1
|
Hindle S, Brien MÈ, Pelletier F, Giguère F, Trudel MJ, Dal Soglio D, Kakkar F, Soudeyns H, Girard S, Boucoiran I. Placenta analysis of Hofbauer cell profile according to the class of antiretroviral therapy used during pregnancy in people living with HIV. Placenta 2023; 139:120-126. [PMID: 37364521 DOI: 10.1016/j.placenta.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION The use of antiretroviral therapy drastically reduces vertical transmission of Human Immunodeficiency Virus. However, recent studies demonstrate associations between ART use during pregnancy and placental inflammation, particularly within protease inhibitor (PI)-based regimens. We sought to characterize placental macrophages, namely Hofbauer cells, according to the class of ART used during pregnancy. METHODS Using immunofluorescence and immunohistochemistry, placentas from 79 pregnant people living with HIV (PPLWH) and 29 HIV-uninfected people were analyzed to quantify the numbers and frequencies of leukocytes (CD45+) and Hofbauer cells (CD68+ and/or CD163+). PPLWH were stratified into three groups based on class of ART: non-nucleoside reverse transcriptase inhibitor (NNRTI)-based, integrase strand-transfer inhibitor (INSTI)-based, and PI-based regimens. RESULTS Placentas of PPLWH contained significantly more leukocytes and Hofbauer cells than controls. Multivariable analyses revealed that this increase in immune cells was associated with a predominantly CD163+ profile in all ART subgroups compared to the HIV-negative group. This was characterized by an increase in total CD163+ cells in the PI and INSTI subgroups, and a higher frequency of CD163+ cells and CD163+/CD68+ ratio in the NNRTI and PI subgroups. DISCUSSION Placentas of PPLWH treated with any ART regimen during their entire pregnancy displayed a selection for CD163+ cells compared to the HIV-negative group, regardless of class of ART, suggesting that class of ART does not intrinsically affect selection of CD163+ and CD68+ Hofbauer cells. Further investigations into the role of Hofbauer cells in ART-associated placental inflammation are warranted to identify the mechanisms behind their potential involvement in maternal-fetal tolerance maintenance.
Collapse
Affiliation(s)
- Stephanie Hindle
- Department of Pharmacology and Physiology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada; CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Marie-Ève Brien
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Florence Pelletier
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Frédérique Giguère
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Mei Juan Trudel
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Dorothée Dal Soglio
- Department of Pathology, CHU Sainte-Justine, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada.
| | - Fatima Kakkar
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada; Department of Pediatrics, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| | - Hugo Soudeyns
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada; Department of Microbiology, Infectiology and Immunology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| | - Sylvie Girard
- Department of Pharmacology and Physiology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada; CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada; Department of Obstetrics and Gynecology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada; Department of Microbiology, Infectiology and Immunology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada; Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Isabelle Boucoiran
- CHU Sainte-Justine Research Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1S2, Canada; Department of Obstetrics and Gynecology, Université de Montréal, 2900 boul. Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
2
|
Chandrasekar V, Singh AV, Maharjan RS, Dakua SP, Balakrishnan S, Dash S, Laux P, Luch A, Singh S, Pradhan M. Perspectives on the Technological Aspects and Biomedical Applications of Virus‐Like Particles/Nanoparticles in Reproductive Biology: Insights on the Medicinal and Toxicological Outlook. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
| | - Ajay Vikram Singh
- German Federal Institute for Risk Assessment (BfR) Department of Chemical and Product Safety Max-Dohrn-Straße 8-10 10589 Berlin Germany
| | - Romi Singh Maharjan
- German Federal Institute for Risk Assessment (BfR) Department of Chemical and Product Safety Max-Dohrn-Straße 8-10 10589 Berlin Germany
| | | | | | - Sagnika Dash
- Obstetrics and Gynecology Apollo Clinic Qatar 23656 Doha Qatar
| | - Peter Laux
- German Federal Institute for Risk Assessment (BfR) Department of Chemical and Product Safety Max-Dohrn-Straße 8-10 10589 Berlin Germany
| | - Andreas Luch
- German Federal Institute for Risk Assessment (BfR) Department of Chemical and Product Safety Max-Dohrn-Straße 8-10 10589 Berlin Germany
| | - Suyash Singh
- Department of Neurosurgery All India Institute of Medical Sciences Raebareli UP 226001 India
| | - Mandakini Pradhan
- Department of Fetal Medicine Sanjay Gandhi Post Graduate Institute of Medical Sciences Reabareli Road Lucknow UP 226014 India
| |
Collapse
|
3
|
Girsch JH, Mejia Plazas MC, Olivier A, Farah M, Littlefield D, Behl S, Punia S, Sakemura R, Hemsath JR, Norgan A, Enninga EAL, Johnson EL, Chakraborty R. Host-Viral Interactions at the Maternal-Fetal Interface. What We Know and What We Need to Know. FRONTIERS-A JOURNAL OF WOMEN STUDIES 2022; 2:833106. [PMID: 36742289 PMCID: PMC9894500 DOI: 10.3389/fviro.2022.833106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In humans, the hemochorial placenta is a unique temporary organ that forms during pregnancy to support fetal development, gaseous exchange, delivery of nutrition, removal of waste products, and provides immune protection, while maintaining tolerance to the HLA-haploidentical fetus. In this review, we characterize decidual and placental immunity during maternal viral (co)-infection with HIV-1, human cytomegalovirus (HCMV), and Zika virus. We discuss placental immunology, clinical presentation, and epidemiology, before characterizing host susceptibility and cellular tropism, and how the three viruses gain access into specific placental target cells. We describe current knowledge on host-viral interactions with decidual and stromal human placental macrophages or Hofbauer cells, trophoblasts including extra villous trophoblasts, T cells, and decidual natural killer (dNK) cells. These clinically significant viral infections elicit both innate and adaptive immune responses to control replication. However, the three viruses either during mono- or co-infection (HIV-1 and HCMV) escape detection to initiate placental inflammation associated with viral transmission to the developing fetus. Aside from congenital or perinatal infection, other adverse pregnancy outcomes include preterm labor and spontaneous abortion. In addition, maternal HIV-1 and HCMV co-infection are associated with impaired fetal and infant immunity in postnatal life and poor clinical outcomes during childhood in exposed infants, even in the absence of vertical transmission of HIV-1. Given the rapidly expanding numbers of HIV-1-exposed uninfected infants and children globally, further research is urgently needed on neonatal immune programming during maternal mono-and co-infection. This review therefore includes sections on current knowledge gaps that may prompt future research directions. These gaps reflect an emerging but poorly characterized field. Their significance and potential investigation is underscored by the fact that although viral infections result in adverse consequences in both mother and developing fetus/newborn, antiviral and immunomodulatory therapies can improve clinical outcomes in the dyad.
Collapse
Affiliation(s)
- James H. Girsch
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Mayo Clinic Graduate School of Biomedical Science, Rochester, MN, United States
| | - Maria C. Mejia Plazas
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Amanda Olivier
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Mohamed Farah
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Dawn Littlefield
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Supriya Behl
- Department of Pediatric Research, Mayo Clinic, Rochester, MN, United States
| | - Sohan Punia
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Reona Sakemura
- Department of Hematology Research, Mayo Clinic, Rochester, MN, United States
| | - Jack R. Hemsath
- Department of Infectious Diseases Research, Mayo Clinic, Rochester, MN, United States
| | - Andrew Norgan
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Elizabeth A. L. Enninga
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Erica L. Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States,Correspondence: Rana Chakraborty
| |
Collapse
|
4
|
Fakonti G, Pantazi P, Bokun V, Holder B. Placental Macrophage (Hofbauer Cell) Responses to Infection During Pregnancy: A Systematic Scoping Review. Front Immunol 2022; 12:756035. [PMID: 35250964 PMCID: PMC8895398 DOI: 10.3389/fimmu.2021.756035] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Congenital infection of the fetus via trans-placental passage of pathogens can result in severe morbidity and mortality. Even without transmission to the fetus, infection of the placenta itself is associated with pregnancy complications including pregnancy loss and preterm birth. Placental macrophages, also termed Hofbauer cells (HBCs), are fetal-origin macrophages residing in the placenta that are likely involved in responding to placental infection and protection of the developing fetus. As HBCs are the only immune cell present in the villous placenta, they represent one of the final opportunities for control of infection and prevention of passage to the developing fetus. OBJECTIVE AND RATIONALE The objective of this review was to provide a systematic overview of the literature regarding HBC responses during infection in pregnancy, including responses to viral, bacterial, and parasitic pathogens. METHODS PubMed and Scopus were searched on May 20th, 2021, with no limit on publication date, to identify all papers that have studied placental macrophages/Hofbauer cells in the context of infection. The following search strategy was utilized: (hofbauer* OR "hofbauer cells" OR "hofbauer cell" OR "placental macrophage" OR "placental macrophages") AND [infect* OR virus OR viral OR bacteri* OR parasite* OR pathogen* OR LPS OR "poly(i:c)" OR toxoplasm* OR microb* OR HIV)]. OUTCOMES 86 studies were identified for review. This included those that investigated HBCs in placentas from pregnancies complicated by maternal infection and in vitro studies investigating HBC responses to pathogens or Pathogen-Associated Molecular Patterns (PAMPs). HBCs can be infected by a variety of pathogens, and HBC hyperplasia was a common observation. HBCs respond to pathogen infection and PAMPs by altering their transcriptional, translational and secretion profiles. Co-culture investigations demonstrate that they can replicate and transmit pathogens to other cells. In other cases, they may eliminate the pathogen through a variety of mechanisms including phagocytosis, cytokine-mediated pathogen elimination, release of macrophage extracellular traps and HBC-antibody-mediated neutralization. HBC responses differ across gestation and may be influenced by pre-existing immunity. Clinical information, including gestational age at infection, gestational age of the samples, mode of sample collection and pregnancy outcome were missing for the majority of studies.
Collapse
Affiliation(s)
| | | | | | - Beth Holder
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Yu W, Hu X, Cao B. Viral Infections During Pregnancy: The Big Challenge Threatening Maternal and Fetal Health. MATERNAL-FETAL MEDICINE 2022; 4:72-86. [PMID: 35187500 PMCID: PMC8843053 DOI: 10.1097/fm9.0000000000000133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/11/2021] [Indexed: 12/18/2022] Open
Abstract
Viral infections during pregnancy are associated with adverse pregnancy outcomes, including maternal and fetal mortality, pregnancy loss, premature labor, and congenital anomalies. Mammalian gestation encounters an immunological paradox wherein the placenta balances the tolerance of an allogeneic fetus with protection against pathogens. Viruses cannot easily transmit from mother to fetus due to physical and immunological barriers at the maternal-fetal interface posing a restricted threat to the fetus and newborns. Despite this, the unknown strategies utilized by certain viruses could weaken the placental barrier to trigger severe maternal and fetal health issues especially through vertical transmission, which was not fully understood until now. In this review, we summarize diverse aspects of the major viral infections relevant to pregnancy, including the characteristics of pathogenesis, related maternal-fetal complications, and the underlying molecular and cellular mechanisms of vertical transmission. We highlight the fundamental signatures of complex placental defense mechanisms, which will prepare us to fight the next emerging and re-emerging infectious disease in the pregnancy population.
Collapse
Affiliation(s)
- Wenzhe Yu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaoqian Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
6
|
Senft AD, Macfarlan TS. Transposable elements shape the evolution of mammalian development. Nat Rev Genet 2021; 22:691-711. [PMID: 34354263 DOI: 10.1038/s41576-021-00385-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Transposable elements (TEs) promote genetic innovation but also threaten genome stability. Despite multiple layers of host defence, TEs actively shape mammalian-specific developmental processes, particularly during pre-implantation and extra-embryonic development and at the maternal-fetal interface. Here, we review how TEs influence mammalian genomes both directly by providing the raw material for genetic change and indirectly via co-evolving TE-binding Krüppel-associated box zinc finger proteins (KRAB-ZFPs). Throughout mammalian evolution, individual activities of ancient TEs were co-opted to enable invasive placentation that characterizes live-born mammals. By contrast, the widespread activity of evolutionarily young TEs may reflect an ongoing co-evolution that continues to impact mammalian development.
Collapse
Affiliation(s)
- Anna D Senft
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, MD, USA.
| | - Todd S Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Amin O, Powers J, Bricker KM, Chahroudi A. Understanding Viral and Immune Interplay During Vertical Transmission of HIV: Implications for Cure. Front Immunol 2021; 12:757400. [PMID: 34745130 PMCID: PMC8566974 DOI: 10.3389/fimmu.2021.757400] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Despite the significant progress that has been made to eliminate vertical HIV infection, more than 150,000 children were infected with HIV in 2019, emphasizing the continued need for sustainable HIV treatment strategies and ideally a cure for children. Mother-to-child-transmission (MTCT) remains the most important route of pediatric HIV acquisition and, in absence of prevention measures, transmission rates range from 15% to 45% via three distinct routes: in utero, intrapartum, and in the postnatal period through breastfeeding. The exact mechanisms and biological basis of these different routes of transmission are not yet fully understood. Some infants escape infection despite significant virus exposure, while others do not, suggesting possible maternal or fetal immune protective factors including the presence of HIV-specific antibodies. Here we summarize the unique aspects of HIV MTCT including the immunopathogenesis of the different routes of transmission, and how transmission in the antenatal or postnatal periods may affect early life immune responses and HIV persistence. A more refined understanding of the complex interaction between viral, maternal, and fetal/infant factors may enhance the pursuit of strategies to achieve an HIV cure for pediatric populations.
Collapse
Affiliation(s)
- Omayma Amin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Jenna Powers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine M. Bricker
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| |
Collapse
|
8
|
Johnson EL, Swieboda D, Olivier A, Enninga EAL, Chakraborty R. Robust innate immune responses at the placenta during early gestation may limit in utero HIV transmission. PLoS Pathog 2021; 17:e1009860. [PMID: 34432853 PMCID: PMC8437274 DOI: 10.1371/journal.ppat.1009860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/13/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022] Open
Abstract
In 2019, >90% of new HIV infections in infants globally occurred vertically. Studies suggest intrauterine transmission most often occurs in the third trimester; however, there are no mechanistic studies to support these observations. We therefore obtained early/mid-gestation and term placentae from 20 HIV/Hepatitis B/CMV negative women. Isolated primary placental macrophages (Hofbauer cells [HCs]) were exposed to HIV-1BaL and/or interferon (IFN)-α, IFN-β, IFN-λ1, and RIG-I-like receptor (RLR) agonists. qRT-PCR, FACS, ELISA, Luminex, and Western blot analyses determined expression of activation markers, co-receptors, viral antigen, cytokines, antiviral genes, and host proteins. Early gestation HCs express higher levels of CCR5 and exhibit a more activated phenotype. Despite downregulation of CCR5, term HCs were more susceptible to HIV replication. Early gestation HCs displayed a more activated phenotype than term HCs and HIV exposure lead to the further up-regulation of T-cell co-stimulatory and MHC molecules. Limited HIV replication in early/mid gestation HCs was associated with increased secretion of anti-inflammatory cytokines, chemokines, and a more robust antiviral immune response. In contrast, term HCs were more susceptible to HIV replication, associated with dampening of IFN-induced STAT1 and STAT2 protein activation. Treatment of early/mid gestation and term HCs, with type I IFNs or RLR agonists reduced HIV replication, underscoring the importance of IFN and RLR signaling in inducing an antiviral state. Viral recognition and antiviral immunity in early gestation HCs may prevent in utero HIV infection, whereas diminished antiviral responses at term can facilitate transmission. Defining mechanisms and specific timing of vertical transmission are critical for the development of specific vaccines and antiviral therapeutics to prevent new HIV infections in children globally.
Collapse
Affiliation(s)
- Erica L. Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Dominika Swieboda
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amanda Olivier
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Elizabeth Ann L. Enninga
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| |
Collapse
|
9
|
Cerveny L, Murthi P, Staud F. HIV in pregnancy: Mother-to-child transmission, pharmacotherapy, and toxicity. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166206. [PMID: 34197912 DOI: 10.1016/j.bbadis.2021.166206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/18/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022]
Abstract
An estimated 1.3 million pregnant women were living with HIV in 2018. HIV infection is associated with adverse pregnancy outcomes and all HIV-positive pregnant women, regardless of their clinical stage, should receive a combination of antiretroviral drugs to suppress maternal viral load and prevent vertical fetal infection. Although antiretroviral treatment in pregnant women has undoubtedly minimized mother-to-child transmission of HIV, several uncertainties remain. For example, while pregnancy is accompanied by changes in pharmacokinetic parameters, relevant data from clinical studies are lacking. Similarly, long-term adverse effects of exposure to antiretrovirals on fetuses have not been studied in detail. Here, we review current knowledge on HIV effects on the placenta and developing fetus, recommended antiretroviral regimens, and pharmacokinetic considerations with particular focus on placental transport. We also discuss recent advances in antiretroviral research and potential effects of antiretroviral treatment on placental/fetal development and programming.
Collapse
Affiliation(s)
- Lukas Cerveny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Padma Murthi
- Department of Medicine, School of Clinical Sciences, and Department of Pharmacology, Monash Biomedicine Discovery Institute Monash University, Clayton, Victoria, Australia; Hudson Institute of Medical Research, The Ritchie Centre, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
10
|
Tang Y, Woodward BO, Pastor L, George AM, Petrechko O, Nouvet FJ, Haas DW, Jiang G, Hildreth JEK. Endogenous Retroviral Envelope Syncytin Induces HIV-1 Spreading and Establishes HIV Reservoirs in Placenta. Cell Rep 2021; 30:4528-4539.e4. [PMID: 32234485 DOI: 10.1016/j.celrep.2020.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/16/2020] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Radical cure of HIV-1 (HIV) is hampered by the establishment of HIV reservoirs and persistent infection in deep tissues despite suppressive antiretroviral therapy (ART). Here, we show that among HIV-positive women receiving suppressive ART, cells from placental tissues including trophoblasts contain HIV RNA and DNA. These viruses can be reactivated by latency reversal agents. We find that syncytin, the envelope glycoprotein of human endogenous retrovirus family W1 expressed on placental trophoblasts, triggers cell fusion with HIV-infected T cells. This results in cell-to-cell spread of HIV to placental trophoblasts. Such cell-to-cell spread of HIV is less sensitive to ART than free virus. Replication in syncytin-expressing cells can also produce syncytin-pseudotyped HIV, further expanding its ability to infect non-CD4 cells. These previously unrecognized mechanisms of HIV entry enable the virus to bypass receptor restriction to infect host barrier cells, thereby facilitating viral transmission and persistent infection in deep tissues.
Collapse
Affiliation(s)
- Yuyang Tang
- University of North Carolina at Chapel Hill, HIV Cure Center and Institute of Global Health & Infectious Diseases, Chapel Hill, NC 27599, USA; Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Beverly O Woodward
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Lorena Pastor
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Alvin M George
- Department of Internal Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Oksana Petrechko
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Franklin J Nouvet
- Department of Internal Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - David W Haas
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, TN 37240, USA; Department of Internal Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Guochun Jiang
- University of North Carolina at Chapel Hill, HIV Cure Center and Institute of Global Health & Infectious Diseases, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - James E K Hildreth
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Department of Internal Medicine, Meharry Medical College, Nashville, TN 37208, USA.
| |
Collapse
|
11
|
Narang K, Cheek EH, Enninga EAL, Theiler RN. Placental Immune Responses to Viruses: Molecular and Histo-Pathologic Perspectives. Int J Mol Sci 2021; 22:2921. [PMID: 33805739 PMCID: PMC7998619 DOI: 10.3390/ijms22062921] [Citation(s) in RCA: 7] [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: 02/17/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
As most recently demonstrated by the SARS-CoV-2 pandemic, congenital and perinatal infections are of significant concern to the pregnant population as compared to the general population. These outcomes can range from no apparent impact all the way to spontaneous abortion or fetal infection with long term developmental consequences. While some pathogens have developed mechanisms to cross the placenta and directly infect the fetus, other pathogens lead to an upregulation in maternal or placental inflammation that can indirectly cause harm. The placenta is a temporary, yet critical organ that serves multiple important functions during gestation including facilitation of fetal nutrition, oxygenation, and prevention of fetal infection in utero. Here, we review trophoblast cell immunology and the molecular mechanisms utilized to protect the fetus from infection. Lastly, we discuss consequences in the placenta when these protections fail and the histopathologic result following infection.
Collapse
Affiliation(s)
- Kavita Narang
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA;
| | - Elizabeth H. Cheek
- Department of Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA;
| | - Elizabeth Ann L. Enninga
- Departments of Immunology, Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA;
| | - Regan N. Theiler
- Division of Obstetrics, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
12
|
Mezouar S, Katsogiannou M, Ben Amara A, Bretelle F, Mege JL. Placental macrophages: Origin, heterogeneity, function and role in pregnancy-associated infections. Placenta 2020; 103:94-103. [PMID: 33120051 PMCID: PMC7568513 DOI: 10.1016/j.placenta.2020.10.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/22/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022]
Abstract
Placental macrophages are a heterogenous population of immune cells present throughout pregnancy. They are essential for maintenance of the homeostatic placenta environment and host defense against infections. The characterization of placental macrophages as well as their activation have been limited for a long time by the lack of convenient tools. The emergence of unbiased methods makes it possible to reappraise the study of placental macrophages. In this review, we discuss the diversity and the functions of placental macrophages to better understand their dysfunctions during placental infections.
Collapse
Affiliation(s)
- Soraya Mezouar
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France.
| | - Maria Katsogiannou
- Hôpital Saint Joseph, Department of Obstetrics and Gynecology, FR-13008, Marseille, France
| | - Amira Ben Amara
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France
| | - Florence Bretelle
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France; AP-HM, Gynecology Department, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France; AP-HM, UF Immunology, Marseille, France.
| |
Collapse
|
13
|
Hoo R, Nakimuli A, Vento-Tormo R. Innate Immune Mechanisms to Protect Against Infection at the Human Decidual-Placental Interface. Front Immunol 2020; 11:2070. [PMID: 33013876 PMCID: PMC7511589 DOI: 10.3389/fimmu.2020.02070] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/29/2020] [Indexed: 12/25/2022] Open
Abstract
During pregnancy, the placenta forms the anatomical barrier between the mother and developing fetus. Infectious agents can potentially breach the placental barrier resulting in pathogenic transmission from mother to fetus. Innate immune responses, orchestrated by maternal and fetal cells at the decidual-placental interface, are the first line of defense to avoid vertical transmission. Here, we outline the anatomy of the human placenta and uterine lining, the decidua, and discuss the potential capacity of pathogen pattern recognition and other host defense strategies present in the innate immune cells at the placental-decidual interface. We consider major congenital infections that access the placenta from hematogenous or decidual route. Finally, we highlight the challenges in studying human placental responses to pathogens and vertical transmission using current experimental models and identify gaps in knowledge that need to be addressed. We further propose novel experimental strategies to address such limitations.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Annettee Nakimuli
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Obstetrics and Gynecology, School of Medicine, Makerere University, Kampala, Uganda
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
14
|
Gabriel B, Medin C, Alves J, Nduati R, Bosire RK, Wamalwa D, Farquhar C, John-Stewart G, Lohman-Payne BL. Analysis of the TCR Repertoire in HIV-Exposed but Uninfected Infants. Sci Rep 2019; 9:11954. [PMID: 31420576 PMCID: PMC6697688 DOI: 10.1038/s41598-019-48434-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 08/02/2019] [Indexed: 01/10/2023] Open
Abstract
Maternal human immunodeficiency virus (HIV) infection has been shown to leave profound and lasting impacts on the HIV-exposed uninfected (HEU) infant, including increased mortality and morbidity, immunological changes, and developmental delays compared to their HIV-unexposed (HU) counterparts. Exposure to HIV or antiretroviral therapy may influence immune development, which could increase morbidity and mortality. However, a direct link between the increased mortality and morbidity and the infant's immune system has not been identified. To provide a global picture of the neonatal T cell repertoire in HEU versus HU infants, the diversity of the T cell receptor beta chain (TRB) expressed in cord blood samples from HEU infants was determined using next-generation sequencing and compared to healthy (HU) infants collected from the same community. While the TRB repertoire of HU infants was broadly diverse, in line with the expected idea of a naïve T cell repertoire, samples of HEU infants showed a significantly reduced TRB diversity. This study is the first to demonstrate differences in TRB diversity between HEU and HU cord blood samples and provides evidence that maternal HIV, in the absence of transmission, influences the adaptive immune system of the unborn child.
Collapse
Affiliation(s)
- Benjamin Gabriel
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, 02903, USA
| | - Carey Medin
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, 02903, USA
| | - Jeremiah Alves
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, 02903, USA
| | - Ruth Nduati
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, 30197, Kenya
| | - Rose Kerubo Bosire
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Dalton Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, 30197, Kenya
| | - Carey Farquhar
- Department of Global Health, University of Washington, Seattle, Washington, 98104, USA
- Departments of Medicine, University of Washington, Seattle, Washington, 98104, USA
- Departments of Epidemiology, University of Washington, Seattle, Washington, 98104, USA
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, 98104, USA
- Departments of Medicine, University of Washington, Seattle, Washington, 98104, USA
- Departments of Epidemiology, University of Washington, Seattle, Washington, 98104, USA
- Departments of Pediatrics, University of Washington, Seattle, Washington, 98104, USA
| | - Barbara L Lohman-Payne
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, 02903, USA.
| |
Collapse
|
15
|
Zulu MZ, Martinez FO, Gordon S, Gray CM. The Elusive Role of Placental Macrophages: The Hofbauer Cell. J Innate Immun 2019; 11:447-456. [PMID: 30970346 DOI: 10.1159/000497416] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/19/2019] [Indexed: 01/07/2023] Open
Abstract
In this review, we discuss the often overlooked tissue-resident fetal macrophages, Hofbauer cells, which are found within the chorionic villi of the human placenta. Hofbauer cells have been shown to have a phenotype associated with regulatory and anti-inflammatory functions. They are thought to play a crucial role in the regulation of pregnancy and in the maintenance of a homeostatic environment that is crucial for fetal development. Even though the numbers of these macrophages are some of the most abundant immune cells in the human placenta, which are sustained throughout pregnancy, there are very few studies that have identified their origin, their phenotype, and functions and why they are maintained throughout gestation. It is not yet understood how Hofbauer cells may change in function throughout normal pregnancy, and especially in those complicated by maternal gestational diabetes, preeclampsia, and viral infections, such as Zika, cytomegalovirus, and human immunodeficiency virus. We review what is known about the origin of these macrophages and explore how common complications of pregnancy dysregulate these cells leading to adverse birth outcomes in humans. Our synthesis sheds light on areas for human studies that can further define these innate regulatory cells.
Collapse
Affiliation(s)
- Michael Z Zulu
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fernando O Martinez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Siamon Gordon
- Chang Gung University, Graduate Institute of Biomedical Sciences, College of Medicine, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Clive M Gray
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa, .,National Health Laboratory Services/Groote Schuur Hospital, Cape Town, South Africa,
| |
Collapse
|
16
|
Milligan C, Slyker JA, Overbaugh J. The Role of Immune Responses in HIV Mother-to-Child Transmission. Adv Virus Res 2017; 100:19-40. [PMID: 29551137 DOI: 10.1016/bs.aivir.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
HIV mother-to-child transmission (MTCT) represents a success story in the HIV/AIDS field given the significant reduction in number of transmission events with the scale-up of antiretroviral treatment and other prevention methods. Nevertheless, MTCT still occurs and better understanding of the basic biology and immunology of transmission will aid in future prevention and treatment efforts. MTCT is a unique setting given that the transmission pair is known and the infant receives passively transferred HIV-specific antibodies from the mother while in utero. Thus, infant exposure to HIV occurs in the face of HIV-specific antibodies, especially during delivery and breastfeeding. This review highlights the immune correlates of protection in HIV MTCT including humoral (neutralizing antibodies, antibody-dependent cellular cytotoxicity, and binding epitopes), cellular, and innate immune factors. We further discuss the future implications of this research as it pertains to opportunities for passive and active vaccination with the ultimate goal of eliminating HIV MTCT.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States.
| | | | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Mother-to-child transmission (MTCT) of HIV-1 remains a significant global health concern despite implementation of maternal combination antiretroviral therapy for treatment as prevention to offset transmission. The risk of in-utero HIV-1 transmission in the absence of interventions is ∼7%. This low rate of transmission points to innate and adaptive mechanisms to restrict lentiviral infection within the placenta. RECENT FINDINGS Placental macrophages (Hofbauer cells) are key mediators in in-utero transmission of HIV-1. Hofbauer cells constitutively express elevated concentrations of regulatory cytokines, which inhibit HIV-1 replication in vitro, and possess intrinsic antiviral properties. Hofbauer cells sequester HIV-1 in intracellular compartments that can be accessed by HIV-1-specific antibodies and may occur in vivo to offset MTCT. Intriguingly, studies have reported strong associations between maternal human cytomegalovirus (HCMV) viremia and MTCT of HIV-1. HCMV infection at the placenta promotes inflammation, chronic villitis, and trophoblast damage, providing potential HIV-1 access into CD4CCR5 target cells. The placenta exhibits a variety of mechanisms to limit HIV-1 replication, yet viral-induced activation with maternal HCMV may override this protection to facilitate in-utero transmission of HIV-1. SUMMARY Understanding immune correlates of protection or transmission at the placenta during on-going HIV-1 exposure may contribute to understanding HIV pathogenesis and the development of effective immunotherapies.
Collapse
|
18
|
Eastlund T. Infectious Disease Transmission through Cell, Tissue, and Organ Transplantation: Reducing the Risk through Donor Selection. Cell Transplant 2017; 4:455-77. [PMID: 8520830 DOI: 10.1177/096368979500400507] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The incidence of cell transplant-transmitted infection is unknown and can only be inferred from prospective studies–that have not yet been performed and reported. The possibility of donor-to-recipient disease transmission through cell transplant therapy can be considered by reviewing the risk associated with other transplanted tissues and organs. Viral, bacterial, and fungal infections have been transmitted via transplantation of organs, tissue allografts such as bone, skin, cornea, and heart valves, and cells such as islets, hematopoietic stem cells, and semen. Several types of protozoan and worm parasites have been transferred via organ transplants. Bone allografts have transmitted hepatitis, tuberculosis, and human immunodeficiency virus (HIV-1). Corneas have transmitted rabies, Creutzfeldt-Jakob disease (CJD), hepatitis B (HBV), cytomegalovirus (CMV), herpes simplex virus (HSV), bacteria, and fungi. Heart valves have been implicated in transmitting tuberculosis and hepatitis B. HIV-1 and CMV seroconversion has been reported in patients receiving skin from seropositive donors. CJD has been transmitted by dura and pericardium transplants. Over the past several years, improvements in donor screening criteria, such as excluding potential donors with infection and those with behaviors risky for HIV-1 and hepatitis infection, and introduction of new donor blood tests have greatly reduced the risk of HIV-1 and hepatitis and may have nearly eliminated the risk of tuberculosis and CJD. Prior to use, many tissues are exposed to antibiotics, disinfectants, and sterilants, which further reduce or remove the risk of transmitted disease. Because organs, cells, and some tissue grafts cannot be subjected to sterilization steps, the risk of infectious disease transmission remains and thorough donor screening and testing is especially important.
Collapse
Affiliation(s)
- T Eastlund
- American Red Cross, North Central Tissue Services, St. Paul, MN 55107, USA
| |
Collapse
|
19
|
Dorsamy V, Vallen C, Haffejee F, Moodley J, Naicker T. The role of trophoblast cell receptor expression in HIV-1 passage across the placenta in pre-eclampsia: an observational study. BJOG 2016; 124:920-928. [PMID: 27700010 DOI: 10.1111/1471-0528.14311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To compare expression of markers of HIV and associated receptors (p24, CD4, CCR5 and ICAM-2) in placentae and umbilical cords of HIV-associated and pre-eclamptic pregnancies to elucidate any association between these conditions in mother-to-child transmission. DESIGN Cross-sectional immunohistochemical analysis of target receptor expression. SETTING Laboratory-based study of primigravidae attending a district hospital in South Africa. POPULATION OR SAMPLE Retrospectively collected placental tissue (stratified into four groups according to HIV status of normotensive and pre-eclamptic participants (n = 20/group). METHOD Immunohistochemistry utilising CD4 (1:1), p24 (1:10), CCR5 (1:80) and ICAM-2 (1:100) antibodies was performed using light microscopy for image acquisition and analysis. MAIN OUTCOME MEASURES Evaluate the expression of receptors on syncytiotrophoblast involved in in utero transmission of HIV. RESULTS Syncytiotrophoblast was immunopositive for CD4 and CCR5 antibody with greater expression of CCR5 in HIV-positive versus HIV-negative groups (F1,159 = 6.979, P = 0.009) and normotensive versus pre-eclamptic groups (F1,159 = 8.803, P = 0.003). p24 was present in both placentae and umbilical cords of babies that were HIV-negative at 6 weeks. ICAM-2 immunostaining was observed in the syncytiotrophoblast across study groups and was significantly higher in the HIV-negative pre-eclamptic group (χ2 (3) = 45.3; P < 0.001). CONCLUSION Concurrent CD4 and CCR5 receptor expression demonstrates possible in utero viral entry routes across the placental barrier. ICAM-2 expression may influence HIV passage across the placenta or restoration of risk of pre-eclampsia in HAART-treated mothers. HIV was found in fetal circulation regardless of antiretroviral treatment. Further confirmatory ultrastructural and molecular work is warranted. TWEETABLE ABSTRACT CD4, CCR5 and ICAM-2 on syncytiotrophoblast may facilitate HIV infection of passage across the placenta.
Collapse
Affiliation(s)
- V Dorsamy
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - C Vallen
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - F Haffejee
- Department of Basic Medical Sciences, Durban University of Technology, Durban, South Africa
| | - J Moodley
- Women's Health and HIV Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - T Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
20
|
Kudoh A, Miyakawa K, Matsunaga S, Matsushima Y, Kosugi I, Kimura H, Hayakawa S, Sawasaki T, Ryo A. H11/HSPB8 Restricts HIV-2 Vpx to Restore the Anti-Viral Activity of SAMHD1. Front Microbiol 2016; 7:883. [PMID: 27379031 PMCID: PMC4904303 DOI: 10.3389/fmicb.2016.00883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/25/2016] [Indexed: 11/13/2022] Open
Abstract
Virus-host interactions play vital roles in viral replication and virus-induced pathogenesis. Viruses rely entirely upon host cells to reproduce progeny viruses; however, host factors positively or negatively regulate virus replication by interacting with viral proteins. The elucidation of virus-host protein interaction not only provides a better understanding of the molecular mechanisms by which host cells combat viral infections, but also facilitates the development of new anti-viral therapeutics. Identification of relevant host factors requires techniques that enable comprehensive characterization of virus-host protein interactions. In this study, we developed a proteomic approach to systematically identify human protein kinases that interact potently with viral proteins. For this purpose, we synthesized 412 full-length human protein kinases using the wheat germ cell-free protein synthesis system, and screened them for their association with a virus protein using the amplified luminescent proximity homogenous assay (AlphaScreen). Using this system, we attempted to discover a robust anti-viral host restriction mechanism targeting virus protein X (Vpx) of HIV-2. The screen identified H11/HSPB8 as a Vpx-binding protein that negatively regulates the stability and function of Vpx. Indeed, overexpression of H11/HSPB8 promoted the degradation of Vpx via the ubiquitin-proteasome pathway and inhibited its interaction with SAMHD1, a host restriction factor responsible for blocking replication of HIV. Conversely, targeted knockdown of H11/HSPB8 in human trophoblast cells, which ordinarily express high levels of this protein, restored the expression and function of Vpx, making the cells highly susceptible to viral replication. These results demonstrate that our proteomic approach represents a powerful tool for revealing virus-host interaction not yet identified by conventional methods. Furthermore, we showed that H11/HSPB8 could be a potential host regulatory factor that may prevent placental infection of HIV-2 during pregnancy.
Collapse
Affiliation(s)
- Ayumi Kudoh
- Department of Microbiology, School of Medicine, Yokohama City University Yokohama, Japan
| | - Kei Miyakawa
- Department of Microbiology, School of Medicine, Yokohama City University Yokohama, Japan
| | - Satoko Matsunaga
- Department of Microbiology, School of Medicine, Yokohama City University Yokohama, Japan
| | - Yuki Matsushima
- Kawasaki City Health and Safety Research Center Kanagawa, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Hirokazu Kimura
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine Tokyo, Japan
| | | | - Akihide Ryo
- Department of Microbiology, School of Medicine, Yokohama City University Yokohama, Japan
| |
Collapse
|
21
|
Evans C, Humphrey JH, Ntozini R, Prendergast AJ. HIV-Exposed Uninfected Infants in Zimbabwe: Insights into Health Outcomes in the Pre-Antiretroviral Therapy Era. Front Immunol 2016; 7:190. [PMID: 27375613 PMCID: PMC4893498 DOI: 10.3389/fimmu.2016.00190] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/02/2016] [Indexed: 11/13/2022] Open
Abstract
The ZVITAMBO trial recruited 14,110 mother-infant pairs to a randomized controlled trial of vitamin A between 1997 and 2000, before the availability of antiretroviral therapy for HIV prophylaxis or treatment in Zimbabwe. The HIV status of mothers and infants was well characterized through 1-2 years of follow-up, leading to the largest cohort to date of HIV-exposed uninfected (HEU) infants (n = 3135), with a suitable comparison group of HIV-unexposed infants (n = 9510). Here, we draw on 10 years of published findings from the ZVITAMBO trial. HEU infants had increased morbidity compared to HIV-unexposed infants, with 50% more hospitalizations in the neonatal period and 30% more sick clinic visits during infancy, particularly for skin infections, lower respiratory tract infections, and oral thrush. HEU children had 3.9-fold and 2.0-fold higher mortality than HIV-unexposed children during the first and second years of life, respectively, most commonly due to acute respiratory infections, diarrhea/dysentery, malnutrition, sepsis, and meningitis. Infant morbidity and mortality were strongly related to maternal HIV disease severity, and increased morbidity remained until maternal CD4 counts were >800 cells/μL. HEU infants were more likely to be premature and small-for-gestational age than HIV-unexposed infants, and had more postnatal growth failure. Here, we propose a conceptual framework to explain the increased risk of infectious morbidity, mortality, and growth failure among HEU infants, hypothesizing that immune activation and inflammation are key drivers of both infection susceptibility and growth failure. Future studies should further dissect the causes of infection susceptibility and growth failure and determine the impact of ART and cotrimoxazole on outcomes of this vulnerable group of infants in the current era.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research , Harare , Zimbabwe
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
22
|
Quicke KM, Bowen JR, Johnson EL, McDonald CE, Ma H, O'Neal JT, Rajakumar A, Wrammert J, Rimawi BH, Pulendran B, Schinazi RF, Chakraborty R, Suthar MS. Zika Virus Infects Human Placental Macrophages. Cell Host Microbe 2016; 20:83-90. [PMID: 27247001 DOI: 10.1016/j.chom.2016.05.015] [Citation(s) in RCA: 360] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 12/28/2022]
Abstract
The recent Zika virus (ZIKV) outbreak in Brazil has been directly linked to increased cases of microcephaly in newborns. Current evidence indicates that ZIKV is transmitted vertically from mother to fetus. However, the mechanism of intrauterine transmission and the cell types involved remain unknown. We demonstrate that the contemporary ZIKV strain PRVABC59 (PR 2015) infects and replicates in primary human placental macrophages, called Hofbauer cells, and to a lesser extent in cytotrophoblasts, isolated from villous tissue of full-term placentae. Viral replication coincides with induction of type I interferon (IFN), pro-inflammatory cytokines, and antiviral gene expression, but with minimal cell death. Our results suggest a mechanism for intrauterine transmission in which ZIKV gains access to the fetal compartment by directly infecting placental cells and disrupting the placental barrier.
Collapse
Affiliation(s)
- Kendra M Quicke
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - James R Bowen
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Erica L Johnson
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Circe E McDonald
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Huailiang Ma
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Justin T O'Neal
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Augustine Rajakumar
- Department of Gynecology and Obstetrics, Division of Maternal Fetal Medicine and Reproductive Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Bassam H Rimawi
- Department of Gynecology and Obstetrics, Division of Maternal Fetal Medicine and Reproductive Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Bali Pulendran
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Raymond F Schinazi
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for AIDS Research, Laboratory of Biochemical Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rana Chakraborty
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mehul S Suthar
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| |
Collapse
|
23
|
Milligan C, Overbaugh J. The role of cell-associated virus in mother-to-child HIV transmission. J Infect Dis 2015; 210 Suppl 3:S631-40. [PMID: 25414417 DOI: 10.1093/infdis/jiu344] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mother-to-child transmission (MTCT) of human immunodeficiency virus (HIV) continues to contribute to the global burden of disease despite great advances in antiretroviral (ARV) treatment and prophylaxis. In this review, we discuss the proposed mechanisms of MTCT, evidence for cell-free and cell-associated transmission in different routes of MTCT, and the impact of ARVs on virus levels and transmission. Many population-based studies support a role for cell-associated virus in transmission and in vitro studies also provide some support for this mode of transmission. However, animal model studies provide proof-of-principle that cell-free virus can establish infection in infants, and studies of ARVs in HIV-infected pregnant women show a strong correlation with reduction in cell-free virus levels and protection. ARV treatment in MTCT potentially provides opportunities to better define the infectious form of virus, but these studies will require better tools to measure the infectious cell reservoir.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center Medical Scientist Training Program, University of Washington School of Medicine Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, Washington
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center Medical Scientist Training Program, University of Washington School of Medicine
| |
Collapse
|
24
|
Placental Hofbauer cells assemble and sequester HIV-1 in tetraspanin-positive compartments that are accessible to broadly neutralizing antibodies. J Int AIDS Soc 2015; 18:19385. [PMID: 25623930 PMCID: PMC4308659 DOI: 10.7448/ias.18.1.19385] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/11/2014] [Accepted: 12/16/2014] [Indexed: 12/23/2022] Open
Abstract
Introduction Within monocyte-derived macrophages, HIV-1 accumulates in intracellular virus-containing compartments (VCCs) that are inaccessible to the external environment, which implicate these cells as latently infected HIV-1 reservoirs. During mother-to-child transmission of HIV-1, human placental macrophages (Hofbauer cells (HCs)) are viral targets, and have been shown to be infected in vivo and sustain low levels of viral replication in vitro; however, the risk of in utero transmission is less than 7%. The role of these primary macrophages as viral reservoirs is largely undefined. The objective of this study is to define potential sites of viral assembly, accumulation and neutralization in HCs given the pivotal role of the placenta in preventing HIV-1 infection in the mother-infant dyad. Methods Term placentae from 20 HIV-1 seronegative women were obtained following caesarian section. VCCs were evaluated by 3D confocal and electron microscopy. Colocalization R values (Pearson's correlation) were quantified with colocalization module of Volocity 5.2.1. Replication kinetics and neutralization studies were evaluated using p24 ELISA. Results We demonstrate that primary HCs assemble and sequester HIV-1BaL in intracellular VCCs, which are enriched in endosomal/lysosomal markers, including CD9, CD81, CD63 and LAMP-1. Following infection, we observed HIV-1 accumulation in potentially acidic compartments, which stained intensely with Lysotracker-Red. Remarkably, these compartments are readily accessible via the cell surface and can be targeted by exogenously applied small molecules and HIV-1-specific broadly neutralizing antibodies. In addition, broadly neutralizing antibodies (4E10 and VRC01) limited viral replication by HIV-1-infected HCs, which may be mediated by FcγRI. Conclusions These findings suggest that placental HCs possess intrinsic adaptations facilitating unique sequestration of HIV-1, and may serve as a protective viral reservoir to permit viral neutralization and/or antiretroviral drug entry in utero.
Collapse
|
25
|
Abstract
The mammalian placenta exhibits elevated expression of endogenous retroviruses (ERVs), but the evolutionary significance of this feature remains unclear. I propose that ERV-mediated regulatory evolution was, and continues to be, an important mechanism underlying the evolution of placental development. Many recent studies have focused on the co-option of ERV-derived genes for specific functional adaptations in the placenta. However, the co-option of ERV-derived regulatory elements could potentially lead to the incorporation of entire gene regulatory networks, which, I argue, would facilitate relatively rapid developmental evolution of the placenta. I suggest a model in which an ancient retroviral infection led to the establishment of the ancestral placental developmental gene network through the co-option of ERV-derived regulatory elements. Consequently, placental development would require elevated tolerance to ERV activity. This in turn would expose a continuous stream of novel ERV mutations that may have catalyzed the developmental diversification of the mammalian placenta.
Collapse
Affiliation(s)
- Edward B Chuong
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
26
|
Singh P, Agnihotri SK, Tewari MC, Kumar S, Sachdev M, Tripathi RK. HIV-1 Nef breaches placental barrier in rat model. PLoS One 2012; 7:e51518. [PMID: 23240037 PMCID: PMC3519864 DOI: 10.1371/journal.pone.0051518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 11/01/2012] [Indexed: 11/18/2022] Open
Abstract
The vertical transmission of HIV-1 from the mother to fetus is known, but the molecular mechanism regulating this transmission is not fully characterized. The fetus is highly protected by the placenta, which does not permit microbial pathogens to cross the placental barrier. In the present study, a rat model was established to observe the effect of HIV-1 protein Nef on placental barrier. Evans blue dye was used to assay permeability of placental barrier and fourteen day pregnant Sprague Dawley rats were injected intravenously with 2% Evans blue dye along with various concentrations of recombinant Nef. After an hour, animals were sacrificed and dye migration was observed through the assimilation of peripheral blood into fetus. Interestingly, traces of recombinant Nef protein were detected in the embryo as well as amniotic fluid and amniotic membrane along with placenta and uterus. Our study indicates that recombinant HIV-1-Nef protein breaches the placental barrier and allows the migration of Evans blue dye to the growing fetus. Further the concentration of Nef protein in blood is directly proportional to the intensity of dye migration and to the amount of Nef protein detected in uterus, placenta, amniotic membrane, amniotic fluid and embryo. Based on this study, it can be concluded that the HIV-1 Nef protein has a direct effect on breaching of the placental barrier in the model we have established in this study. Our observations will be helpful to understand the molecular mechanisms related to this breach of placental barrier by Nef in humans and may be helpful to identify specific Nef inhibitors.
Collapse
Affiliation(s)
- Poonam Singh
- Toxicology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
| | - Saurabh Kumar Agnihotri
- Endocrinology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
| | - Mahesh Chandra Tewari
- Endocrinology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
| | - Sadan Kumar
- Toxicology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
| | - Monika Sachdev
- Endocrinology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
- * E-mail: (MS); (RK)
| | - Raj Kamal Tripathi
- Toxicology Division, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow, Uttar Pradesh, India
- * E-mail: (MS); (RK)
| |
Collapse
|
27
|
Johnson EL, Chakraborty R. Placental Hofbauer cells limit HIV-1 replication and potentially offset mother to child transmission (MTCT) by induction of immunoregulatory cytokines. Retrovirology 2012; 9:101. [PMID: 23217137 PMCID: PMC3524025 DOI: 10.1186/1742-4690-9-101] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 11/16/2012] [Indexed: 12/26/2022] Open
Abstract
Background Despite readily detectable levels of the HIV-1 (co)-receptors CD4, CCR5 and DC-SIGN on placental macrophages (Hofbauer Cells [HCs]), the rate of HIV-1 infection in utero in the absence of interventions is only 7% of exposed infants. Here, we examine the replication kinetics of human HCs to the primary isolate HIV-1BaL. We also determined the infectivity of HIV-1-exposed HCs by co-culturing with isolated cord and peripheral blood mononuclear cells [CBMCs, PBMCs]. To understand the limiting nature of HCs to HIV-1 replication, we examined the effect of endogenously secreted cytokines on replication kinetics. Results HCs have reduced ability to replicate HIV-1 in vitro (p < 0.01) and to transmit virus to CBMCs and PBMCs (p < 0.001 for both) compared to standard infections of MDMs. HCs were shown to release HIV-1 particles at levels comparable to MDMs, however exhibit significant decreases in viral transcription (gag and env), which may account for lower levels of HIV-1 replication. Un-stimulated HCs constitutively express significantly higher levels of regulatory cytokines, IL-10 and TGF-β, compared to MDMs (p < 0.01), which may contribute to immunoregulatory predominance at the placenta and possibly account for down-regulation of HIV-1 replication and infectivity by HCs. We further demonstrate that these regulatory cytokines inhibit HIV-1 replication within HCs in vitro. Conclusion HCs have reduced ability to replicate and disseminate R5-tropic HIV-1BaLin vitro and potentially offset mother to child transmission (MTCT) of HIV-1 by the induction of immunoregulatory cytokines. Despite the potential for migration and infectivity, HCs are not present in the neighboring fetal circulation. These results implicate HCs as important mediators of protection at the feto-maternal interface during ongoing HIV-1 exposure.
Collapse
Affiliation(s)
- Erica L Johnson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | | |
Collapse
|
28
|
|
29
|
Cavarelli M, Scarlatti G. Human immunodeficiency virus type 1 mother-to-child transmission and prevention: successes and controversies. J Intern Med 2011; 270:561-79. [PMID: 21929711 DOI: 10.1111/j.1365-2796.2011.02458.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The World Health Organization (WHO) and United Nations Programme on HIV/AIDS (UNAIDS) estimated that an additional 370 000 new human immunodeficiency virus type 1 (HIV-1) infections occurred in children in 2009, mainly through mother-to-child transmission (MTCT). Intrapartum transmission contributes to approximately 20-25% of infections, in utero transmission to 5-10% and postnatal transmission to an additional 10-15% of cases. MTCT accounts for only a few hundred infected newborns in those countries in which services are established for voluntary counselling and testing of pregnant women, and a supply of antiretroviral drugs is available throughout pregnancy with recommendations for elective Caesarean section and avoidance of breastfeeding. The single-dose nevirapine regimen has provided the momentum to initiate MTCT programmes in many resource-limited countries; however, regimens using a combination of antiretroviral drugs are needed also to effectively reduce transmission via breastfeeding.
Collapse
Affiliation(s)
- M Cavarelli
- Unit of Viral Evolution and Transmission, DITID, San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
30
|
Tandon R, SenGupta D, Ndhlovu LC, Vieira RGS, Jones RB, York VA, Vieira VA, Sharp ER, Wiznia AA, Ostrowski MA, Rosenberg MG, Nixon DF. Identification of human endogenous retrovirus-specific T cell responses in vertically HIV-1-infected subjects. J Virol 2011; 85:11526-31. [PMID: 21880743 PMCID: PMC3194955 DOI: 10.1128/jvi.05418-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/18/2011] [Indexed: 11/20/2022] Open
Abstract
Human endogenous retrovirus (HERV)-specific T cell responses in HIV-1-infected adults have been reported. Whether HERV-specific immunity exists in vertically HIV-1-infected children is unknown. We performed a cross-sectional analysis of HERV-specific T cell responses in 42 vertically HIV-1-infected children. HERV (-H, -K, and -L family)-specific T cell responses were identified in 26 of 42 subjects, with the greatest magnitude observed for the responses to HERV-L. These HERV-specific T cell responses were inversely correlated with the HIV-1 plasma viral load and positively correlated with CD4(+) T cell counts. These data indicate that HERV-specific T cells may participate in controlling HIV-1 replication and that certain highly conserved HERV-derived proteins may serve as promising therapeutic vaccine targets in HIV-1-infected children.
Collapse
Affiliation(s)
- Ravi Tandon
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Devi SenGupta
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Lishomwa C. Ndhlovu
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Raphaella G. S. Vieira
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - R. Brad Jones
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Vanessa A. York
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Vinicius A. Vieira
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Elizabeth R. Sharp
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| | - Andrew A. Wiznia
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Mario A. Ostrowski
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael G. Rosenberg
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Douglas F. Nixon
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California 94110
| |
Collapse
|
31
|
Weinberg A, Naglik JR, Kohli A, Tugizov SM, Fidel PL, Liu Y, Herzberg M. Innate immunity including epithelial and nonspecific host factors: workshop 1B. Adv Dent Res 2011; 23:122-9. [PMID: 21441493 DOI: 10.1177/0022034511399917] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The majority of HIV infections are initiated at mucosal sites. The oral mucosal tissue has been shown to be a potential route of entry in humans and primates. Whereas HIV RNA, proviral DNA, and infected cells are detected in the oral mucosa and saliva of infected individuals, it appears that the oral mucosa is not permissive for efficient HIV replication and therefore may differ in susceptibility to infection when compared to other mucosal sites. Since there is no definitive information regarding the fate of the HIV virion in mucosal epithelium, there is a pressing need to understand what occurs when the virus is in contact with this tissue, what mechanisms are in play to determine the outcome, and to what degree the mechanisms and outcomes differ between mucosal sites. Workshop 1B tackled 5 important questions to define current knowledge about epithelial cell-derived innate immune agents, commensal and endogenous pathogens, and epithelial cells and cells of the adaptive immune system and how they contribute to dissemination or resistance to HIV infection. Discovering factors that explain the differential susceptibility and resistance to HIV infection in mucosal sites will allow for the identification and development of novel protective strategies.
Collapse
Affiliation(s)
- A Weinberg
- Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Chen XQ, Liu C, Kong XH. The role of HIV replicative fitness in perinatal transmission of HIV. Virol Sin 2011; 26:147-55. [PMID: 21667335 DOI: 10.1007/s12250-011-3180-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/31/2011] [Indexed: 11/28/2022] Open
Abstract
Perinatal transmission of Human immunodeficiency virus (HIV), also called mother-to-child transmission (MTCT), accounts for 90% of infections in infants worldwide and occurs in 30%-45% of children born to untreated HIV-1 infected mothers. Among HIV-1 infected mothers, some viruses are transmitted from mothers to their infants while others are not. The relationship between virologic properties and the pathogenesis caused by HIV-1 remains unclear. Previous studies have demonstrated that one obvious source of selective pressure in the perinatal transmission of HIV-1 is maternal neutralizing antibodies. Recent studies have shown that viruses which are successfully transmitted to the child have growth advantages over those not transmitted, when those two viruses are grown together. Furthermore, the higher fitness is determined by the gp120 protein of the virus envelope. This suggests that the selective transmission of viruses with higher fitness occurred exclusively, regardless of transmission routes. There are many factors contributing to the selective transmission and HIV replicative fitness is an important one that should not be neglected. This review summarizes current knowledge of the role of HIV replicative fitness in HIV MTCT transmission and the determinants of viral fitness upon MTCT.
Collapse
Affiliation(s)
- Xue-Qing Chen
- Laboratory of Medical Molecular Virology, School of Medicine, Nankai University, Tianjin, China
| | | | | |
Collapse
|
33
|
Expresión diferencial en placenta de beta-defensinas humanas y detección de variantes alélicas en el gen DEFB1 de madres positivas para VIH-1. BIOMEDICA 2011. [DOI: 10.7705/biomedica.v31i1.335] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
34
|
Cavarelli M, Scarlatti G. HIV-1 co-receptor usage: influence on mother-to-child transmission and pediatric infection. J Transl Med 2011; 9 Suppl 1:S10. [PMID: 21284900 PMCID: PMC3105501 DOI: 10.1186/1479-5876-9-s1-s10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Viral CCR5 usage is not a predictive marker of mother to child transmission (MTCT) of HIV-1. CXCR4-using viral variants are little represented in pregnant women, have an increased although not significant risk of transmission and can be eventually also detected in the neonates. Genetic polymorphisms are more frequently of relevance in the child than in the mother. However, specific tissues as the placenta or the intestine, which are involved in the prevalent routes of infection in MTCT, may play an important role of selective barriers. The virus phenotype of the infected children, like that of adults, can evolve from R5 to CXCR4-using phenotype or remain R5 despite clinical progression to overt immune deficiency. The refined classification of R5 viruses into R5(narrow) and R5(broad) resolves the enigma of the R5 phenotype being associated with the state of immune deficiency. Studies are needed to address more in specific the relevance of these factors in HIV-1 MTCT and pediatric infection of non-B subtypes.
Collapse
Affiliation(s)
- Mariangela Cavarelli
- Unit of Viral Evolution and Transmission, Division of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | | |
Collapse
|
35
|
Coats KS, Boudreaux CE, Clay BT, Lockett NN, Scott VL. Placental immunopathology in the FIV-infected cat: a role for inflammation in compromised pregnancy? Vet Immunol Immunopathol 2009; 134:39-47. [PMID: 19896219 DOI: 10.1016/j.vetimm.2009.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In utero transmission of feline immunodeficiency virus (FIV) occurs frequently in queens experimentally infected with FIV-B-2542 and other FIV isolates. Fetal infection has been detected as early as 3-4 weeks gestation, and the incidence of fetal infection increases with progressing gestation. Reproductive failure occurs commonly, including fetal resorptions and developmentally-arrested fetuses, demonstrating that fetal demise occurs early in gestation. Precise, temporal immunomodulation within the placenta is essential for successful pregnancy. Placental Th1 and Th2 cytokines must be appropriately balanced, typically favoring Th2 cytokines at the maternal-fetal interface. Abnormal inflammatory cytokine expression often accompanies miscarriage. Regulatory T cells (Tregs) play an essential role in maternal tolerance of the semi-allogeneic fetus by suppressing inflammation. We are using the FIV-infected cat to examine the relationship between lentivirus-induced placental immunopathology and reproductive outcome. Using TaqMan real time reverse transcriptase (RT)-PCR, we measured relative expression of key immunomodulators in the placentas of FIV-B-2542-infected and control cats, including placentas from both viable and nonviable pregnancies. Our data associate significantly-increased expression of inflammatory cytokines with failed pregnancies, identify Treg markers in the placentas, and provide preliminary evidence that Tregs or other cells bearing similar activation markers may be involved in pregnancy maintenance. Our data suggest that placental inflammation in the FIV-infected cat may compromise pregnancy.
Collapse
Affiliation(s)
- Karen S Coats
- Department of Biological Sciences, P.O. Box GY, Mississippi State University, Mississippi State, MS 39762, USA.
| | | | | | | | | |
Collapse
|
36
|
Risk of equine infectious anemia virus disease transmission through in vitro embryo production using somatic cell nuclear transfer. Theriogenology 2009; 72:289-99. [DOI: 10.1016/j.theriogenology.2009.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 03/20/2009] [Accepted: 03/28/2009] [Indexed: 11/23/2022]
|
37
|
Abstract
OBJECTIVE Review the role and mechanism of in utero placental transmission of HIV-1. STUDY DESIGN A thorough review based on a literature search for publications relevant to this subject was performed using relevant search terms. Articles that describe the genetic and pathophysiology of vertical transmission have been acknowledged. The articles pertinent to the topic were selected to support the discussion. RESULTS Vertical transmission may occur through CD4+ endothelial tissues or CD4+ Hofbauer cells. Trophoblasts and villi have CD4 receptors, which make them potential candidates for HIV infection. Placental cytokines and chemokines influence HIV replication in trophoblasts. Genetic analysis of HIV-1 sequences verify the interaction of HIV-1 and placental tissue. The vertical transmission of HIV-1 characterized by selection of genotype variant that escape the mother's immune system. CONCLUSION Placental transmission of HIV-1 is a complex incompletely understood process which requires advanced studies. The available literature provides information with regards to the interactions of placental cells with HIV.
Collapse
|
38
|
de Vries BS, Peek MJ. Exploring the mechanisms of intrapartum transmission of HIV. Does elective caesarean section hold the key? BJOG 2008; 115:677-80. [PMID: 18410649 DOI: 10.1111/j.1471-0528.2008.01693.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- B S de Vries
- Department of Women and Babies, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| | | |
Collapse
|
39
|
Al-Adnani M, Sebire NJ. The role of perinatal pathological examination in subclinical infection in obstetrics. Best Pract Res Clin Obstet Gynaecol 2007; 21:505-21. [PMID: 17448728 DOI: 10.1016/j.bpobgyn.2007.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Infectious agents are associated with a wide range of obstetric complications and pathological processes affecting the placenta, membranes and fetus. In some cases there will be associated maternal symptoms and signs indicating an infectious aetiology, but in the majority such infection is subclinical, and specific diagnosis or confirmation is achieved following pathological examination of the delivered placenta and/or fetus. There are two major groups of microorganism-related mechanisms associated with significant perinatal morbidity and mortality. First, ascending genital-tract infection, almost always bacterial, which ranges from localized choriodecidual inflammation to frank chorioamnionitis with fetal sepsis; this is a major cause of mid-trimester miscarriage and severe preterm delivery, and more recent data suggest that it may also have potentially important effects via cytokine release mediating neonatal cerebral injury. Second, haematogenous spread of maternal systemic infection--bacterial, viral or parasitic--which may result in isolated placental effects or transmission to the fetus with associated developmental abnormalities and neonatal complications. In many cases distinctive histopathological findings are described, and in addition a wide range of techniques is now available for culture and microscopy to confirm these diagnoses; such techniques include highly specific immunohistochemical markers and sensitive molecular diagnostic techniques such as the polymerase chain reaction. It is likely that with increasingly widespread availability of these investigative approaches to obstetric pathology, a greater understanding of the role of infectious agents in obstetric complications will become apparent.
Collapse
Affiliation(s)
- M Al-Adnani
- Department of Paediatric Pathology, Great Ormond Street Hospital, Camelia Botnar Laboratories, Great Ormond Street, London WC1N 3JH, UK
| | | |
Collapse
|
40
|
Vidricaire G, Tremblay MJ. A Clathrin, Caveolae, and Dynamin-independent Endocytic Pathway Requiring Free Membrane Cholesterol Drives HIV-1 Internalization and Infection in Polarized Trophoblastic Cells. J Mol Biol 2007; 368:1267-83. [PMID: 17395200 DOI: 10.1016/j.jmb.2007.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/02/2007] [Accepted: 03/02/2007] [Indexed: 02/06/2023]
Abstract
In human trophoblastic cells, a correlation between early endosomal trafficking of HIV-1 and virus infection was previously documented. However, if HIV-1 is massively internalized in these cells, the endocytic pathway(s) responsible for viral uptake is still undefined. Here we address this vital question. Amongst all the putative endocytic pathways present in polarized trophoblastic cells, we demonstrate that HIV-1 infection of these cells is independent of clathrin-mediated endocytosis and macropinocytosis. Importantly, treatment with the cholesterol-sequestering drug filipin severely impairs virus internalization, whereas the cholesterol-depleting compound methyl-beta-cyclodextrin has no impact on this pathway. Moreover, viral internalization is unaffected by overexpression of a mutant dynamin 2 or treatment with a kinase or tyrosine phosphatase inhibitor. Thus, HIV-1 infection in polarized trophoblastic cells occurs primarily via a clathrin, caveolae, and dynamin-independent pathway requiring free cholesterol. Notably, even though HIV-1 did not initially co-localize with transferrin, some virions migrate at later time points to transferrin-enriched endosomes, suggesting an unusual transit from the non-classical pathway to early endosomes. Finally, virus internalization in these cells does not involve the participation of microtubules but relies partly on actin filaments. Collectively these findings provide unprecedented information on the route of HIV-1 internalization in polarized human trophoblasts.
Collapse
Affiliation(s)
- Gaël Vidricaire
- Research Center in Infectious Diseases, CHUL Research Center, and Department of Medical Biology, Faculty of Medicine, Laval University, Quebec, Canada G1V 4G2
| | | |
Collapse
|
41
|
Lehman DA, Farquhar C. Biological mechanisms of vertical human immunodeficiency virus (HIV-1) transmission. Rev Med Virol 2007; 17:381-403. [PMID: 17542053 DOI: 10.1002/rmv.543] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the absence of interventions, 30-45% of exposed infants acquire human immunodeficiency virus type 1 (HIV-1) through mother-to-child transmission. It remains unclear why some infants become infected while others do not, despite significant exposure to HIV-1 in utero, during delivery and while breastfeeding. Here we discuss the correlates of vertical transmission with an emphasis on factors that increase maternal HIV-1 levels, either systemically or locally in genital secretions and breast milk. Immune responses may influence maternal viral load, and data suggest that maternal neutralising antibodies reduce infection rates. In addition, infants may be capable of mounting HIV-specific cellular immune responses. We propose that both humoral and cellular responses are necessary to reduce infection because cell-free as well as cell-associated virus appears to play a role in vertical transmission. These distinct forms of the virus may be targeted most effectively by different components of the immune system. We also discuss the use of antiretrovirals to reduce transmission, focusing on the mechanisms of action of regimens currently used in developing country settings. We conclude that prevention relies not only on reducing maternal HIV-1 levels within blood, genital tract and breast milk, but also on pre- and/or post-exposure prophylaxis to the infant. However, HIV-1 has the capacity to mutate under drug pressure and rapidly acquires mutations conferring antiretroviral resistance. This review concludes with data on persistence of low-level resistance after delivery as well as recent guidelines for maternal and infant regimens designed to limit resistance.
Collapse
Affiliation(s)
- Dara A Lehman
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | |
Collapse
|
42
|
|
43
|
Huang Y, Zhu XY, Du MR, Wu X, Wang MY, Li DJ. Chemokine CXCL16, a scavenger receptor, induces proliferation and invasion of first-trimester human trophoblast cells in an autocrine manner. Hum Reprod 2006; 21:1083-91. [PMID: 16431903 DOI: 10.1093/humrep/dei436] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate whether CXCL16/CXCR6, a newly identified chemokine pair, is expressed in first-trimester human placenta and whether they affect the trophoblast cell biology, since we have found CXCR6 highly transcribed in first-trimester human trophoblast cells previously. METHODS We analysed the transcription and translation of CXCR6 and CXCL16 in purified first-trimester human trophoblast cells by real-time RT-PCR and immunochemical staining. We then examined the kinetic secretion of CXCL16 in the supernatant of primary-cultured trophoblast by enzyme-linked immunosorbent assay. We further investigated effects of CXCL16 on the proliferation and invasion of trophoblast cells in vitro. RESULTS We found the chemokine pair CXCL16/CXCR6 was transcribed and translated in first-trimester trophoblast cells and JAR line. In addition, the primary-cultured trophoblasts secreted CXCL16 spontaneously and continuously in 100-h culture. Treating trophoblasts with CXCL16 induced marked proliferation and invasion in vitro. CONCLUSION The findings from this study have demonstrated for the first time that CXCR6 and CXCL16 are co-expressed by first-trimester human trophoblast cells and stimulate their proliferation and invasion in an autocrine/paracrine manner. It suggests that CXCL16 plays important roles in human extravillous cytotrophoblast invasion and placentation.
Collapse
MESH Headings
- Autocrine Communication
- Cell Line
- Cell Proliferation/drug effects
- Chemokine CXCL16
- Chemokines, CXC/metabolism
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Female
- Humans
- Immunohistochemistry
- Placenta/cytology
- Placenta/metabolism
- Placentation/drug effects
- Placentation/physiology
- Pregnancy
- Pregnancy Trimester, First/physiology
- RNA, Messenger/metabolism
- Receptors, CXCR6
- Receptors, Chemokine/metabolism
- Receptors, Scavenger/metabolism
- Receptors, Scavenger/physiology
- Receptors, Virus/metabolism
- Trophoblasts/cytology
- Trophoblasts/metabolism
- Trophoblasts/physiology
Collapse
Affiliation(s)
- Yu Huang
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
44
|
Bhoopat L, Khunamornpong S, Lerdsrimongkol P, Sirivatanapa P, Sethavanich S, Limtrakul A, Gomutbuthra V, Kajanavanich S, Thorner PS, Bhoopat T. Effectiveness of Short-Term and Long-Term Zidovudine Prophylaxis on Detection of HIV-1 Subtype E in Human Placenta and Vertical Transmission. J Acquir Immune Defic Syndr 2005; 40:545-50. [PMID: 16284530 DOI: 10.1097/01.qai.0000185572.90849.4d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antiretroviral treatment with zidovudine (ZDV) from the 14th week until the end of pregnancy has markedly reduced the vertical transmission rate of HIV-1 in Europe and North America. A shorter duration of treatment has reduced this rate in Africa and Southeast Asia to a lesser degree. In Southeast Asia, subtype E is the major subtype rather than subtype B as in Western countries. The goals of this study were to determine the optimal duration of ZDV prophylaxis for subtype E and to confirm its effectiveness at the histologic level. Fifty pregnant women seropositive for HIV-1 subtype E were given ZDV prophylaxis consisting of 300 mg administered twice daily, switching to 300 mg administered every 3 hours from the onset of labor until delivery. Twenty-seven received "short-term" ZDV lasting 14 to 35 days before delivery, whereas the other 23 received "long-term" ZDV lasting 62 to 92 days. The effectiveness of ZDV prophylaxis was assessed by detection of HIV-1 in the placenta using in situ polymerase chain reaction (PCR). All babies in this study were tested up to one year of age. Three were not positive until after one month of age, but one was positive as a neonate. Four neonates were positive for HIV-1 as detected by PCR on peripheral blood, including one in the neonatal period. All cases were from the short-term prophylaxis group. Decidual glandular epithelial cells were the only cell type in the placenta that expressed HIV proviral DNA under ZDV prophylaxis. Sixty-seven percent of placentas in the short-term ZDV group showed more than occasional positive cells compared with 22% in the group receiving long-term ZDV prophylaxis (P < 0.02). This is first study to compare the effectiveness of short-term and long-term ZDV prophylaxis with respect to the presence of HIV in the placenta. Our study shows that longer (at least 60 days) prophylaxis is more effective in reducing HIV expression in the placenta and is associated with reduced transmission to neonates.
Collapse
Affiliation(s)
- Lertlakana Bhoopat
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Vidricaire G, Tremblay MJ. Rab5 and Rab7, but Not ARF6, Govern the Early Events of HIV-1 Infection in Polarized Human Placental Cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:6517-30. [PMID: 16272306 DOI: 10.4049/jimmunol.175.10.6517] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Trophoblasts, the structural cells of the placenta, are thought to play a determinant role in in utero HIV type 1 (HIV-1) transmission. We have accumulated evidence suggesting that HIV-1 infection of these cells is associated with uptake by an unusual clathrin/caveolae-independent endocytic pathway and that endocytosis is followed by trafficking through multiple organelles. Furthermore, part of this trafficking involves the transit of HIV-1 from transferrin-negative to EEA1 and transferrin-positive endosomes, suggesting a merger from nonclassical to classical endocytic pathways in these cells. In the present article, the relationship between the presence of HIV-1 within specific endosomes and infection was studied. We demonstrate that viral infection is virtually lost when endosome inhibitors are added shortly after exposure to HIV-1. Thus, contrary to what is seen in CD4+ T lymphocytes, the initial presence of HIV-1 within the endosomes is mandatory for infection to take place. Importantly, this process is independent of the viral envelope proteins gp120 and gp41. The Rab family of small GTPases coordinates the vesicular transport between the different endocytic organelles. Experiments performed with various expression vectors indicated that HIV-1 infection in polarized trophoblasts relies on Rab5 and Rab7 without the contribution of Arf6 or Rab11. Furthermore, we conclude that Rab5 drives movements from raft-rich region to early endosomes, and this transit is required for subsequently reaching late endosomes via Rab7. This complex trafficking is mandatory for HIV-1 infection to proceed in human polarized trophoblasts.
Collapse
Affiliation(s)
- Gaël Vidricaire
- Research Center in Infectious Diseases, Centre Hospitalier de l'Université Laval Research Center, and Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | | |
Collapse
|
46
|
Bhoopat L, Khunamornpong S, Sirivatanapa P, Rithaporn T, Lerdsrimongkol P, Thorner PS, Bhoopat T. Chorioamnionitis is associated with placental transmission of human immunodeficiency virus-1 subtype E in the early gestational period. Mod Pathol 2005; 18:1357-64. [PMID: 15846390 DOI: 10.1038/modpathol.3800418] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The frequency and the cellular basis for HIV-1 transmission from mother to child in the early gestational period are poorly understood. We compared the placentas of 24 women seropositive for HIV-1 subtype E and who had not received any antiretroviral drugs, to placentas of 25 seronegative women. All placentas were obtained during therapeutic abortion at 6-23 weeks gestation. Placentas and fetal organs were examined by routine light microscopy, immunostaining for p24 capsid protein, and in situ PCR to localize which cells were infected with HIV-1 subtype E. The number of previous abortions was not a factor in placental HIV infection since this number was higher in seronegative women (P < 0.01). There were no significant differences between the placentas of the two groups with respect to presence of chorioamnionitis, villitis, villous stromal fibrosis, infarction, abnormal villous maturation, deciduitis or decidual necrosis. HIV-1 subtype E was detected in up to 83% of placentas, either by immunostaining or in situ PCR, in trophoblast, villous stromal cells, Hofbauer cells, decidual and decidual glandular epithelium. Fetal organs were positive for HIV in 30% (6/20) of cases. There was a significant association between transmission of HIV to the fetus and the histologic findings of chorioamnionitis, plasmacellular deciduitis and decidual cell necrosis. This is the first report showing an association of chorioamnionitis with early in utero transmission of HIV-1 subtype E. This may help explain the cases of in utero transmission that persist despite antiretroviral prophylaxis, given that therapy is started in the late gestational period.
Collapse
Affiliation(s)
- Lertlakana Bhoopat
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | | | | | | | | | | | | |
Collapse
|
47
|
Coats KS. The Feline Immunodeficiency Virus-Infected Cat: A Model for Lentivirus-induced Placental Immunopathology and Reproductive Failure (Mini-Review). Am J Reprod Immunol 2005; 54:169-85. [PMID: 16135008 DOI: 10.1111/j.1600-0897.2005.00296.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Pediatric human immunodeficiency virus (HIV) infection is largely a result of transplacental transmission, and pregnancy perturbation is more frequent in HIV-infected women. Dysregulation of placental immunology may occur during HIV infection, possibly facilitating HIV vertical transfer and miscarriage. The (FIV)-infected cat is a useful small-animal model for HIV pathogenesis because the viruses share common biological and clinical features. Transplacental transmission is readily achieved experimentally, resulting in a high proportion of infected offspring and frequent reproductive failure. METHOD OF STUDY We are using this model to examine lentivirus-induced placental immunopathology to determine the role aberrant immunology plays in intrauterine transmission and pregnancy perturbation. RESULTS Kittens were cesarean delivered from FIV-B-2542-infected and control queens at week 8 gestation (1 week short of term), and placental and fetal specimens were collected. On average, control queens delivered 3.8 kittens/litter, and 1 of 31 kittens (3.2%) was non-viable. FIV-infected queens produced 2.7 kittens/litter with 15 of 25 fetuses (60%) non-viable. The virus was detected in 14 of 15 placentas (93%) and 21 of 22 fetuses (95%) using polymerase chain reaction (PCR). Using a one-step, real time reverse transcriptase (RT)-PCR, we measured expression of representative placental T helper 1 (Th1) cytokines, interleukin (IL)-1beta and interferon (IFN)-gamma, a Th2 cytokine, IL-10, and chemokine receptor CXCR4. A comparison of placental cytokine expression between infected and control queens did not reveal differences between the two groups. However, elevated expression of Th1 cytokines and increased Th1/Th2 ratios (IL-1beta/IL-10) occurred in placentas from resorptions, indicating that increased placental Th1 cytokine expression was associated with pregnancy failure in the FIV-infected cat. CONCLUSION The potential to establish efficient FIV in utero transmission, coupled with the parallels in immunopathology between FIV-infected cats and HIV-infected humans, suggests the usefulness of the FIV-infected cat as a cost-effective, small-animal model to study lentivirus-induced immunopathology, transplacental infection, and reproductive failure.
Collapse
Affiliation(s)
- Karen S Coats
- Department of Biological Sciences and College of Veterinary Medicine, Mississippi State University, PO Box GY, MS 39762, USA.
| |
Collapse
|
48
|
Vidricaire G, Tremblay MJ. [For a better understanding of the mechanisms involved in vertical transmission of HIV]. Med Sci (Paris) 2005; 20:784-7. [PMID: 15361345 DOI: 10.1051/medsci/2004208-9784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Maternal-infant transmission of human immunodeficiency virus-1 (HIV) is the primary cause of this retrovirus infection in neonates. The mechanisms of vertical transmission of HIV, in particular in utero transmission, remain poorly defined. Trophoblastic cells from the placenta are thought to be a target of HIV infection and/or may be utilized by the virus to be transported across the placental barrier by a process known as transcytosis. The vertical transmission of HIV (via infection or transcytosis) may be either favoured or inhibited by factors related to both the viral phenotype and the cellular environment.
Collapse
Affiliation(s)
- Gaël Vidricaire
- Laboratoire d'immunorétrovirologie humaine, Centre de recherche en infectiologie, Hôpital CHUL, Centre Hospitalier Universitaire de Québec, Département de biologie médicale, Faculté de Médecine, Université Laval, Sainte-Foy, Canada
| | | |
Collapse
|
49
|
Weaver CC, Burgess SC, Nelson PD, Wilkinson M, Ryan PL, Nail CA, Kelly-Quagliana KA, May ML, Reeves RK, Boyle CR, Coats KS. Placental immunopathology and pregnancy failure in the FIV-infected cat. Placenta 2005; 26:138-47. [PMID: 15708115 DOI: 10.1016/j.placenta.2004.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2004] [Indexed: 11/23/2022]
Abstract
Placental HIV infections frequently result in infected babies or miscarriage. Aberrant placental cytokine expression during HIV infections may facilitate transplacental viral transmission or pregnancy perturbation. The feline immunodeficiency virus (FIV)-infected cat is a model for HIV infections due to similarities in biology and clinical disease. The purpose of this study was to evaluate placental immunomodulator expression and reproductive outcome using the FIV-infected cat model. Kittens were cesarean delivered from FIV-B-2542-infected and control queens near term; placental and fetal tissues were collected. Real-time RT-PCR was used to measure expression of representative placental Th1 cytokines, interleukin-1beta (IL-1beta) and interferon-gamma (IFN-gamma), a Th2 cytokine, IL-10, and chemokine receptor CXCR4. On average, control queens delivered 3.8 kittens/litter; 1 of 31 kittens (3.2%) was non-viable. FIV-infected queens produced 2.7 kittens/litter; 15 of 25 concepti (60%) were non-viable. FIV was detected in 14 of 15 placentas (93%) and 21 of 22 fetuses (95%) using PCR. Placental immunomodulator expression did not differ significantly when placentas from infected cats were compared to those of control cats. However, elevated expression of Th1 cytokines and increased Th1/Th2 ratios (IL-1beta/IL-10) occurred in placentas from resorptions. Therefore, increased placental Th1 cytokine expression was associated with pregnancy failure in the FIV-infected cat.
Collapse
Affiliation(s)
- C C Weaver
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Vidricaire G, Tardif MR, Tremblay MJ. The low viral production in trophoblastic cells is due to a high endocytic internalization of the human immunodeficiency virus type 1 and can be overcome by the pro-inflammatory cytokines tumor necrosis factor-alpha and interleukin-1. J Biol Chem 2003; 278:15832-41. [PMID: 12604606 DOI: 10.1074/jbc.m210470200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maternal-infant transmission of human immunodeficiency virus type-1 (HIV-1) is the primary cause of this retrovirus infection in neonates. Trophoblasts have been proposed to play a critical role in modulating virus spread to the fetus. This paper addresses the mechanism of HIV-1 biology in trophoblastic cells. The trophoblastic cell lines BeWo, JAR, and JEG-3 were infected with reporter HIV-1 particles pseudotyped with envelope glycoproteins from the vesicular stomatitis virus or various strains of HIV-1. We demonstrate that despite a high internalization process of HIV-1 and no block in viral production, HIV-1 established a limited infection of trophoblasts with the production of very few progeny viruses. The factor responsible for this restriction to virus replication in such a cellular microenvironment is that the intracellular p24 is concentrated predominantly in endosomal vesicles following HIV-1 entry. HIV-1 transcription and virus production of infectious particles were both augmented upon treatment of trophoblasts with tumor necrosis factor-alpha and interleukin-1. However, the amount of progeny virions released by trophoblasts infected with native HIV-1 virions was so low even in the presence of pro-inflammatory cytokines that a co-culture step with indicator cells was necessary to detect virus production. Collectively these data illustrate for the first time that the natural low permissiveness of trophoblasts to productive HIV-1 infection is because of a restriction in the mode of entry, and such a limitation can be overcome with physiologic doses of tumor necrosis factor-alpha and interleukin-1, which are both expressed by the placenta, in conjunction with cell-cell contact. Considering that there is a linear correlation between viral load and HIV-1 vertical transmission, the environment may thus contribute to the propagation of HIV-1 across the placenta.
Collapse
Affiliation(s)
- Gaël Vidricaire
- Centre de Recherche en Infectiologie, Hôpital CHUL, Centre Hospitalier Universitaire de Québec, 2705 Boulevard Laurier, Ste-Foy, Québec G1V 4G2, Canada
| | | | | |
Collapse
|