HIV eradication by shielding the CNS viral reservoir from antiviral immunity.
Collapse
Affiliation(s)
- Barbara Tavano
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Vicky Tsipouri
- NIHR Biological Research Unit, Royal Brompton Hospital, London, UK
| | - Gareth A D Hardy
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Caroline M Royle
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Michael R Keegan
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK.,ViiV Healthcare, Middlesex, UK
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Steven Patterson
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Neil Almond
- Division of Virology, National Institute for Biological Standards and Controls (NIBSC), Potters Bar, Hertfordshire, UK
| | - Neil Berry
- Division of Virology, National Institute for Biological Standards and Controls (NIBSC), Potters Bar, Hertfordshire, UK
| | - Claire Ham
- Division of Virology, National Institute for Biological Standards and Controls (NIBSC), Potters Bar, Hertfordshire, UK
| | - Deborah Ferguson
- Division of Virology, National Institute for Biological Standards and Controls (NIBSC), Potters Bar, Hertfordshire, UK
| | - Adriano Boasso
- Centre for Immunology and Vaccinology (CIV), Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
29
|
Koide F, Goebel S, Snyder B, Walters KB, Gast A, Hagelin K, Kalkeri R, Rayner J. Development of a Zika Virus Infection Model in Cynomolgus Macaques. Front Microbiol 2016; 7:2028. [PMID: 28066354 PMCID: PMC5165249 DOI: 10.3389/fmicb.2016.02028] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/02/2016] [Indexed: 12/18/2022] Open
Abstract
Limited availability of Indian rhesus macaques (IRM) is a bottleneck to study Zika virus (ZIKV) pathogenesis and evaluation of appropriate control measures in non-human primates. To address these issues, we report here the Mauritian cynomolgus macaque (MCM) model for ZIKV infection. In brief, six MCMs (seronegative for Dengue and ZIKV) were subdivided into three cohorts with a male and female each and challenged with different doses of Asian [PRVABC59 (Puerto Rico) or FSS13025 (Cambodia)] or African (IBH30656) lineage ZIKV isolates. Clinical signs were monitored; and biological fluids (serum, saliva, and urine) and tissues (testes and brain) were assessed for viral load by quantitative reverse transcription polymerase chain reaction and neutralizing antibodies (Nab) by 50% Plaque Reduction Neutralization Test (PRNT50) at various times post-infection (p.i). PRVABC59 induced viremia detectable up to day 10, with peak viral load at 2–3 days p.i. An intermittent viremia spike was observed on day 30 with titers reaching 2.5 × 103 genomes/mL. Moderate viral load was observed in testes, urine and saliva. In contrast, FSS13025 induced viremia lasting only up to 6 days and detectable viral loads in testes but not in urine and saliva. Recurrent viremia was detected but at lower titers compare to PRVABC59. Challenge with either PRVABC59 or FSS13025 resulted in 100% seroconversion; with mean PRNT50 titers ranging from 597 to 5179. IBH30656 failed to establish infection in MCM suggesting that MCM are susceptible to infection with ZIKV isolates of the Asian lineage but not from Africa. Due to the similarity of biphasic viremia and Nab responses between MCM and IRM models, MCM could be a suitable alternative for evaluation of ZIKV vaccine and therapeutic candidates.
Collapse
Affiliation(s)
- Fusataka Koide
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Scott Goebel
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Beth Snyder
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Kevin B Walters
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Alison Gast
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Kimberly Hagelin
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Raj Kalkeri
- Department of Infectious Disease Research, Drug Development, Southern Research Institute, Frederick MD, USA
| | - Jonathan Rayner
- Department of Infectious Disease Research, Drug Development, Southern Research, Birmingham AL, USA
| |
Collapse
|
30
|
Okamura T, Tsujimura Y, Soma S, Takahashi I, Matsuo K, Yasutomi Y. Simian immunodeficiency virus SIVmac239 infection and simian human immunodeficiency virus SHIV89.6P infection result in progression to AIDS in cynomolgus macaques of Asian origin. J Gen Virol 2016; 97:3413-3426. [PMID: 27902330 DOI: 10.1099/jgv.0.000641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Simian immunodeficiency virus (SIV) infection models in cynomolgus macaques are important for analysis of the pathogenesis of immunodeficiency virus and for studies on the efficacy of new vaccine candidates. However, very little is known about the pathogenesis of SIV or simian human immunodeficiency virus (SHIV) in cynomolgus macaques from different Asian countries. In the present study, we analysed the infectivity and pathogenicity of CCR5-tropic SIVmac and those of dual-tropic SHIV89.6P inoculated into cynomolgus macaques in Indonesian, Malaysian or Philippine origin. The plasma viral loads in macaques infected with either SIVmac239 or SHIV89.6P were maintained at high levels. CD4+ T cell levels in macaques infected with SIVmac239 gradually decreased. All of the macaques infected with SHIV89.6P showed greatly reduced CD4+ T-cell numbers within 6 weeks of infection. Eight of the 11 macaques infected with SIVmac239 were killed due to AIDS symptoms after 2-4.5 years, while four of the five macaques infected with SHIV89.6P were killed due to AIDS symptoms after 1-3.5 years. We also analysed cynomolgus macaques infected intrarectally with repeated low, medium or high doses of SIVmac239, SIVmac251 or SHIV89.6P. Infection was confirmed by quantitative RT-PCR at more than 5000, 300 and 500 TCID50 for SIVmac239, SIVmac251 and SHIV89.6P, respectively. The present study indicates that cynomolgus macaques of Asian origin are highly susceptible to SIVmac and SHIV infection by both intravenous and mucosal routes. These models will be useful for studies on virus pathogenesis, vaccination and therapeutics against human immunodeficiency virus/AIDS.
Collapse
Affiliation(s)
- Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Yusuke Tsujimura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Shogo Soma
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan.,Division of Immunoregulation, Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Ichiro Takahashi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Kazuhiro Matsuo
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo 204-0022, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan.,Division of Immunoregulation, Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
31
|
Russell JNH, Marsh AK, Willer DO, Ambagala APN, Dzamba M, Chan JK, Pilon R, Fournier J, Brudno M, Antony JM, Sandstrom P, Evans BJ, MacDonald KS. A novel strain of cynomolgus macaque cytomegalovirus: implications for host-virus co-evolution. BMC Genomics 2016; 17:277. [PMID: 27044312 PMCID: PMC4820910 DOI: 10.1186/s12864-016-2588-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/14/2016] [Indexed: 12/20/2022] Open
Abstract
Background Cytomegaloviruses belong to a large, ancient, genus of DNA viruses comprised of a wide array of species-specific strains that occur in diverse array of hosts. Methods In this study we sequenced the ~217 Kb genome of a cytomegalovirus isolated from a Mauritius cynomolgus macaque, CyCMV Mauritius, and compared it to previously sequenced cytomegaloviruses from a cynomolgus macaque of Filipino origin (CyCMV Ottawa) and two from Indian rhesus macaques (RhCMV 180.92 and RhCMV 68–1). Results Though more closely related to CyCMV Ottawa, CyCMV Mauritius is less genetically distant from both RhCMV strains than is CyCMV Ottawa. Several individual genes, including homologues of CMV genes RL11B, UL123, UL83b, UL84 and a homologue of mammalian COX-2, show a closer relationship between homologues of CyCMV Mauritius and the RhCMVs than between homologues of CyCMV Mauritius and CyCMV Ottawa. A broader phylogenetic analysis of 12 CMV strains from eight species recovers evolutionary relationships among viral strains that mirror those amongst the host species, further demonstrating co-evolution of host and virus. Conclusions Phylogenetic analyses of rhesus and cynomolgus macaque CMV genome sequences demonstrate co-speciation of the virus and host. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2588-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Angie K Marsh
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, ON, Canada
| | - David O Willer
- Department of Medicine, University of Toronto, Toronto, M5S 1A8, ON, Canada.,Department of Microbiology, Mount Sinai Hospital, Toronto, M5G 1X5, ON, Canada
| | - Aruna P N Ambagala
- Department of Medicine, University of Toronto, Toronto, M5S 1A8, ON, Canada.,Department of Microbiology, Mount Sinai Hospital, Toronto, M5G 1X5, ON, Canada.,Present Address: Canadian Science Centre for Human and Animal Health, National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, R3E 3M4, MB, Canada
| | - Misko Dzamba
- Department of Computer Science, University of Toronto, Toronto, M5S 1A8, ON, Canada
| | - Jacqueline K Chan
- Department of Medicine, University of Toronto, Toronto, M5S 1A8, ON, Canada.,Department of Microbiology, Mount Sinai Hospital, Toronto, M5G 1X5, ON, Canada
| | - Richard Pilon
- National HIV & Retrovirology Laboratories, Public Health Agency of Canada, Ottawa, K1A 0K9, ON, Canada
| | - Jocelyn Fournier
- Scientific Services Division, Health Products & Food Branch, Health Canada, Ottawa, K1A 0L2, ON, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto, M5S 1A8, ON, Canada
| | - Joseph M Antony
- Department of Medicine, University of Toronto, Toronto, M5S 1A8, ON, Canada.,Department of Microbiology, Mount Sinai Hospital, Toronto, M5G 1X5, ON, Canada
| | - Paul Sandstrom
- National HIV & Retrovirology Laboratories, Public Health Agency of Canada, Ottawa, K1A 0K9, ON, Canada
| | - Ben J Evans
- Biology Department, McMaster University, Life Sciences Building, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Kelly S MacDonald
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, ON, Canada. .,Department of Medicine, University of Toronto, Toronto, M5S 1A8, ON, Canada. .,Department of Microbiology, Mount Sinai Hospital, Toronto, M5G 1X5, ON, Canada. .,Section of Infectious Diseases, Department of Internal Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, R3E 0J9, MB, Canada. .,Present Address: University of Manitoba, Basic Medical Sciences Building, Room 501, 745 Bannatyne Ave., Winnipeg, R3E 0J9, MB, Canada.
| |
Collapse
|
32
|
Itoh Y. Translational research on influenza virus infection using a nonhuman primate model. Pathol Int 2016; 66:132-141. [PMID: 26811109 DOI: 10.1111/pin.12385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/28/2015] [Indexed: 12/17/2022]
Abstract
Influenza virus infection is a seasonal infectious disease for humans, whereas it is also a zoonosis that is originally transmitted from animals to humans. Therefore, several animal models are used in research on influenza virus infection. We have used a nonhuman primate (NHP) model to extrapolate pathogenicity of various influenza viruses and efficacy of vaccines and antiviral drugs against the influenza viruses in humans. NHPs have genes, anatomical structure, and immune responses similar to those of humans as compared to other animal models. Using an NHP model, we revealed that the pandemic 2009 influenza A virus caused viral pneumonia as reported in human patients. Thus, it is thought that NHP models can be used to predict replication of emerging viruses in humans. We also examined the pathogenicity of highly pathogenic avian influenza viruses and evaluated a new therapeutic antibody in macaques under an immunocompromised condition. NHP models have provided promising results in research on other infectious diseases including Ebola virus and human/simian immunodeficiency virus infections. Thus, NHPs are important in biomedical research for determining the pathogenesis and for development of treatments, especially when clinical trials are difficult. We summarize the characteristics and advantages of research using NHP models in this review.
Collapse
Affiliation(s)
- Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|