51
|
Abstract
Seminal fluid is often assumed to have just one function in mammalian reproduction, delivering sperm to fertilize oocytes. But seminal fluid also transmits signaling agents that interact with female reproductive tissues to facilitate conception and .pregnancy. Upon seminal fluid contact, female tissues initiate a controlled inflammatory response that affects several aspects of reproductive function to ultimately maximize the chances of a male producing healthy offspring. This effect is best characterized in mice, where the female response involves several steps. Initially, seminal fluid factors cause leukocytes to infiltrate the female reproductive tract, and to selectively target and eliminate excess sperm. Other signals stimulate ovulation, induce an altered transcriptional program in female tract tissues that modulates embryo developmental programming, and initiate immune adaptations to promote receptivity to implantation and placental development. A key result is expansion of the pool of regulatory T cells that assist implantation by suppressing inflammation, mediating tolerance to male transplantation antigens, and promoting uterine vascular adaptation and placental development. Principal signaling agents in seminal fluid include prostaglandins and transforming growth factor-β. The balance of male signals affects the nature of the female response, providing a mechanism of ‟cryptic female choiceˮ that influences female reproductive investment. Male-female seminal fluid signaling is evident in all mammalian species investigated including human, and effects of seminal fluid in invertebrates indicate evolutionarily conserved mechanisms. Understanding the female response to seminal fluid will shed new light on infertility and pregnancy disorders and is critical to defining how events at conception influence offspring health.
Collapse
Affiliation(s)
- John E Schjenken
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
52
|
Scott NA, Mann ER. Regulation of mononuclear phagocyte function by the microbiota at mucosal sites. Immunology 2020; 159:26-38. [PMID: 31777068 PMCID: PMC6904663 DOI: 10.1111/imm.13155] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
Mucosal tissues contain distinct microbial communities that differ drastically depending on the barrier site, and as such, mucosal immune responses have evolved to be tailored specifically for their location. Whether protective or regulatory immune responses against invading pathogens or the commensal microbiota occur is controlled by local mononuclear phagocytes (MNPs). Comprising macrophages and dendritic cells (DCs), the functions of these cells are highly dependent on the local environment. For example, the intestine contains the greatest bacterial load of any site in the body, and hence, intestinal MNPs are hyporesponsive to bacterial stimulation. This is thought to be one of the major mechanisms by which harmful immune responses directed against the trillions of harmless bacteria that line the gut lumen are avoided. Regulation of MNP function by the microbiota has been characterized in the most depth in the intestine but there are several mucosal sites that also contain their own microbiota. In this review, we present an overview of how MNP function is regulated by the microbiota at mucosal sites, highlighting recent novel pathways by which this occurs in the intestine, and new studies elucidating these interactions at mucosal sites that have been characterized in less depth, including the urogenital tract.
Collapse
Affiliation(s)
- Nicholas A. Scott
- Lydia Becker Institute of Immunology and InflammationUniversity of ManchesterManchesterUK
- Manchester Collaborative Centre for Inflammation ResearchFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Elizabeth R. Mann
- Lydia Becker Institute of Immunology and InflammationUniversity of ManchesterManchesterUK
- Manchester Collaborative Centre for Inflammation ResearchFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
53
|
Pudney J, Wangu Z, Panther L, Fugelso D, Marathe JG, Sagar M, Politch JA, Anderson DJ. Condylomata Acuminata (Anogenital Warts) Contain Accumulations of HIV-1 Target Cells That May Provide Portals for HIV Transmission. J Infect Dis 2019; 219:275-283. [PMID: 30137482 DOI: 10.1093/infdis/jiy505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/16/2018] [Indexed: 11/13/2022] Open
Abstract
Background Condylomata acuminata (anogenital warts [AGWs]) are prevalent in human immunodeficiency virus (HIV)-infected individuals and sexually active populations at risk for HIV acquisition and have been associated with HIV transmission. We compared AGW specimens to control tissue specimens for abundance, types, and location of HIV target cells and for susceptibility to HIV infection in vitro, to provide biologic evidence that AGWs facilitate HIV transmission. Methods We used immunohistologic staining to identify HIV target cells in AGW and control specimens. We also inoculated HIV in vitro into AGW and control specimens from HIV-negative men and assessed infection by means of TZM-bl and p24 assays. Results CD1a+ dendritic cells, CD4+ T cells, and macrophages were significantly more abundant in the epidermis of AGW specimens than control specimens. These HIV target cells also often appeared in large focal accumulations in the dermis of AGW specimens. Two of 8 AGW specimens versus 0 of 8 control specimens showed robust infection with HIV in vitro. Conclusions Compared with normal skin, AGWs contain significantly higher concentrations of HIV target cells that may be susceptible to HIV infection. Condylomata may thus promote HIV transmission, especially in the setting of typical lesion vascularity and friability. Prevention or treatment of AGWs may decrease the sexual transmission of HIV.
Collapse
Affiliation(s)
- Jeffrey Pudney
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston
| | - Zoon Wangu
- Division of Pediatric Infectious Diseases and Immunology, UMass Memorial Children's Medical Center, Worcester, Massachusetts
| | - Lori Panther
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston
| | - Dana Fugelso
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston
| | - Jai G Marathe
- Department of Medicine, Boston University School of Medicine, Boston
| | - Manish Sagar
- Department of Medicine, Boston University School of Medicine, Boston
| | - Joseph A Politch
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston
| | - Deborah J Anderson
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston.,Department of Medicine, Boston University School of Medicine, Boston
| |
Collapse
|
54
|
Vickram AS, Samad HA, Latheef SK, Chakraborty S, Dhama K, Sridharan TB, Sundaram T, Gulothungan G. Human prostasomes an extracellular vesicle - Biomarkers for male infertility and prostrate cancer: The journey from identification to current knowledge. Int J Biol Macromol 2019; 146:946-958. [PMID: 31730983 DOI: 10.1016/j.ijbiomac.2019.09.218] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are gaining attention among the cell biologists and researchers over the last two decades. Prostasomes are considered to be (Evs) secreted by prostate epithelial cells into the semen during emission or ejaculation. Prostasomes contain various proteins required for immune regulation namely, amino and dipeptidyl peptidase; endopeptidase (neutral); decay accelerating factor; angiotensin-converting enzyme. Sperm cells need a few prerequisites in order to fertilize the egg. The role of prostasomes in enhancing the male fertility was reviewed extensively throughout the manuscript. Also, prostasomes have an immunosuppressive, immunomodulatory, antibacterial role in the female reproductive tract, and in some cases they can be used as immunocontraceptive agent to regulate the fertility status. This review will give insights to many active researchers in the field of prostasomal research and male infertility/fertility research. This review will open many unanswered mechanisms of prostasomes with respect to structure-function analysis, fatty acids patterns in diagnosis as well as prognosis of male infertility/fertility. More scientific reports are in need to support the mechanism of prostasomes and its role in immunomodulation. The development of prostasomes as a biomarker for the prostate cancer is still miserable with a lot of controversial results by various researchers.
Collapse
Affiliation(s)
- A S Vickram
- Saveetha School of Engineering, Department of Biotechnology, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Chennai 602 105, India.
| | - Hari Abdul Samad
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India
| | - Shyma K Latheef
- Immunology Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243 122, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, R.K. Nagar, West Tripura 799008, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - T B Sridharan
- Department of Biotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Thanigaivel Sundaram
- Saveetha School of Engineering, Department of Biomedical Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Chennai 602 105, India
| | - G Gulothungan
- Saveetha School of Engineering, Department of Biomedical Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Chennai 602 105, India
| |
Collapse
|
55
|
Protective Effect of Probiotic Bacteria and Estrogen in Preventing HIV-1-Mediated Impairment of Epithelial Barrier Integrity in Female Genital Tract. Cells 2019; 8:cells8101120. [PMID: 31546582 PMCID: PMC6829272 DOI: 10.3390/cells8101120] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 01/18/2023] Open
Abstract
Approximately 40% of global HIV-1 transmission occurs in the female genital tract (FGT) through heterosexual transmission. Epithelial cells lining the FGT provide the first barrier to HIV-1 entry. Previous studies have suggested that certain hormonal contraceptives or a dysbiosis of the vaginal microbiota can enhance HIV-1 acquisition in the FGT. We examined the effects of lactobacilli and female sex hormones on the barrier functions and innate immune responses of primary endometrial genital epithelial cells (GECs). Two probiotic strains, Lactobacillus reuteri RC-14 and L. rhamnosus GR-1, were tested, as were sex hormones estrogen (E2), progesterone (P4), and the hormonal contraceptive medroxyprogesterone acetate (MPA). Our results demonstrate that probiotic lactobacilli enhance barrier function without affecting cytokines. Treatment of GECs with MPA resulted in reduced barrier function. In contrast, E2 treatment enhanced barrier function and reduced production of proinflammatory cytokines. Comparison of hormones plus lactobacilli as a pre-treatment prior to HIV exposure revealed a dominant effect of lactobacilli in preventing loss of barrier function by GECs. In summary, the combination of E2 and lactobacilli had the best protective effect against HIV-1 seen by enhancement of barrier function and reduction in proinflammatory cytokines. These studies provide insights into how probiotic lactobacilli in the female genital microenvironment can alter HIV-1-mediated barrier disruption and how the combination of E2 and lactobacilli may decrease susceptibility to primary HIV infection.
Collapse
|
56
|
Comparative transcriptome analysis of the human endocervix and ectocervix during the proliferative and secretory phases of the menstrual cycle. Sci Rep 2019; 9:13494. [PMID: 31530865 PMCID: PMC6749057 DOI: 10.1038/s41598-019-49647-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 08/24/2019] [Indexed: 12/18/2022] Open
Abstract
Despite extensive studies suggesting increased susceptibility to HIV during the secretory phase of the menstrual cycle, the molecular mechanisms involved remain unclear. Our goal was to analyze transcriptomes of the endocervix and ectocervix during the proliferative and secretory phases using RNA sequencing to explore potential molecular signatures of susceptibility to HIV. We identified 202 differentially expressed genes (DEGs) between the proliferative and secretory phases of the cycle in the endocervix (adjusted p < 0.05). The biofunctions and pathways analysis of DEGs revealed that cellular assembly and epithelial barrier function in the proliferative phase and inflammatory response/cellular movement in the secretory phase were among the top biofunctions and pathways. The gene set enrichment analysis of ranked DEGs (score = log fold change/p value) in the endocervix and ectocervix revealed that (i) unstimulated/not activated immune cells gene sets positively correlated with the proliferative phase and negatively correlated with the secretory phase in both tissues, (ii) IFNγ and IFNα response gene sets positively correlated with the proliferative phase in the ectocervix, (iii) HIV restrictive Wnt/β-catenin signaling pathway negatively correlated with the secretory phase in the endocervix. Our data show menstrual cycle phase-associated changes in both endocervix and ectocervix, which may modulate susceptibility to HIV.
Collapse
|
57
|
Sharkey DJ, Glynn DJ, Schjenken JE, Tremellen KP, Robertson SA. Interferon-gamma inhibits seminal plasma induction of colony-stimulating factor 2 in mouse and human reproductive tract epithelial cells. Biol Reprod 2019; 99:514-526. [PMID: 29596569 DOI: 10.1093/biolre/ioy071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/26/2018] [Indexed: 12/26/2022] Open
Abstract
Seminal fluid interacts with the female reproductive tract to initiate a permissive immune response that facilitates embryo implantation and pregnancy success. The immune-regulatory cytokine interferon-γ (IFNG), which can be elevated in seminal plasma, is associated with reduced fertility. Here, we investigated how IFNG influences the female immune response to seminal fluid. In human Ect1 cervical epithelial cells, IFNG added at physiologically relevant concentrations substantially impaired seminal plasma-induced synthesis of key cytokines colony-stimulating factor 2 (CSF2) and interleukin-6 (IL6). Seminal fluid-induced CSF2 synthesis was also suppressed in the uterus of mice in vivo, when IFNG was delivered transcervically 12 h after mating. Transforming growth factor B1 (TGFB1) is the major seminal fluid signaling factor which elicits CSF2 induction, and IFNG exhibited potent dose-dependent suppression of CSF2 synthesis induced by TGFB1 in murine uterine epithelial cells in vitro. Similarly, IFNG suppressed TGFB1-mediated CSF2 induction in Ect1 cells and human primary cervical epithelial cells; however, IL6 regulation by IFNG was independent of TGFB1. Quantitative PCR confirmed that CSF2 regulation by IFNG in Ect1 cells occurs at the gene transcription level, secondary to IFNG suppression of TGFBR2 encoding TGFB receptor 2. Conversely, TGFB1 suppressed IFNG receptor 1 and 2 genes IFNGR1 and IFNGR2. These data identify IFNG as a potent inhibitor of the TGFB-mediated seminal fluid interaction with relevant reproductive tract epithelia in mice and human. These findings raise the prospect that IFNG in the male partner's seminal fluid impairs immune adaptation for pregnancy following coitus in women.
Collapse
Affiliation(s)
- David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Danielle J Glynn
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kelton P Tremellen
- Repromed Pty Ltd, Dulwich, South Australia, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
58
|
Cavrois M, Hilton JF, Roan NR, Takeda M, Seidman D, Averbach S, Chang E, Raman N, Greenblatt R, Shacklett BL, Smith-McCune K. Effects of the levonorgestrel-containing intrauterine device, copper intrauterine device, and levonorgestrel-containing oral contraceptive on susceptibility of immune cells from cervix, endometrium and blood to HIV-1 fusion measured ex vivo. PLoS One 2019; 14:e0221181. [PMID: 31437197 PMCID: PMC6705759 DOI: 10.1371/journal.pone.0221181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
Globally, HIV/AIDS is a leading cause of morbidity worldwide among reproductive-aged cisgender women, highlighting the importance of understanding effects of contraceptives on HIV-1 risk. Some observational studies suggest there may be an increased risk of HIV-1 acquisition among women using the long-acting injectable progestin contraceptive, depo-medroxyprogesterone acetate. The potential mechanism of this susceptibility is unclear. There are few data on the role of the upper female reproductive tract in HIV-1 transmission, and the mechanisms of HIV-1 infection are likely to differ in the upper compared to the lower reproductive tract due to differences in tissue composition and variable effects of sex steroids on mucosal immune cell distribution and activity. In this study, we measured the susceptibility of mucosal immune cells from the upper female reproductive tract to HIV-1 entry using the virion-based HIV-1 fusion assay in samples from healthy female volunteers. We studied 37 infectious molecular clones for their ability to fuse to cells from endometrial biopsies in three participants and found that subtype (B or C) and origin of the virus (transmitted founder or chronic control) had little influence on HIV-1 fusion susceptibility. We studied the effect of contraceptives on HIV-1 susceptibility of immune cells from the cervix, endometrium and peripheral blood by comparing fusion susceptibility in four groups: users of the copper intrauterine device (IUD), levonorgestrel-containing oral contraceptive, levonorgestrel-containing IUD and unexposed controls (n = 58 participants). None of the contraceptives was associated with higher rates of HIV-1 entry into female reproductive tract cells compared to control samples from the mid-luteal phase.
Collapse
Affiliation(s)
- Marielle Cavrois
- Gladstone Institute of Virology and Immunology, San Francisco, California, United States of America
| | - Joan F. Hilton
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| | - Nadia R. Roan
- Gladstone Institute of Virology and Immunology, San Francisco, California, United States of America
- Department of Urology, University of California San Francisco, San Francisco, California, United States of America
| | - Margaret Takeda
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Dominika Seidman
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Sarah Averbach
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California, United States of America
| | - Eric Chang
- Gladstone Institute of Virology and Immunology, San Francisco, California, United States of America
| | - Nandhini Raman
- Gladstone Institute of Virology and Immunology, San Francisco, California, United States of America
| | - Ruth Greenblatt
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
- Departments of Clinical Pharmacy and Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Karen Smith-McCune
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
59
|
Winters AD, Romero R, Gervasi MT, Gomez-Lopez N, Tran MR, Garcia-Flores V, Pacora P, Jung E, Hassan SS, Hsu CD, Theis KR. Does the endometrial cavity have a molecular microbial signature? Sci Rep 2019; 9:9905. [PMID: 31289304 PMCID: PMC6616349 DOI: 10.1038/s41598-019-46173-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/21/2019] [Indexed: 01/15/2023] Open
Abstract
Recent molecular studies concluded that the endometrium has a resident microbiota dominated by Lactobacillus spp. and is therefore similar to that of the vagina. These findings were largely derived from endometrial samples obtained through a transcervical catheter and thus prone to contamination. Herein, we investigated the molecular microbial profiles of mid-endometrial samples obtained through hysterectomy and compared them with those of the cervix, vagina, rectum, oral cavity, and controls for background DNA contamination. Microbial profiles were examined through 16S rRNA gene qPCR and sequencing. Universal bacterial qPCR of total 16S rDNA revealed a bacterial load exceeding that of background DNA controls in the endometrium of 60% (15/25) of the study subjects. Bacterial profiles of the endometrium differed from those of the oral cavity, rectum, vagina, and background DNA controls, but not of the cervix. The bacterial profiles of the endometrium and cervix were dominated by Acinetobacter, Pseudomonas, Cloacibacterium, and Comamonadaceae. Both 16S rRNA gene sequencing and Lactobacillus species-specific (L. iners & L crispatus) qPCR showed that Lactobacillus was rare in the endometrium. In conclusion, if there is a microbiota in the middle endometrium, it is not dominated by Lactobacillus as was previously concluded, yet further investigation using culture and microscopy is necessary.
Collapse
Affiliation(s)
- Andrew D Winters
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA. .,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA. .,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA. .,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA. .,Detroit Medical Center, Detroit, Michigan, USA.
| | - Maria Teresa Gervasi
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Nardhy Gomez-Lopez
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Maria Rosa Tran
- Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sonia S Hassan
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, USA.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Kevin R Theis
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA. .,Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, USA. .,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, Detroit, Michigan, USA.
| |
Collapse
|
60
|
Schumacher A, Sharkey DJ, Robertson SA, Zenclussen AC. Immune Cells at the Fetomaternal Interface: How the Microenvironment Modulates Immune Cells To Foster Fetal Development. THE JOURNAL OF IMMUNOLOGY 2019; 201:325-334. [PMID: 29987001 DOI: 10.4049/jimmunol.1800058] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Immune cells adapt their phenotypic and functional characteristics in response to the tissue microenvironment within which they traffic and reside. The fetomaternal interface, consisting of placental trophoblasts and the maternal decidua, is a highly specialized tissue with a unique and time-limited function: to nourish and support development of the semiallogeneic fetus and protect it from inflammatory or immune-mediated injury. It is therefore important to understand how immune cells within these tissues are educated and adapt to fulfill their biological functions. This review article focuses on the local regulatory mechanisms ensuring that both innate and adaptive immune cells appropriately support the early events of implantation and placental development through direct involvement in promoting immune tolerance of fetal alloantigens, suppressing inflammation, and remodeling of maternal uterine vessels to facilitate optimal placental function and fetal growth.
Collapse
Affiliation(s)
- Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| |
Collapse
|
61
|
Buckner LR, Drobnis EZ, Augustine MS, Rogers LK, Akers J, Mott PD, Hope TJ, Quayle AJ, Schust DJ. Cervical and systemic concentrations of long acting hormonal contraceptive (LARC) progestins depend on delivery method: Implications for the study of HIV transmission. PLoS One 2019; 14:e0214152. [PMID: 31095572 PMCID: PMC6522049 DOI: 10.1371/journal.pone.0214152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/07/2019] [Indexed: 11/28/2022] Open
Abstract
Progestin-only long-acting reversible contraceptives (LARCs) are increasingly popular among women seeking contraception; however, recent epidemiological studies suggest that systemically administered medroxyprogesterone acetate (MPA) may increase HIV acquisition. In order to determine the exact mechanisms underlying increases in transmission specific to MPA use and to test safer, alternative contraceptive progestin types and delivery methods, in vitro modeling studies must be performed. To achieve this, it is imperative that accurate hormone concentrations be utilized when modeling progestin-mediated outcomes, as the down-stream effects are dose-dependent. The local concentrations of progestins to which the lower female genital tract tissues are exposed after initiation of LARCs are unknown, but they likely differ from peripheral concentrations, dependent upon the progestin type and delivery method. Here, we measured in vivo endocervical and plasma concentrations of (1) systemically-delivered depo MPA (DMPA), (2) levonorgestrel (LNG) delivered via intrauterine system (IUS) and (3) etonogestrel (ETG) delivered via vaginal ring in women who recently initiated contraception treatment. Levels of ETG and LNG in cervical secretions were 100-200 fold higher than plasma levels. In contrast, measurable MPA levels were approximately 10-fold higher in plasma compared to cervical secretions. These results will inform the design of accurate in vitro studies on the influence of progestins on epithelial cells, tissue explants, and peripheral blood cells, to be able to better predict in vivo outcomes. Subsequent observations will aid in determining how MPA might influence HIV acquisition and may facilitate identification of optimal progestin-containing LARC alternatives for women at high risk for HIV infection.
Collapse
Affiliation(s)
- Lyndsey R. Buckner
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Erma Z. Drobnis
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, United States of America
| | - Molly S. Augustine
- The Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, United States of America
| | - Lynette K. Rogers
- The Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, United States of America
| | - Jill Akers
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, United States of America
| | - Patricia D. Mott
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Thomas J. Hope
- Cell and Molecular Biology, Northwestern University, Chicago, IL, United States of America
| | - Alison J. Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Danny J. Schust
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, United States of America
| |
Collapse
|
62
|
Tietzel I, Quayle AJ, Carabeo RA. Alternatively Activated Macrophages Are Host Cells for Chlamydia trachomatis and Reverse Anti-chlamydial Classically Activated Macrophages. Front Microbiol 2019; 10:919. [PMID: 31134002 PMCID: PMC6524708 DOI: 10.3389/fmicb.2019.00919] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/11/2019] [Indexed: 12/12/2022] Open
Abstract
The obligate intracellular pathogen Chlamydia trachomatis (Ctr) is the causative agent of the most common form of sexually transmitted disease in the United States. Genital infections with C. trachomatis can lead to inflammatory tissue damage followed by scarring and tissue remodeling during wound healing. Extensive scarring can lead to ectopic pregnancy or infertility. Classically activated macrophages (CA mϕ), with their anti-microbial effector mechanisms, are known to be involved in acute inflammatory processes during the course of infection. In contrast, alternatively activated macrophages (AA mϕ) contribute to tissue repair at sites of wound healing, and have reduced bactericidal functions. They are present during infection, and thus potentially can provide a growth niche for C. trachomatis during a course of infection. To address this question, macrophages derived from CD14-positive monocytes magnetically isolated from peripheral blood mononuclear cells (PBMC) were treated with interferon-γ or interleukin-4 to produce CA mϕ or AA mϕ, respectively. Confocal microscopy of chlamydial inclusions and quantification of infectious yields revealed better pathogen growth and development in AA mϕ than CA mϕ, which correlated with the reduced expression of indoleamine 2,3-dioxygenase, a known anti-chlamydial effector of the host. Furthermore, AA mϕ stained strongly for transferrin receptor and secreted higher amounts of anti-inflammatory interleukin-10 compared to CA mϕ, characteristics that indicate its suitability as host to C. trachomatis. CA, AA, and resting mϕ were infected with Ctr serovar L2. The data suggest that IL-10 produced by infected AA mϕ attenuated the anti-chlamydial function of CA mϕ with growth recovery observed in infected CA mϕ in the presence of infected, but not mock-infected AA mϕ. This could be related to our observation that IL-10 treatment of infected CA mϕ promoted better chlamydial growth. Thus, in addition to serving as an additional niche, AA mϕ might also serve as a means to modulate the immediate environment by attenuating the anti-chlamydial functions of nearby CA mϕ in a manner that could involve IL-10 produced by infected AA mϕ.
Collapse
Affiliation(s)
- Illya Tietzel
- Department of Natural Sciences, Southern University at New Orleans, New Orleans, LA, United States
| | - Alison J Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rey A Carabeo
- School of Molecular Biosciences, Washington State University, Pullman, WA, United States
| |
Collapse
|
63
|
Yang X, Gilman-Sachs A, Kwak-Kim J. Ovarian and endometrial immunity during the ovarian cycle. J Reprod Immunol 2019; 133:7-14. [PMID: 31055226 DOI: 10.1016/j.jri.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
Immune tolerance is crucial for the successful pregnancy, while immune effectors and their products are required to safeguard a fetus from the infectious pathogens. The key immune effectors, such as T, B, and natural killer (NK) cells, monocytes, macrophages, and dendritic cells take part in regulating the immune responses at the maternal-fetal interface. The immune effectors become involved in intraovarian reproductive processes as well, such as ovulation, production of corpus luteum (CL) and its degeneration and determine the quality and evolution of the oocyte during the folliculogenesis. In the cycling endometrium, NK cells are rapidly infiltrated into the endometrium after ovulation and participate in angiogenesis and spiral artery remodeling process. In this study, we reviewed the characteristics and action mechanisms of immune effectors and their products in the peripheral blood, ovary, and endometrium during the ovarian cycle, since a comprehensive understanding of immune responses during the ovarian cycle and the time of implantation can help us to predict the pregnancy outcome and take effective measures for the prevention of potential obstetrical complications.
Collapse
Affiliation(s)
- Xiuhua Yang
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA; Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA; Department of Obstetrics, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA; Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA.
| |
Collapse
|
64
|
Barrios De Tomasi J, Opata MM, Mowa CN. Immunity in the Cervix: Interphase between Immune and Cervical Epithelial Cells. J Immunol Res 2019; 2019:7693183. [PMID: 31143785 PMCID: PMC6501150 DOI: 10.1155/2019/7693183] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The cervix is divided into two morphologically and immunologically distinct regions, namely, (1) the microbe-laden ectocervix, which is proximal to the vagina, and (2) the "sterile" endocervix, which is distal to the uterus. The two cervical regions are bordered by the cervical transformation zone (CTZ), an area of changing cells, and are predominantly composed of cervical epithelial cells. Epithelial cells are known to play a crucial role in the initiation, maintenance, and regulation of innate and adaptive response in collaboration with immune cells in several tissue types, including the cervix, and their dysfunction can lead to a spectrum of clinical syndromes. For instance, epithelial cells block progression and neutralize or kill microorganisms through multiple ways. These (ways) include mounting physical (intercellular junctions, secretion of mucus) and immune barriers (pathogen-recognition receptor-mediated pathways), which collectively and ultimately lead to the release of specific chemokines and or cytokines. The cytokines subsequently recruit subsets of immune cells appropriate to a particular immune context and response, such as dendritic cells (DCs), T, B, and natural killer (NK) cells. The immune response, as most biological processes in the female reproductive tract (FRT), is mainly regulated by estrogen and progesterone and their (immune cells) responses vary during different physiological phases of reproduction, such as menstrual cycle, pregnancy, and post menopause. The purpose of the present review is to compare the immunological profile of the mucosae and immune cells in the ecto- and endocervix and their interphase during the different phases of female reproduction.
Collapse
Affiliation(s)
- Jorgelina Barrios De Tomasi
- Department of Biology, Appalachian State University, Boone 28608, USA
- Departamento de Ciencias de la Medicina, Division de Ciencias de la Salud, Chetumal, Quintana Roo, Mexico
| | | | - Chishimba Nathan Mowa
- Department of Biology, Appalachian State University, Boone 28608, USA
- Rusangu University, Monze, Zambia
| |
Collapse
|
65
|
Abstract
As our understanding of mucosal immunity increases, it is becoming clear that the host response to HIV-1 is more complex and nuanced than originally believed. The mucosal landscape is populated with a variety of specialized cell types whose functions include combating infectious agents while preserving commensal microbiota, maintaining barrier integrity, and ensuring immune homeostasis. Advances in multiparameter flow cytometry, gene expression analysis and bioinformatics have allowed more detailed characterization of these cell types and their roles in host defense than was previously possible. This review provides an overview of existing literature on immunity to HIV-1 and SIVmac in mucosal tissues of the female reproductive tract and the gastrointestinal tract, focusing on major effector cell populations and briefly summarizing new information on tissue resident memory T cells, Treg, Th17, Th22 and innate lymphocytes (ILC), subsets that have been studied primarily in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology.,Division of Infectious Diseases, Department of Medicine, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
66
|
Abstract
Most new HIV infections, over 80%, occur through sexual transmission. During sexual transmission, the virus must bypass specific female and male reproductive tract anatomical barriers to encounter viable target cells. Understanding the generally efficient ability of these barrier to exclude HIV and the precise mechanisms of HIV translocation beyond these genital barriers is essential for vaccine and novel therapeutic development. In this review, we explore the mucosal, barriers of cervico-vaginal and penile tissues that comprise the female and male reproductive tracts. The unique cellular assemblies f the squamous and columnar epithelium are illustrate highlighting their structure and function. Each anatomical tissue offers a unique barrier to virus entry in healthy individuals. Unfortunately barrier dysfunction can lead to HIV transmission. How these diverse mucosal barriers have the potential to fail is considered, highlighting those anatomical areas that are postulated to offer a weaker barrier and are; therefore, more susceptible to viral ingress. Risk factors, such as sexually transmitted infections, microbiome dysbiosis, and high progestin environments are also associated with increased acquisition of HIV. How these states may affect the integrity of mucosal barriers leading to HIV acquisition are discussed suggesting mechanisms of transmission and revealing potential targets for intervention.
Collapse
Affiliation(s)
- Ann M Carias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| | - Thomas J Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| |
Collapse
|
67
|
Pepe G, Locati M, Della Torre S, Mornata F, Cignarella A, Maggi A, Vegeto E. The estrogen-macrophage interplay in the homeostasis of the female reproductive tract. Hum Reprod Update 2019; 24:652-672. [PMID: 30256960 DOI: 10.1093/humupd/dmy026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/10/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Estrogens are known to orchestrate reproductive events and to regulate the immune system during infections and following tissue damage. Recent findings suggest that, in the absence of any danger signal, estrogens trigger the physiological expansion and functional specialization of macrophages, which are immune cells that populate the female reproductive tract (FRT) and are increasingly being recognized to participate in tissue homeostasis beyond their immune activity against infections. Although estrogens are the only female gonadal hormones that directly target macrophages, a comprehensive view of this endocrine-immune communication and its involvement in the FRT is still missing. OBJECTIVE AND RATIONALE Recent accomplishments encourage a revision of the literature on the ability of macrophages to respond to estrogens and induce tissue-specific functions required for reproductive events, with the aim to envision macrophages as key players in FRT homeostasis and mediators of the regenerative and trophic actions of estrogens. SEARCH METHODS We conducted a systematic search using PubMed and Ovid for human, animal (rodents) and cellular studies published until 2018 on estrogen action in macrophages and the activity of these cells in the FRT. OUTCOMES Our search identified the remarkable ability of macrophages to activate biochemical processes in response to estrogens in cell culture experiments. The distribution at specific locations, interaction with selected cells and acquisition of distinct phenotypes of macrophages in the FRT, as well as the cyclic renewal of these properties at each ovarian cycle, demonstrate the involvement of these cells in the homeostasis of reproductive events. Moreover, current evidence suggests an association between estrogen-macrophage signaling and the generation of a tolerant and regenerative environment in the FRT, although a causative link is still missing. WIDER IMPLICATIONS Dysregulation of the functions and estrogen responsiveness of FRT macrophages may be involved in infertility and estrogen- and macrophage-dependent gynecological diseases, such as ovarian cancer and endometriosis. Thus, more research is needed on the physiology and pharmacological control of this endocrine-immune interplay.
Collapse
Affiliation(s)
- Giovanna Pepe
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Segrate, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via fratelli Cervi, Segrate, Italy
| | - Sara Della Torre
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Federica Mornata
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padua, Largo Meneghetti 2, Padua, Italy
| | - Adriana Maggi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| |
Collapse
|
68
|
The contraceptive medroxyprogesterone acetate, unlike norethisterone, directly increases R5 HIV-1 infection in human cervical explant tissue at physiologically relevant concentrations. Sci Rep 2019; 9:4334. [PMID: 30867477 PMCID: PMC6416361 DOI: 10.1038/s41598-019-40756-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/19/2019] [Indexed: 02/06/2023] Open
Abstract
The intramuscular progestin-only injectable contraceptive, depo-medroxyprogesterone acetate (DMPA-IM), is more widely used in Sub-Saharan Africa than another injectable contraceptive, norethisterone enanthate (NET-EN). Epidemiological data show a significant 1.4-fold increased risk of HIV-1 acquisition for DMPA-IM usage, while no such association is shown from limited data for NET-EN. We show that MPA, unlike NET, significantly increases R5-tropic but not X4-tropic HIV-1 replication ex vivo in human endocervical and ectocervical explant tissue from pre-menopausal donors, at physiologically relevant doses. Results support a mechanism whereby MPA, unlike NET, acts via the glucocorticoid receptor (GR) to increase HIV-1 replication in cervical tissue by increasing the relative frequency of CD4+ T cells and activated monocytes. We show that MPA, unlike NET, increases mRNA expression of the CD4 HIV-1 receptor and CCR5 but not CXCR4 chemokine receptors, via the GR. However, increased density of CD4 on CD3+ cells was not observed with MPA by flow cytometry of digested tissue. Results suggest that DMPA-IM may increase HIV-1 acquisition in vivo at least in part via direct effects on cervical tissue to increase founder R5-tropic HIV-1 replication. Our findings support differential biological mechanisms and disaggregation of DMPA-IM and NET-EN regarding HIV-1 acquisition risk category for use in high risk areas.
Collapse
|
69
|
Marlin R, Nugeyre MT, Tchitchek N, Parenti M, Lefebvre C, Hocini H, Benjelloun F, Cannou C, Nozza S, Dereuddre-Bosquet N, Levy Y, Barré-Sinoussi F, Scarlatti G, Le Grand R, Menu E. Seminal Plasma Exposures Strengthen Vaccine Responses in the Female Reproductive Tract Mucosae. Front Immunol 2019; 10:430. [PMID: 30915079 PMCID: PMC6423065 DOI: 10.3389/fimmu.2019.00430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/18/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-1 sexual transmission occurs mainly via mucosal semen exposures. In the female reproductive tract (FRT), seminal plasma (SP) induces physiological modifications, including inflammation. An effective HIV-1 vaccine should elicit mucosal immunity, however, modifications of vaccine responses by the local environment remain to be characterized. Using a modified vaccinia virus Ankara (MVA) as a vaccine model, we characterized the impact of HIV-1+ SP intravaginal exposure on the local immune responses of non-human primates. Multiple HIV-1+ SP exposures did not impact the anti-MVA antibody responses. However, SP exposures revealed an anti-MVA responses mediated by CD4+ T cells, which was not observed in the control group. Furthermore, the frequency and the quality of specific anti-MVA CD8+ T cell responses increased in the FRT exposed to SP. Multi-parameter approaches clearly identified the cervix as the most impacted compartment in the FRT. SP exposures induced a local cell recruitment of antigen presenting cells, especially CD11c+ cells, and CD8+ T cell recruitment in the FRT draining lymph nodes. CD11c+ cell recruitment was associated with upregulation of inflammation-related gene expression after SP exposures in the cervix. We thus highlight the fact that physiological conditions, such as SP exposures, should be taken into consideration to test and to improve vaccine efficacy against HIV-1 and other sexually transmitted infections.
Collapse
Affiliation(s)
- Romain Marlin
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France.,Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France
| | - Marie-Thérèse Nugeyre
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France.,Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France
| | - Nicolas Tchitchek
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France
| | - Matteo Parenti
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France
| | - Cécile Lefebvre
- Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France.,Équipe 16 Physiopathologie et Immunothérapies dans l'Infection VIH, Institut Mondor de Recherche Biomédicale - INSERM U955, Créteil, France
| | - Hakim Hocini
- Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France.,Équipe 16 Physiopathologie et Immunothérapies dans l'Infection VIH, Institut Mondor de Recherche Biomédicale - INSERM U955, Créteil, France
| | - Fahd Benjelloun
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Claude Cannou
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Silvia Nozza
- Infectious Diseases Department, San Raffaele Scientific Institute, Milan, Italy
| | - Nathalie Dereuddre-Bosquet
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France
| | - Yves Levy
- Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France.,Équipe 16 Physiopathologie et Immunothérapies dans l'Infection VIH, Institut Mondor de Recherche Biomédicale - INSERM U955, Créteil, France.,Groupe Henri-Mondor Albert-Chenevier, Service d'Immunologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France
| | - Françoise Barré-Sinoussi
- Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France.,International Division, Institut Pasteur, Paris, France
| | - Gabriella Scarlatti
- Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France.,Viral Evolution and Transmission Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Roger Le Grand
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France
| | - Elisabeth Menu
- IDMIT Department, U1184 ≪ Immunology of Viral Infections and Autoimmune Diseases ≫ (IMVA), CEA, IBFJ, Université Paris-Sud, Inserm, Fontenay-Aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France.,Vaccine Research Institute - VRI, Hôpital Henri Mondor, Créteil, France
| |
Collapse
|
70
|
Li L, Zhou J, Wang W, Huang L, Tu J, Baiamonte L, Stark M, Mills M, Hope TJ, Drobnis EZ, Quayle AJ, Schust DJ. Effects of three long-acting reversible contraceptive methods on HIV target cells in the human uterine cervix and peripheral blood. Reprod Biol Endocrinol 2019; 17:26. [PMID: 30795774 PMCID: PMC6387540 DOI: 10.1186/s12958-019-0469-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/12/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hormonal contraceptives, particularly depot medroxyprogesterone acetate (DMPA), have been reported to be associated with substantially enhanced HIV acquisition; however, the biological mechanisms of this risk remain poorly understood. We aimed to investigate the effects of different hormonal contraceptives on the expression of the HIV co-receptors, CXCR4 and CCR5, on female endocervical and peripheral blood T cells. METHODS A total of 59 HIV-negative women were enrolled, including 15 initiating DMPA, 28 initiating a levonorgestrel-releasing intrauterine device (LNG-IUD) and 16 initiating an etonogestrel (ETG)-delivering vaginal ring. Peripheral blood and endocervical cytobrush specimens were collected at enrollment and 3-4 weeks after contraception initiation to analyze the expression of CXCR4 and CCR5, on CD4+ and CD8+ T cells using flow cytometry. RESULTS Administration of DMPA increased the percentages of CD4+ and CD8+ T cells expressing CCR5 in the endocervix but not in the peripheral blood. Administration of the LNG-IUD or the ETG vaginal ring did not affect the percentages of T lymphocytes expressing CXCR4 or CCR5 in the female cervix or peripheral blood. CONCLUSIONS Increase in the percentage of endocervical T cells expressing CCR5 upon DMPA exposure provides a plausible biological explanation for the association between DMPA use and an elevated risk of HIV infection.
Collapse
Affiliation(s)
- Liping Li
- Department of Obstetrics and Gynecology, Guangzhou First People’s Hospital, South China University of Technology School of Medicine, Guangzhou, China
| | - Jie Zhou
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO USA
| | - Weijia Wang
- Department of Obstetrics and Gynecology, Guangzhou First People’s Hospital, South China University of Technology School of Medicine, Guangzhou, China
| | - Lina Huang
- Department of Obstetrics and Gynecology, Guangzhou First People’s Hospital, South China University of Technology School of Medicine, Guangzhou, China
| | - Jiaoqin Tu
- Department of Obstetrics and Gynecology, Guangzhou First People’s Hospital, South China University of Technology School of Medicine, Guangzhou, China
| | - Lyndsey Baiamonte
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana USA
| | - Moselle Stark
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO USA
| | - Mistie Mills
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO USA
| | - Thomas J. Hope
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Erma Z. Drobnis
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO USA
| | - Alison J. Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana USA
| | - Danny J. Schust
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO USA
| |
Collapse
|
71
|
Erneholm K, Lorenzen E, Bøje S, Olsen AW, Jungersen G, Jensen HE, Cassidy JP, Andersen P, Agerholm JS, Follmann F. Genital Infiltrations of CD4 + and CD8 + T Lymphocytes, IgA + and IgG + Plasma Cells and Intra-Mucosal Lymphoid Follicles Associate With Protection Against Genital Chlamydia trachomatis Infection in Minipigs Intramuscularly Immunized With UV-Inactivated Bacteria Adjuvanted With CAF01. Front Microbiol 2019; 10:197. [PMID: 30800114 PMCID: PMC6375829 DOI: 10.3389/fmicb.2019.00197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/24/2019] [Indexed: 11/15/2022] Open
Abstract
The development of a vaccine against genital chlamydia in women is advancing, and the evaluation of in situ immune responses following vaccination and challenge infections is crucial for development of a safe and protective vaccine. This study employs the sexually mature minipig model to characterize the genital in situ immune response to Chlamydia trachomatis infection in pigs previously immunized intramuscularly with UV-inactivated C. trachomatis serovar D (UV-SvD) adjuvanted/formulated with CAF01 adjuvant compared to a CAF01-alone control group. Pigs immunized with UV-SvD were significantly protected against vaginal challenge with C. trachomatis on day 3 post inoculation and showed significantly higher cervical infiltrations of approximately equal numbers of CD4+ and CD8+ T-cells, and IgG+ and IgA+ plasma cells compared to adjuvant-alone immunized controls. These immunological signatures correspond to findings in mice and are similar to those described in female chlamydia patients. This proves important potential for the pig model in elucidating immunological in situ signatures in future translational research in chlamydia vaccinology.
Collapse
Affiliation(s)
- Karin Erneholm
- Section of Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Emma Lorenzen
- Section of Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Sarah Bøje
- Section of Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Weinreich Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Henrik E. Jensen
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Joseph P. Cassidy
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Jørgen S. Agerholm
- Section of Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
72
|
Gonzalez SM, Aguilar-Jimenez W, Su RC, Rugeles MT. Mucosa: Key Interactions Determining Sexual Transmission of the HIV Infection. Front Immunol 2019; 10:144. [PMID: 30787929 PMCID: PMC6373783 DOI: 10.3389/fimmu.2019.00144] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 01/17/2019] [Indexed: 12/26/2022] Open
Abstract
In the context of HIV sexual transmission at the genital mucosa, initial interactions between the virus and the mucosal immunity determine the outcome of the exposure. Hence, these interactions have been deeply explored in attempts to undercover potential targets for developing preventative strategies. The knowledge gained has led to propose a hypothetical model for mucosal HIV transmission. Subsequent research studies on this topic further revealed new mechanisms and identified new host-HIV interactions. This review aims at integrating these findings to inform better and update the current model of HIV transmission. At the earliest stage of virus exposure, the epithelial integrity and the presence of antiviral factors are critical in preventing viral entry to the submucosa. However, the virus has been shown to enter to the submucosa in the presence of physical abrasion or via epithelial transmigration using paracellular passage or transcytosis mechanisms. The efficiency of these processes is greater with cell-associated viral inoculums and can be influenced by the presence of viral and immune factors, and by the structure of the exposed epithelium. Once the virus reaches the submucosa, dendritic cells and fibroblasts, as recently described, have been shown in vitro of being capable of facilitating the transfer of viral particles to susceptible cells, leading to viral dissemination, most likely in a trans-infection manner. The presence of activated CD4+ T cells in submucosa increases the probability of infection, where the predominant microbiota could be implicated through the modulation of an inflammatory microenvironment. Other factors such as genital fluids and hormones could also play an essential role in HIV transmission. Here, we review the most recent evidence described for mucosal HIV-transmission contributing with the understanding of this phenomenon.
Collapse
Affiliation(s)
- Sandra M Gonzalez
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,National HIV and Retrovirology Laboratory, JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada
| | | | - Ruey-Chyi Su
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Maria T Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
73
|
|
74
|
Introini A, Vanpouille C, Fitzgerald W, Broliden K, Margolis L. Ex Vivo Infection of Human Lymphoid Tissue and Female Genital Mucosa with Human Immunodeficiency Virus 1 and Histoculture. J Vis Exp 2018. [PMID: 30371673 DOI: 10.3791/57013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Histocultures allow studying intercellular interactions within human tissues, and they can be employed to model host-pathogen interactions under controlled laboratory conditions. Ex vivo infection of human tissues with human immunodeficiency virus (HIV), among other viruses, has been successfully used to investigate early disease pathogenesis, as well as a platform to test the efficacy and toxicity of antiviral drugs. In the present protocol, we explain how to process and infect with HIV-1 tissue explants from human tonsils and cervical mucosae, and maintain them in culture on top of gelatin sponges at the liquid-air interface for about two weeks. This non-polarized culture setting maximizes access to nutrients in culture medium and oxygen, although progressive loss of tissue integrity and functional architectures remains its main limitation. This method allows monitoring HIV-1 replication and pathogenesis using several techniques, including immunoassays, qPCR, and flow cytometry. Of importance, the physiologic variability between tissue donors, as well as between explants from different areas of the same specimen, may significantly affect experimental results. To ensure result reproducibility, it is critical to use an adequate number of explants, technical replicates, and donor-matched control conditions to normalize the results of the experimental treatments when compiling data from multiple experiments (i.e., conducted using tissue from different donors) for statistical analysis.
Collapse
Affiliation(s)
- Andrea Introini
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet; Section of Intercellular Interactions, Eunice Shriver National Institute of Child Health and Human Development, National Institutes of Health;
| | - Christophe Vanpouille
- Section of Intercellular Interactions, Eunice Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Wendy Fitzgerald
- Section of Intercellular Interactions, Eunice Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Kristina Broliden
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet
| | - Leonid Margolis
- Section of Intercellular Interactions, Eunice Shriver National Institute of Child Health and Human Development, National Institutes of Health
| |
Collapse
|
75
|
Melis GB, Piras B, Marotto MF, Neri M, Corda V, Vallerino V, Saba A, Lello S, Pilloni M, Zedda P, Paoletti AM, Mais V. The stimulation of the vaginal immune system with short-term administration of a vaginal gel containing fraction of Propionibacterium acnes, hyaluronic acid and polycarbophil is efficacious in vaginal infections dependent on disorders in the vaginal ecosystem. Gynecol Endocrinol 2018; 34:880-883. [PMID: 29648469 DOI: 10.1080/09513590.2018.1460345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/26/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022] Open
Abstract
The vaginal immune system (VIS) is the first defense against antigens recognized as foreign. Substances capable of locally activating the VIS could be a valid strategy to treat vulvo-vaginal infections (VVI), caused by changes in the vaginal ecosystem, such as bacterial vaginosis (BV), vulvo-vaginal candidiasis (CA), and mixed vaginitis (MV). Bacterial lysates, obtained by crushing bacterial cultures, exert immuno-modulatory activities. The parietal fraction from Propionibacterium acnes is a patent of Depofarma (MoglianoVeneto, Italy). The preparation that associates such fraction to hyaluronic acid and polycarbophil is a registered trademark, commercially available in Italy as vaginal gel, Immunovag®. The study aimed to evaluate whether a 5-day-treatment with Immunovag® improves the symptoms and signs of VVI, in 60 women with Gardnerella vaginalis (GV), 154 with CA, 95 with MV, diagnosed with vulvar vaginal swab (VVS), and in 283 with BV, diagnosed with the Amsel criteria. At the end of the treatment (visit 2), the symptoms and signs of VVI disappeared in a significant number of subjects (χ2p < .02 vs pre-treatment) in all VVI groups, and their intensity was significantly (p < .0002) reduced in the subjects in which they were still present. Immunovag® represents a valid treatment of VVI induced by changes in the vaginal ecosystem.
Collapse
Affiliation(s)
- Gian Benedetto Melis
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Bruno Piras
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Maria Francesca Marotto
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Manuela Neri
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Valentina Corda
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Valerio Vallerino
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Alessandra Saba
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Stefano Lello
- c Department of Health Protection for Women and Children , Gemelli Polyclinic Foundation, Rome , Italy
| | - Monica Pilloni
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Pierina Zedda
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Anna Maria Paoletti
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| | - Valerio Mais
- a Department of Surgical Sciences , University of Cagliari , Cagliari , CA , Italy
- b Department of Obstetrics and Gynecology , University Hospital of Cagliari (AOUCA) , Cagliari , CA , Italy
| |
Collapse
|
76
|
Santegoets SJ, van Ham VJ, Ehsan I, Charoentong P, Duurland CL, van Unen V, Höllt T, van der Velden LA, van Egmond SL, Kortekaas KE, de Vos van Steenwijk PJ, van Poelgeest MIE, Welters MJP, van der Burg SH. The Anatomical Location Shapes the Immune Infiltrate in Tumors of Same Etiology and Affects Survival. Clin Cancer Res 2018; 25:240-252. [PMID: 30224343 DOI: 10.1158/1078-0432.ccr-18-1749] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/27/2018] [Accepted: 09/10/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The tumor immune microenvironment determines clinical outcome. Whether the original tissue in which a primary tumor develops influences this microenvironment is not well understood. EXPERIMENTAL DESIGN We applied high-dimensional single-cell mass cytometry [Cytometry by Time-Of-Flight (CyTOF)] analysis and functional studies to analyze immune cell populations in human papillomavirus (HPV)-induced primary tumors of the cervix (cervical carcinoma) and oropharynx (oropharyngeal squamous cell carcinoma, OPSCC). RESULTS Despite the same etiology of these tumors, the composition and functionality of their lymphocytic infiltrate substantially differed. Cervical carcinoma displayed a 3-fold lower CD4:CD8 ratio and contained more activated CD8+CD103+CD161+ effector T cells and less CD4+CD161+ effector memory T cells than OPSCC. CD161+ effector cells produced the highest cytokine levels among tumor-specific T cells. Differences in CD4+ T-cell infiltration between cervical carcinoma and OPSCC were reflected in the detection rate of intratumoral HPV-specific CD4+ T cells and in their impact on OPSCC and cervical carcinoma survival. The peripheral blood mononuclear cell composition of these patients, however, was similar. CONCLUSIONS The tissue of origin significantly affects the overall shape of the immune infiltrate in primary tumors.
Collapse
Affiliation(s)
- Saskia J Santegoets
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vanessa J van Ham
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ilina Ehsan
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pornpimol Charoentong
- Department of Medical Oncology, National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Chantal L Duurland
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas Höllt
- Department of Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands.,Computer Graphics and Visualization Group, Delft University of Technology, Delft, the Netherlands
| | - Lilly-Ann van der Velden
- Department of Otorhinolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Sylvia L van Egmond
- Department of Otorhinolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim E Kortekaas
- Department of Gynaecology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
77
|
Increased degranulation of immune cells is associated with higher cervical viral load in HIV-infected women. AIDS 2018; 32:1939-1949. [PMID: 29912065 DOI: 10.1097/qad.0000000000001925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The activation of effector immune cells at the cervicovaginal mucosa (CVM) might influence the cervical HIV load and thus the secondary transmission; however, limited information is available about the innate effector cells at CVM during HIV infection. In this study, we quantified and assessed the activation of the effector immune cells at the CVM of HIV-infected women with different disease outcomes: nonprogressive HIV disease (LTNPs) and chronic HIV-infected (CHI) and their relationship with cervical viral shedding. METHOD The phenotype and frequency of cytobrush-derived effector immune cells like natural killer cells, T cells, and dendritic cells and their degranulation status (CD107a expression as a surrogate marker of activation) was determined using flow cytometry in age-matched HIV- infected and uninfected women and their association with cervical HIV load was determined. RESULT The frequencies of dendritic cells, CD56, CD56 natural killer cell subsets were similar in both the study groups and also within the HIV-infected women with and without progressive disease. The frequencies of CD56CD16 natural killer cells (P = 0.04) and degranulating CD56 natural killer cells were significantly higher among HIV-infected women (P < 0.05). Among HIV-infected women, LTNP women showed reduced degranulation of natural killer and CD8 T cells than seen in the CHI women, which was also associated with lower cervical viral load (P < 0.05). CONCLUSION The present study showed that increased degranulation of natural killer and T cells is associated with higher HIV shedding at the CVM of HIV-infected women. Hence reduction of the local immune activation at CVM could be an effective strategy to reduce the cervical viral load.
Collapse
|
78
|
Wessels JM, Felker AM, Dupont HA, Kaushic C. The relationship between sex hormones, the vaginal microbiome and immunity in HIV-1 susceptibility in women. Dis Model Mech 2018; 11:dmm035147. [PMID: 30154116 PMCID: PMC6177003 DOI: 10.1242/dmm.035147] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The role of sex hormones in regulating immune responses in the female genital tract has been recognized for decades. More recently, it has become increasingly clear that sex hormones regulate susceptibility to sexually transmitted infections through direct and indirect mechanisms involving inflammation and immune responses. The reproductive cycle can influence simian/human immunodeficiency virus (SHIV) infections in primates and HIV-1 infection in ex vivo cervical tissues from women. Exogenous hormones, such as those found in hormonal contraceptives, have come under intense scrutiny because of the increased susceptibility to sexually transmitted infections seen in women using medroxyprogesterone acetate, a synthetic progestin-based contraceptive. Recent meta-analyses concluded that medroxyprogesterone acetate enhanced HIV-1 susceptibility in women by 40%. In contrast, estradiol-containing hormonal contraceptives were not associated with increased susceptibility and some studies reported a protective effect of estrogen on HIV/SIV infection, although the underlying mechanisms remain incompletely understood. Recent studies describe a key role for the vaginal microbiota in determining susceptibility to sexually transmitted infections, including HIV-1. While Lactobacillus spp.-dominated vaginal microbiota is associated with decreased susceptibility, complex microbiota, such as those seen in bacterial vaginosis, correlates with increased susceptibility to HIV-1. Interestingly, sex hormones are inherently linked to microbiota regulation in the vaginal tract. Estrogen has been postulated to play a key role in establishing a Lactobacillus-dominated microenvironment, whereas medroxyprogesterone acetate is linked to hypo-estrogenic effects. The aim of this Review is to contribute to a better understanding of the sex-hormone-microbiome-immunity axis, which can provide key information on the determinants of HIV-1 susceptibility in the female genital tract and, consequently, inform HIV-1 prevention strategies.
Collapse
Affiliation(s)
- Jocelyn M Wessels
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Allison M Felker
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Haley A Dupont
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
79
|
Endocervical and vaginal microbiota in South African adolescents with asymptomatic Chlamydia trachomatis infection. Sci Rep 2018; 8:11109. [PMID: 30038262 PMCID: PMC6056523 DOI: 10.1038/s41598-018-29320-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/06/2018] [Indexed: 12/12/2022] Open
Abstract
Adolescent girls and young women represent a key risk group for sexually transmitted infections (STIs). The vaginal microbiota is thought to play an important role in susceptibility to STIs such as Chlamydia trachomatis. We compared the microbiota of the lateral vaginal wall and endocervix, and assessed associations with C. trachomatis infection in South African adolescents. The endocervical and vaginal lateral wall microbiota were characterized by amplifying and sequencing the V4 region of the 16S rRNA gene and C. trachomatis diagnosed using molecular methods. Of the 72 girls included, 30 had asymptomatic C. trachomatis infections. Three major vaginal community types were identified; one Lactobacillus crispatus, one L. iners and one diverse, Gardnerella vaginalis dominant. The microbiota of the endocervix was significantly different from that of the lateral wall in terms of diversity. There were many differentially abundant taxa between the endocervix and lateral vaginal wall, including Achromobacter spanius and Enterococcus faecium. Women with C. trachomatis had higher relative abundance of G. vaginalis and other anaerobes. In this African adolescent cohort, significant differences between the lateral vaginal wall and endocervical microbiota diversity and composition were evident, although neither were strongly associated with C. trachomatis infection.
Collapse
|
80
|
Salinas-Muñoz L, Campos-Fernández R, Mercader E, Olivera-Valle I, Fernández-Pacheco C, Matilla L, García-Bordas J, Brazil JC, Parkos CA, Asensio F, Muñoz-Fernández MA, Hidalgo A, Sánchez-Mateos P, Samaniego R, Relloso M. Estrogen Receptor-Alpha (ESR1) Governs the Lower Female Reproductive Tract Vulnerability to Candida albicans. Front Immunol 2018; 9:1033. [PMID: 29881378 PMCID: PMC5976782 DOI: 10.3389/fimmu.2018.01033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/25/2018] [Indexed: 01/04/2023] Open
Abstract
Estradiol-based therapies predispose women to vaginal infections. Moreover, it has long been known that neutrophils are absent from the vaginal lumen during the ovulatory phase (high estradiol). However, the mechanisms that regulate neutrophil influx to the vagina remain unknown. We investigated the neutrophil transepithelial migration (TEM) into the vaginal lumen. We revealed that estradiol reduces the CD44 and CD47 epithelial expression in the vaginal ectocervix and fornix, which retain neutrophils at the apical epithelium through the estradiol receptor-alpha. In contrast, luteal progesterone increases epithelial expression of CD44 and CD47 to promote neutrophil migration into the vaginal lumen and Candida albicans destruction. Distinctive to vaginal mucosa, neutrophil infiltration is contingent to sex hormones to prevent sperm from neutrophil attack; although it may compromise immunity during ovulation. Thus, sex hormones orchestrate tolerance and immunity in the vaginal lumen by regulating neutrophil TEM.
Collapse
Affiliation(s)
- Laura Salinas-Muñoz
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Raúl Campos-Fernández
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Enrique Mercader
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Servicio de Cirugía General, Sección Cirugía Endocrino-Metabólica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Irene Olivera-Valle
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Carlota Fernández-Pacheco
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Animalario, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Lara Matilla
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Julio García-Bordas
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Fernando Asensio
- Animalario, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maria A Muñoz-Fernández
- Laboratorio InmunoBiología Molecular, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Paloma Sánchez-Mateos
- Laboratorio de Inmuno-oncología, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Rafael Samaniego
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Miguel Relloso
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
81
|
Ellegård R, Khalid M, Svanberg C, Holgersson H, Thorén Y, Wittgren MK, Hinkula J, Nyström S, Shankar EM, Larsson M. Complement-Opsonized HIV-1 Alters Cross Talk Between Dendritic Cells and Natural Killer (NK) Cells to Inhibit NK Killing and to Upregulate PD-1, CXCR3, and CCR4 on T Cells. Front Immunol 2018; 9:899. [PMID: 29760706 PMCID: PMC5936988 DOI: 10.3389/fimmu.2018.00899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/11/2018] [Indexed: 01/07/2023] Open
Abstract
Dendritic cells (DCs), natural killer (NK) cells, and T cells play critical roles during primary HIV-1 exposure at the mucosa, where the viral particles become coated with complement fragments and mucosa-associated antibodies. The microenvironment together with subsequent interactions between these cells and HIV at the mucosal site of infection will determine the quality of immune response that ensues adaptive activation. Here, we investigated how complement and immunoglobulin opsonization influences the responses triggered in DCs and NK cells, how this affects their cross talk, and what T cell phenotypes are induced to expand following the interaction. Our results showed that DCs exposed to complement-opsonized HIV (C-HIV) were less mature and had a poor ability to trigger IFN-driven NK cell activation. In addition, when the DCs were exposed to C-HIV, the cytotolytic potentials of both NK cells and CD8 T cells were markedly suppressed. The expression of PD-1 as well as co-expression of negative immune checkpoints TIM-3 and LAG-3 on PD-1 positive cells were increased on both CD4 as well as CD8 T cells upon interaction with and priming by NK–DC cross talk cultures exposed to C-HIV. In addition, stimulation by NK–DC cross talk cultures exposed to C-HIV led to the upregulation of CD38, CXCR3, and CCR4 on T cells. Together, the immune modulation induced during the presence of complement on viral surfaces is likely to favor HIV establishment, dissemination, and viral pathogenesis.
Collapse
Affiliation(s)
- Rada Ellegård
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mohammad Khalid
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Pharmaceutics, College of Pharmacy, King Khalid University, Asir-Abha, Saudi Arabia
| | - Cecilia Svanberg
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Hanna Holgersson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ylva Thorén
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mirja Karolina Wittgren
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Clinical Immunology and Transfusion Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Esaki M Shankar
- Division of Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India.,Center of Excellence for Research in AIDS (CERiA), University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia.,Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| | - Marie Larsson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
82
|
Gao Y, McKay PF, Mann JFS. Advances in HIV-1 Vaccine Development. Viruses 2018; 10:E167. [PMID: 29614779 PMCID: PMC5923461 DOI: 10.3390/v10040167] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 03/30/2018] [Accepted: 03/30/2018] [Indexed: 02/07/2023] Open
Abstract
An efficacious HIV-1 vaccine is regarded as the best way to halt the ongoing HIV-1 epidemic. However, despite significant efforts to develop a safe and effective vaccine, the modestly protective RV144 trial remains the only efficacy trial to provide some level of protection against HIV-1 acquisition. This review will outline the history of HIV vaccine development, novel technologies being applied to HIV vaccinology and immunogen design, as well as the studies that are ongoing to advance our understanding of vaccine-induced immune correlates of protection.
Collapse
Affiliation(s)
- Yong Gao
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, N6A 5C1, Canada.
| | - Paul F McKay
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London, W2 1PG, UK.
| | - Jamie F S Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
83
|
Dezzutti CS, Park SY, Marks KM, Lawlor SE, Russo JR, Macio I, Chappell CA, Bunge KE. Heterogeneity of HIV-1 Replication in Ectocervical and Vaginal Tissue Ex Vivo. AIDS Res Hum Retroviruses 2018; 34:185-192. [PMID: 28982249 DOI: 10.1089/aid.2017.0107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In clinical trials evaluating HIV-1 prevention products, ex vivo exposure of mucosal tissue to HIV-1 is performed to inform drug levels needed to suppress viral infection. Understanding assay and participant variables that influence HIV-1 replication will help with assay implementation. Demographic and behavioral data were obtained from 61 healthy women aged 21-45. Paired cervical tissue (CT) and vaginal tissue (VT) biopsies were collected and treated with HIV-1BaL or HIV-1JR-CSF, washed, and cultured. On days 3, 7, and/or 11, culture supernatant was collected, and viral replication was monitored by p24 ELISA. Tissue was extracted at study end, and HIV-1 relative RNA copies were determined by polymerase chain reaction. Cumulative p24 and RNA were log-transformed and analyzed using a linear mixed model, t-test, and an intraclass correlation coefficient (ICC). HIV replication was similar between CT and VT for each virus, but HIV-1BaL had 1.5 log10 and 0.9 log10 higher levels of p24 than HIV-1JR-CSF in CT and VT, respectively (p < .001), which correlated with HIV-1 relative RNA copies. Cumulative p24 and RNA copies in both tissues demonstrated low intraperson correlation for both viruses (ICC ≤0.513 HIV-1BaL; ICC ≤0.419 HIV-1JR-CSF). Enrollment into previous clinical studies in which genital biopsies were collected modestly decreased the HIV-1BaL cumulative p24 for CT, but not for VT. To improve the ex vivo challenge assay, viruses should be evaluated for replication in mucosal tissue before study implementation, baseline mucosal tissue is not needed if a placebo/no treatment group is included within the clinical trial, and previous biopsy sites should be avoided.
Collapse
Affiliation(s)
- Charlene S. Dezzutti
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Seo Young Park
- Department of Medicine Bio Statisics, and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | - Julie R. Russo
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Ingrid Macio
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Catherine A. Chappell
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Katherine E. Bunge
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| |
Collapse
|
84
|
Akbalik ME, Liman N, Sagsoz H, Guney Saruhan B. Tissue distribution of some immune cells in bovine reproductive tract during follicular and luteal phase. Microsc Res Tech 2018; 81:315-331. [PMID: 29318745 DOI: 10.1002/jemt.22983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/08/2017] [Accepted: 12/16/2017] [Indexed: 01/21/2023]
Abstract
More recent studies indicate that immune cells which secrete their secretory products or cytokines play an important role in reproductive system. In our study, immune cell populations (CD8+ T lymphocytes, CD68+ macrophages, plasma cells, siderophages, eosinophils) and expression of major histocompatibility complex (MHC) class I and class II were examined in female reproductive tract during follicular (n = 13) and luteal phase (n = 10). Plasma cells and eosinophil granulocytes are present in few numbers in luminal epithelium, but abundant in longitudinal muscle layer of uterus, whereas siderophages are the dominant cell type in stroma. Moreover, MHC-I and -II+ cells are expressed by individual cells in organ layers, while CD8+ T cells and CD68+ macrophages are dominant in epithelium and muscle layer, respectively. In conclusion, we did not found significant changes in immune cells according to follicular and luteal phases, but localization and numbers in each organ have changed according to both organ and layers. These results indicate that these factors may play a crucial role not only to generate an immune response but also to have a role in regulation of physiological functions in female reproductive organs.
Collapse
Affiliation(s)
- Mehmet Erdem Akbalik
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakir, Turkey
| | - Narin Liman
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Hakan Sagsoz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakir, Turkey
| | - Berna Guney Saruhan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
85
|
In vitro models for deciphering the mechanisms underlying the sexual transmission of viruses at the mucosal level. Virology 2017; 515:1-10. [PMID: 29220713 DOI: 10.1016/j.virol.2017.11.023] [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] [Received: 10/02/2017] [Revised: 11/13/2017] [Accepted: 11/28/2017] [Indexed: 01/31/2023]
Abstract
Sexually transmitted viruses infect the genital and colorectal mucosa of the partner exposed to contaminated genital secretions through a wide range of mechanisms, dictated in part by the organization of the mucosa. Because understanding the modes of entry into the organism of viruses transmitted through sexual intercourse is a necessary prerequisite to the design of treatments to block those infections, in vitro modeling of the transmission is essential. The aim of this review is to present the models and methodologies available for the in vitro study of the interactions between viruses and mucosal tissue and for the preclinical evaluation of antiviral compounds, and to point out their advantages and limitations according to the question being studied.
Collapse
|
86
|
|
87
|
The human female urogenital microbiome: complexity in normality. Emerg Top Life Sci 2017; 1:363-372. [PMID: 33525775 DOI: 10.1042/etls20170042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 09/13/2017] [Accepted: 09/29/2017] [Indexed: 12/19/2022]
Abstract
Microbial communities of the urogenital tract have long been recognised to play an important role in disease states. A revolution in methodological approaches is permitting the assessment of complex urogenital tract microbiota-host interactions and the metabolic and protein milieu of the mucosal interface. There is now great potential for significant advances in biomarker discovery and disease risk stratification, and for the elucidation of mechanisms underpinning the microbial community dynamics involved in urogenital tract pathology. Microbiota-host interactions in the female genital tract have a particular significance, because unlike in the male, there is direct communication between the external genitalia, the uterus and the peritoneal cavity. This review examines the microbial community composition at differing sites of the female urogenital tract and its relationship with health and disease. Key factors involved in the modulation of vaginal microbiome stability and structure, such as endocrine, immune and inflammatory pathways, are considered in the context of a woman's life cycle and disease pathogenesis.
Collapse
|
88
|
Vitali D, Wessels JM, Kaushic C. Role of sex hormones and the vaginal microbiome in susceptibility and mucosal immunity to HIV-1 in the female genital tract. AIDS Res Ther 2017; 14:39. [PMID: 28893284 PMCID: PMC5594427 DOI: 10.1186/s12981-017-0169-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
While the prevalence of Human immunodeficiency virus-1 (HIV-1) infection has stabilized globally, it continues to be the leading cause of death among women of reproductive age. The majority of new infections are transmitted heterosexually, and women have consistently been found to be more susceptible to HIV-1 infection during heterosexual intercourse compared to men. This emphasizes the need for a deeper understanding of how the microenvironment in the female genital tract (FGT) could influence HIV-1 acquisition. This short review focuses on our current understanding of the interplay between estrogen, progesterone, and the cervicovaginal microbiome and their immunomodulatory effects on the FGT. The role of hormonal contraceptives and bacterial vaginosis on tissue inflammation, T cell immunity and HIV-1 susceptibility is discussed. Taken together, this review provides valuable information for the future development of multi-purpose interventions to prevent HIV-1 infection in women.
Collapse
|
89
|
Effects of depot-medroxyprogesterone acetate on the immune microenvironment of the human cervix and endometrium: implications for HIV susceptibility. Mucosal Immunol 2017; 10:1270-1278. [PMID: 28051087 PMCID: PMC5496803 DOI: 10.1038/mi.2016.121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 11/21/2016] [Indexed: 02/04/2023]
Abstract
Depot-medroxyprogesterone acetate is a commonly used injectable contraceptive that has been associated with an increased risk of HIV acquisition. This study compares effects of depot-medroxyprogesterone acetate on immune parameters from several upper reproductive tract compartments relevant to HIV-1 susceptibility in repetitive samples from 15 depot-medroxyprogesterone acetate users and 27 women not on hormonal contraceptives. Compared with samples from unexposed women in the mid-luteal phase, depot-medroxyprogesterone acetate use was associated with: increased endocervical concentrations of MCP1 and IFNalpha2; decreased endocervical concentrations of IL1beta and IL6; increased proportions of endometrial CD4+ and CD8+ cells expressing the activation marker HLADR; increased density of endometrial macrophages; and decreased density of endometrial regulatory T cells. Unlike previous reports with samples from the vagina, we did not observe increased expression of the HIV co-receptor CCR5 on CD4+ T cells in the endocervix or endometrium. Our results indicate important differences in anatomic compartments regarding mechanisms by which depot-medroxyprogesterone acetate could be associated with increased risk of HIV acquisition, including increased recruitment of macrophages to the endometrium, decreased levels of pro-inflammatory cytokines in the endocervix possibly leading to enhanced susceptibility to viral infection, and activation of endometrial T cells.
Collapse
|
90
|
Marlin R, Nugeyre MT, Tchitchek N, Parenti M, Hocini H, Benjelloun F, Cannou C, Dereuddre-Bosquet N, Levy Y, Barré-Sinoussi F, Scarlatti G, Le Grand R, Menu E. Modified Vaccinia Virus Ankara Vector Induces Specific Cellular and Humoral Responses in the Female Reproductive Tract, the Main HIV Portal of Entry. THE JOURNAL OF IMMUNOLOGY 2017; 199:1923-1932. [PMID: 28760882 DOI: 10.4049/jimmunol.1700320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/03/2017] [Indexed: 11/19/2022]
Abstract
The female reproductive tract (FRT) is one of the major mucosal invasion sites for HIV-1. This site has been neglected in previous HIV-1 vaccine studies. Immune responses in the FRT after systemic vaccination remain to be characterized. Using a modified vaccinia virus Ankara (MVA) as a vaccine model, we characterized specific immune responses in all compartments of the FRT of nonhuman primates after systemic vaccination. Memory T cells were preferentially found in the lower tract (vagina and cervix), whereas APCs and innate lymphoid cells were mainly located in the upper tract (uterus and fallopian tubes). This compartmentalization of immune cells in the FRT was supported by transcriptomic analyses and a correlation network. Polyfunctional MVA-specific CD8+ T cells were detected in the blood, lymph nodes, vagina, cervix, uterus, and fallopian tubes. Anti-MVA IgG and IgA were detected in cervicovaginal fluid after a second vaccine dose. Thus, systemic vaccination with an MVA vector elicits cellular and Ab responses in the FRT.
Collapse
Affiliation(s)
- Romain Marlin
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France.,Mucosal Innate Immunity and Sexually Transmitted Infections Control Group, Department of Virology, Institut Pasteur, 75015 Paris, France.,Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France
| | - Marie-Thérèse Nugeyre
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France.,Mucosal Innate Immunity and Sexually Transmitted Infections Control Group, Department of Virology, Institut Pasteur, 75015 Paris, France.,Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France
| | - Nicolas Tchitchek
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France
| | - Matteo Parenti
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France
| | - Hakim Hocini
- Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France.,Faculté de Médecine, Université Paris-Est, INSERM U955, 94010 Créteil, France
| | - Fahd Benjelloun
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France.,Mucosal Innate Immunity and Sexually Transmitted Infections Control Group, Department of Virology, Institut Pasteur, 75015 Paris, France
| | - Claude Cannou
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France.,Mucosal Innate Immunity and Sexually Transmitted Infections Control Group, Department of Virology, Institut Pasteur, 75015 Paris, France
| | - Nathalie Dereuddre-Bosquet
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France
| | - Yves Levy
- Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France.,Faculté de Médecine, Université Paris-Est, INSERM U955, 94010 Créteil, France.,Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Assistance Publique-Hôpitaux de Paris, 94010 Créteil, France
| | - Françoise Barré-Sinoussi
- Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France.,Division Internationale, Institut Pasteur, 75015 Paris, France; and
| | - Gabriella Scarlatti
- Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France.,Viral Evolution and Transmission Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roger Le Grand
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France
| | - Elisabeth Menu
- Immunologie des Infections Virales et des Maladies Auto-immunes (ImVA)/Infrastructure Nationale pour la Modélisation des Maladies Infectieuses Humaines et les Thérapies Innovantes (IDMIT)/Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut des Maladies Emergentes et des Traitements Innovants (IMETI), Université Paris-Sud, INSERM U1184, 92265 Fontenay-Aux-Roses, France; .,Mucosal Innate Immunity and Sexually Transmitted Infections Control Group, Department of Virology, Institut Pasteur, 75015 Paris, France.,Vaccine Research Institute, Henri Mondor Hospital, 94010 Créteil, France
| |
Collapse
|
91
|
HIV-Enhancing and HIV-Inhibiting Properties of Cationic Peptides and Proteins. Viruses 2017; 9:v9050108. [PMID: 28505117 PMCID: PMC5454421 DOI: 10.3390/v9050108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 12/26/2022] Open
Abstract
Cationic antimicrobial peptides and proteins have historically been ascribed roles in innate immunity that infer killing of microbial and viral pathogens and protection of the host. In the context of sexually transmitted HIV-1, we take an unconventional approach that questions this paradigm. It is becoming increasingly apparent that many of the cationic polypeptides present in the human genital or anorectal mucosa, or human semen, are capable of enhancing HIV-1 infection, often in addition to other reported roles as viral inhibitors. We explore how the in vivo environment may select for or against the HIV-enhancing aspects of these cationic polypeptides by focusing on biological relevance. We stress that the distinction between enhancing and inhibiting HIV-1 infection is not mutually exclusive to specific classes of cationic polypeptides. Understanding how virally enhancing peptides and proteins act to promote sexual transmission of HIV-1 would be important for the design of topical microbicides, mucosal vaccines, and other preventative measures.
Collapse
|
92
|
Herbst-Kralovetz MM, Pyles RB, Ratner AJ, Sycuro LK, Mitchell C. New Systems for Studying Intercellular Interactions in Bacterial Vaginosis. J Infect Dis 2017; 214 Suppl 1:S6-S13. [PMID: 27449872 DOI: 10.1093/infdis/jiw130] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bacterial vaginosis (BV) affects almost a quarter of US women, making it a condition of major public health relevance. Key questions remain regarding the etiology of BV, mechanisms for its association with poor reproductive health outcomes, and reasons for high rates of treatment failure. New model systems are required to answer these remaining questions, elucidate the complex host-microbe and microbe-microbe interactions, and develop new, effective interventions. In this review, we cover the strengths and limitations of in vitro and in vivo model systems to study these complex intercellular interactions. Furthermore, we discuss advancements needed to maximize the translational utility of the model systems. As no single model can recapitulate all of the complex physiological and environmental conditions of the human vaginal microenvironment, we conclude that combinatorial approaches using in vitro and in vivo model systems will be required to address the remaining fundamental questions surrounding the enigma that is BV.
Collapse
Affiliation(s)
| | - Richard B Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston
| | - Adam J Ratner
- Departments of Pediatrics and Microbiology, New York University School of Medicine, New York
| | - Laura K Sycuro
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Caroline Mitchell
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston
| |
Collapse
|
93
|
Deruaz M, Murooka TT, Ji S, Gavin MA, Vrbanac VD, Lieberman J, Tager AM, Mempel TR, Luster AD. Chemoattractant-mediated leukocyte trafficking enables HIV dissemination from the genital mucosa. JCI Insight 2017; 2:e88533. [PMID: 28405607 DOI: 10.1172/jci.insight.88533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HIV vaginal transmission accounts for the majority of newly acquired heterosexual infections. However, the mechanism by which HIV spreads from the initial site of viral entry at the mucosal surface of the female genital tract to establish a systemic infection of lymphoid and peripheral tissues is not known. Once the virus exits the mucosa it rapidly spreads to all tissues, leading to CD4+ T cell depletion and the establishment of a viral reservoir that cannot be eliminated with current treatments. Understanding the molecular and cellular requirements for viral dissemination from the genital tract is therefore of great importance, as it could reveal new strategies to lengthen the window of opportunity to target the virus at its entry site in the mucosa where it is the most vulnerable and thus prevent systemic infection. Using HIV vaginal infection of humanized mice as a model of heterosexual transmission, we demonstrate that blocking the ability of leukocytes to respond to chemoattractants prevented HIV from leaving the female genital tract. Furthermore, blocking lymphocyte egress from lymph nodes prevented viremia and infection of the gut. Leukocyte trafficking therefore plays a major role in viral dissemination, and targeting the chemoattractant molecules involved can prevent the establishment of a systemic infection.
Collapse
Affiliation(s)
- Maud Deruaz
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas T Murooka
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sophina Ji
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Vladimir D Vrbanac
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew M Tager
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
94
|
Richardson-Harman N, Parody R, Anton P, McGowan I, Doncel G, Thurman AR, Herrera C, Kordy K, Fox J, Tanner K, Swartz G, Dezzutti CS. Analytical Advances in the Ex Vivo Challenge Efficacy Assay. AIDS Res Hum Retroviruses 2017; 33:395-403. [PMID: 27841671 PMCID: PMC5372762 DOI: 10.1089/aid.2016.0073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The ex vivo challenge assay is being increasingly used as an efficacy endpoint during early human clinical trials of HIV prevention treatments. There is no standard methodology for the ex vivo challenge assay, although the use of different data collection methods and analytical parameters may impact results and reduce the comparability of findings between trials. In this analysis, we describe the impact of data imputation methods, kit type, testing schedule and tissue type on variability, statistical power, and ex vivo HIV growth kinetics. Data were p24 antigen (pg/ml) measurements collected from clinical trials of candidate microbicides where rectal (n = 502), cervical (n = 88), and vaginal (n = 110) tissues were challenged with HIV-1BaL ex vivo. Imputation of missing data using a nonlinear mixed effect model was found to provide an improved fit compared to imputation using half the limit of detection. The rectal virus growth period was found to be earlier and of a relatively shorter duration than the growth period for cervical and vaginal tissue types. On average, only four rectal tissue challenge assays in each treatment and control group would be needed to find a one log difference in p24 to be significant (alpha = 0.05), but a larger sample size was predicted to be needed for either cervical (n = 21) or vaginal (n = 10) tissue comparisons. Overall, the results indicated that improvements could be made in the design and analysis of the ex vivo challenge assay to provide a more standardized and powerful assay to compare efficacy of microbicide products.
Collapse
Affiliation(s)
| | - Robert Parody
- Alpha StatConsult, LLC, Damascus, Maryland
- School of Mathematical Sciences, Rochester Institute of Technology, Rochester, New York
| | - Peter Anton
- Department of Medicine, David Geffen School of Medicine at UCLA, Center for HIV Prevention Research, UCLA AIDS Institute, Los Angeles, California
| | - Ian McGowan
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee Womens Research Institute, Pittsburgh, Pennsylvania
| | - Gustavo Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk and Arlington, Virginia
| | | | - Carolina Herrera
- Division of Infectious Diseases, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Kattayoun Kordy
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Southern California, Los Angeles, California
| | - Julie Fox
- Guys and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Karen Tanner
- Department of Medicine, David Geffen School of Medicine at UCLA, Center for HIV Prevention Research, UCLA AIDS Institute, Los Angeles, California
| | - Glenn Swartz
- Advanced Bioscience Laboratories, Gaithersburg, Maryland
| | - Charlene S. Dezzutti
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee Womens Research Institute, Pittsburgh, Pennsylvania
| |
Collapse
|
95
|
Shang L, Duan L, Perkey KE, Wietgrefe S, Zupancic M, Smith AJ, Southern PJ, Johnson RP, Haase AT. Epithelium-innate immune cell axis in mucosal responses to SIV. Mucosal Immunol 2017; 10:508-519. [PMID: 27435105 PMCID: PMC5250613 DOI: 10.1038/mi.2016.62] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/18/2016] [Indexed: 02/04/2023]
Abstract
In the SIV (simian immunodeficiency virus)-rhesus macaque model of HIV-1 (human immunodeficiency virus type I) transmission to women, one hallmark of the mucosal response to exposure to high doses of SIV is CD4 T-cell recruitment that fuels local virus expansion in early infection. In this study, we systematically analyzed the cellular events and chemoattractant profiles in cervical tissues that precede CD4 T-cell recruitment. We show that vaginal exposure to the SIV inoculum rapidly induces chemokine expression in cervical epithelium including CCL3, CCL20, and CXCL8. The chemokine expression is associated with early recruitment of macrophages and plasmacytoid dendritic cells that are co-clustered underneath the cervical epithelium. Production of chemokines CCL3 and CXCL8 by these cells in turn generates a chemokine gradient that is spatially correlated with the recruitment of CD4 T cells. We further show that the protection of SIVmac239Δnef vaccination against vaginal challenge is correlated with the absence of this epithelium-innate immune cell-CD4 T-cell axis response in the cervical mucosa. Our results reveal a critical role for cervical epithelium in initiating early mucosal responses to vaginal infection, highlight an important role for macrophages in target cell recruitment, and provide further evidence of a paradoxical dampening effect of a protective vaccine on these early mucosal responses.
Collapse
Affiliation(s)
- L Shang
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - L Duan
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - K E Perkey
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - S Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - M Zupancic
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - A J Smith
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - P J Southern
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - R P Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - A T Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
96
|
Botting RA, Rana H, Bertram KM, Rhodes JW, Baharlou H, Nasr N, Cunningham AL, Harman AN. Langerhans cells and sexual transmission of HIV and HSV. Rev Med Virol 2017; 27. [PMID: 28044388 DOI: 10.1002/rmv.1923] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/01/2016] [Accepted: 12/02/2016] [Indexed: 12/14/2022]
Abstract
Langerhans cells (LCs) situated in stratified squamous epithelium of the skin and mucosal tissue are amongst the first cells that sexually transmitted pathogens encounter during transmission. They are potent antigen presenting cells and play a key role in the host mounting an appropriate immune response. As such, viruses have evolved complex strategies to manipulate these cells to facilitate successful transmission. One of best studied examples is HIV, which manipulates the natural function of these cells to interact with CD4 T cells, which are the main target cell for HIV in which rapid replication occurs. However, there is controversy in the literature as to the role that LCs play in this process. Langerhans cells also play a key role in the way the body mounts an immune response to HSV, and there is also a complex interplay between the transmission of HSV and HIV that involves LCs. In this article, we review both past and present literatures with a particular focus on a few very recent studies that shed new light on the role that LCs play in the transmission and immune response to these 2 pathogens.
Collapse
Affiliation(s)
- Rachel A Botting
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Hafsa Rana
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Kirstie M Bertram
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Jake W Rhodes
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Najla Nasr
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Anthony L Cunningham
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew N Harman
- The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
97
|
Gibbs A, Leeansyah E, Introini A, Paquin-Proulx D, Hasselrot K, Andersson E, Broliden K, Sandberg JK, Tjernlund A. MAIT cells reside in the female genital mucosa and are biased towards IL-17 and IL-22 production in response to bacterial stimulation. Mucosal Immunol 2017; 10:35-45. [PMID: 27049062 PMCID: PMC5053908 DOI: 10.1038/mi.2016.30] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/25/2016] [Indexed: 02/07/2023]
Abstract
The female genital tract (FGT) mucosa is a critically important site for immune defense against microbes. Mucosal-associated invariant T (MAIT) cells are an innate-like T-cell population that recognizes microbial riboflavin metabolite antigens in an MR1-dependent manner. The role of MAIT cells in the FGT mucosa is unknown. Here, we found that MAIT cells and MR1+ antigen-presenting cells were present in the upper and lower FGT, with distinct tissue localization of MAIT cells in endometrium vs. cervix. The MAIT cells from the FGT and blood displayed a distinct phenotype with expression of interleukin (IL)-18Rα, CD127, α4β7, PD-1, as well as the transcription factors promyelocytic leukemia zinc finger (PLZF), RORγt, Helios, Eomes, and T-bet. Their expression levels of PLZF and Eomes were lower in the FGT compared with blood. When stimulated with Escherichia coli, MAIT cells from the FGT displayed a bias towards IL-17 and IL-22 expression, whereas blood MAIT cells produced primarily IFN-γ, TNF, and Granzyme B. Furthermore, both FGT- and blood-derived MAIT cells were polyfunctional and contributed to the T-cell-mediated response to E. coli. Thus, MAIT cells in the genital mucosa have a distinct IL-17/IL-22 profile and may have an important role in the immunological homeostasis and control of microbes at this site.
Collapse
Affiliation(s)
- Anna Gibbs
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Edwin Leeansyah
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Andrea Introini
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Dominic Paquin-Proulx
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Klara Hasselrot
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
- Department of Obstetrics and Gynecology, Danderyd Hospital, 182 88 Stockholm, Sweden
| | - Emilia Andersson
- Clinical Pathology/Cytology, Capio St. Göran Hospital, Stockholm, Sweden
| | - Kristina Broliden
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Annelie Tjernlund
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| |
Collapse
|
98
|
Abstract
Urinary tract infections (UTI) are among the most common bacterial infections in humans, affecting millions of people every year. UTI cause significant morbidity in women throughout their lifespan, in infant boys, in older men, in individuals with underlying urinary tract abnormalities, and in those that require long-term urethral catheterization, such as patients with spinal cord injuries or incapacitated individuals living in nursing homes. Serious sequelae include frequent recurrences, pyelonephritis with sepsis, renal damage in young children, pre-term birth, and complications of frequent antimicrobial use including high-level antibiotic resistance and Clostridium difficile colitis. Uropathogenic E. coli (UPEC) cause the vast majority of UTI, but less common pathogens such as Enterococcus faecalis and other enterococci frequently take advantage of an abnormal or catheterized urinary tract to cause opportunistic infections. While antibiotic therapy has historically been very successful in controlling UTI, the high rate of recurrence remains a major problem, and many individuals suffer from chronically recurring UTI, requiring long-term prophylactic antibiotic regimens to prevent recurrent UTI. Furthermore, the global emergence of multi-drug resistant UPEC in the past ten years spotlights the need for alternative therapeutic and preventative strategies to combat UTI, including anti-infective drug therapies and vaccines. In this chapter, we review recent advances in the field of UTI pathogenesis, with an emphasis on the identification of promising drug and vaccine targets. We then discuss the development of new UTI drugs and vaccines, highlighting the challenges these approaches face and the need for a greater understanding of urinary tract mucosal immunity.
Collapse
|
99
|
Edwards JL, Jennings MP, Apicella MA, Seib KL. Is gonococcal disease preventable? The importance of understanding immunity and pathogenesis in vaccine development. Crit Rev Microbiol 2016; 42:928-41. [PMID: 26805040 PMCID: PMC4958600 DOI: 10.3109/1040841x.2015.1105782] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/01/2015] [Accepted: 10/06/2015] [Indexed: 12/31/2022]
Abstract
Gonorrhea is a major, global public health problem for which there is no vaccine. The continuing emergence of antibiotic-resistant strains raises concerns that untreatable Neisseria gonorrhoeae may become widespread in the near future. Consequently, there is an urgent need for increased efforts towards the development of new anti-gonococcal therapeutics and vaccines, as well as suitable models for potential pre-clinical vaccine trials. Several current issues regarding gonorrhea are discussed herein, including the global burden of disease, the emergence of antibiotic-resistance, the status of vaccine development and, in particular, a focus on the model systems available to evaluate drug and vaccine candidates. Finally, alternative approaches to evaluate vaccine candidates are presented. Such approaches may provide valuable insights into the protective mechanisms, and correlates of protection, required to prevent gonococcal transmission, local infection and disease sequelae.
Collapse
Affiliation(s)
- Jennifer L. Edwards
- Department of Pediatrics, The Research Institute at Nationwide Children's Hospital and The Ohio State UniversityColumbus,
OH,
USA
| | | | | | - Kate L. Seib
- Institute for Glycomics, Griffith University,
Gold Coast,
Australia
| |
Collapse
|
100
|
Salpingectomy and prevention of ovarian carcinoma. Gynecol Minim Invasive Ther 2016. [DOI: 10.1016/j.gmit.2015.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|