1
|
Zhang Y, Klein K, Ratcliff A, Galappaththi SL, Hathaway N, Twells N, Patel M, Temesy S, Bailey J, Mahal L, Creuzenet C, Arts E. Transmitted/founder (T/F) HIV-1 derived from sexual contact exhibits greater transmission fitness in human cervical tissue than T/F HIV-1 from blood-to-blood contact: Unique glycan profiles on T/F envelopes associated with transmission phenotypes. PLoS Pathog 2025; 21:e1013177. [PMID: 40408432 DOI: 10.1371/journal.ppat.1013177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) risk groups include, but are not limited to, heterosexual individuals (HET), men-who-have-sex-with-men (MSM), and people who inject drugs (PWID). Although genetically diverse HIV-1 populations are transferred from donor to recipient, systemic infection is often established by a single clone, the transmitted/founder (T/F) virus. This phenomenon is especially prevalent in sexual transmission, but less stringent in blood-to-blood contact transmission. Specific traits that permit successful transmission have not been well characterized. Thus, HIV-1 containing the chimeric T/F envelope (Env) from different transmission routes was assessed for ex vivo transmission fitness by performing mixed competition assays (also referred to as mixed competitions) on human cervical tissues. We found that chimeric T/F viruses isolated from the PWID exhibit limited replication capacity in cervical tissues when compared to those from MSM and HET, diminishing their chances of transmission to T helper type 1 (Th1) and Th17 cells. This reduced transmission fitness of T/F HIV-1 from PWID was not observed when infecting Th1 and Th17 cells directly, bypassing cervical tissues. Phenotypic assays showed that the chimeric T/F viruses from PWID differed from other groups by having an enhanced ability to utilize diverse CCR5 conformations, while Env expression level, CD4/CCR5 utilization, and entry speed did not differ. Different glycosylation profiles were detected on T/F compared to chronic Env with increased complex, fucosylated N- and O-glycans found more frequently on the T/F Env. Furthermore, the increased presence of these fucosylated glycans correlated with replication fitness in cervical tissues. In contrast, bisecting branched N-glycan found more frequently on chronic Env was associated with decreased entry efficiency and more stringent usage of CCR5. These findings suggest that glycosylation patterns/levels and/or Env structure greatly impact the differences in transmission fitness of T/F HIV-1.
Collapse
Affiliation(s)
- Yiying Zhang
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Katja Klein
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, United Kingdom
| | - Annette Ratcliff
- Department of Molecular Biology and Microbiology and Division of Infectious Diseases, Case Western Reserve University, Cleveland, United States of America
| | | | - Nicholas Hathaway
- Department of Pathology and Laboratory Medicine, Brown University, Providence, United States of America
| | - Nicholas Twells
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Mukti Patel
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Stephen Temesy
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Jeffrey Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, United States of America
| | - Lara Mahal
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Carole Creuzenet
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Eric Arts
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Department of Molecular Biology and Microbiology and Division of Infectious Diseases, Case Western Reserve University, Cleveland, United States of America
| |
Collapse
|
2
|
Izadifar Z, Stejskalova A, Gulati A, Gutzeit O, Ingber DE. Human Cervix Chip: A Preclinical Model for Studying the Role of the Cervical Mucosa and Microbiome in Female Reproductive Health. Bioessays 2025:e70014. [PMID: 40401380 DOI: 10.1002/bies.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Advancements in women's reproductive health have been hindered by insufficient knowledge and the underrepresentation of women in research, leading to symptom-focused care with poor outcomes. Modeling female reproductive biology and disease pathophysiology has been challenging due to the complexity and dynamic nature of the female organs. Here, we briefly review recent advancements made with a new in vitro microfluidic organ-on-a-chip model of the human cervix (Cervix Chip) that faithfully mimics key features of the cervix, including mucus production and physiological responses to hormonal, environmental, and microbial stimuli. We also discuss how this preclinical platform can provide a way to obtain unique insights into the role of mucosal immunity, genetic and risk factors, as well as microbiome and pathogen interactions in human cervix health and disease, while bridging knowledge gaps in fertility and pregnancy-related conditions. By enabling preclinical drug screening and accelerating translational research, the Cervix Chip holds the potential to improve the development of therapeutics, diagnostics, and ultimately, the sexual and reproductive health of millions of women globally.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Urology Department, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Ola Gutzeit
- IVF Unit, Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
- Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
Avitabile E, Menotti L, Croatti V, Giordani B, Parolin C, Vitali B. Protective Mechanisms of Vaginal Lactobacilli against Sexually Transmitted Viral Infections. Int J Mol Sci 2024; 25:9168. [PMID: 39273118 PMCID: PMC11395631 DOI: 10.3390/ijms25179168] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The healthy cervicovaginal microbiota is dominated by various Lactobacillus species, which support a condition of eubiosis. Among their many functions, vaginal lactobacilli contribute to the maintenance of an acidic pH, produce antimicrobial compounds, and modulate the host immune response to protect against vaginal bacterial and fungal infections. Increasing evidence suggests that these beneficial bacteria may also confer protection against sexually transmitted infections (STIs) caused by viruses such as human papillomavirus (HPV), human immunodeficiency virus (HIV) and herpes simplex virus (HSV). Viral STIs pose a substantial public health burden globally, causing a range of infectious diseases with potentially severe consequences. Understanding the molecular mechanisms by which lactobacilli exert their protective effects against viral STIs is paramount for the development of novel preventive and therapeutic strategies. This review aims to provide more recent insights into the intricate interactions between lactobacilli and viral STIs, exploring their impact on the vaginal microenvironment, host immune response, viral infectivity and pathogenesis, and highlighting their potential implications for public health interventions and clinical management strategies.
Collapse
Affiliation(s)
- Elisa Avitabile
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Laura Menotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Vanessa Croatti
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Barbara Giordani
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| |
Collapse
|
4
|
Tuniyazi M, Zhang N. Possible Therapeutic Mechanisms and Future Perspectives of Vaginal Microbiota Transplantation. Microorganisms 2023; 11:1427. [PMID: 37374929 PMCID: PMC10305445 DOI: 10.3390/microorganisms11061427] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Microbial communities inhabiting the human body play a crucial role in protecting the host against pathogens and inflammation. Disruptions to the microbial composition can lead to various health issues. Microbial transfer therapy (MTT) has emerged as a potential treatment option to address such issues. Fecal microbiota transplantation (FMT) is the most widely used form of MTT and has been successful in treating several diseases. Another form of MTT is vaginal microbiota transplantation (VMT), which involves transferring vaginal microbiota from a healthy female donor to a diseased patient's vaginal cavity with the goal of restoring normal vaginal microbial composition. However, VMT has not been extensively studied due to safety concerns and a lack of research. This paper explores the therapeutic mechanisms of VMT and discusses future perspectives. Further research is necessary to advance the clinical applications and techniques of VMT.
Collapse
Affiliation(s)
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| |
Collapse
|
5
|
Short CES, Quinlan R, Lee YS, Preda VG, Smith A, Marchesi JR, Shattock R, Bennett PR, MacIntyre DA, Taylor GP. Comparative analysis of vaginal microbiota sampling using menstrual cups and high vaginal swabs in pregnant women living with HIV-1 infection. Front Cell Infect Microbiol 2023; 13:1190160. [PMID: 37228662 PMCID: PMC10204588 DOI: 10.3389/fcimb.2023.1190160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Background Menstrual cups (MCs) are increasingly used to collect cervicovaginal secretions to characterise vaginal mucosal immunology, in conjunction with high vaginal swabs (HVS) for metataxonomics, particularly in HIV transmission studies. We hypothesised that both methods of collecting bacterial biomass are equivalent for 16S rRNA gene sequencing. Material and Methods Cervicovaginal fluid (CVF) samples from 16 pregnant women with HIV-1 (PWWH) were included to represent the major vaginal bacterial community state types (CST I-V). Women underwent sampling during the second trimester by liquid amies HVS followed by a MC (Soft disc™) and samples were stored at -80°C. Bacterial cell pellets obtained from swab elution and MC (500 µL, 1 in 10 dilution) were resuspended in 120 µL PBS for DNA extraction. Bacterial 16S rRNA gene sequencing was performed using V1-V2 primers and were analysed using MOTHUR. Paired total DNA, bacterial load, amplicon read counts, diversity matrices and bacterial taxa were compared by sampling method using MicrobiomeAnalyst, SPSS and R. Results The total DNA eluted from one aliquot of diluted CVF from an MC was similar to that of a HVS (993ng and 609ng, p=0.18); the mean bacterial loads were also comparable for both methods (MC: 8.0 log10 16S rRNA gene copies versus HVS: 7.9 log10 16S rRNA gene copies, p=0.27). The mean number of sequence reads generated from MC samples was lower than from HVS (MC: 12730; HVS:14830, p=0.05). The α-diversity metrices were similar for both techniques; MC Species Observed: 41 (range 12-96) versus HVS: 47 (range 16-96), p=0.15; MC Inverse Simpson Index: 1.98 (range 1.0-4.0) versus HVS: 0.48 (range 1.0-4.4), p=0.22). The three most abundant species observed were: Lactobacillus iners, Lactobacillus crispatus and Gardnerella vaginalis. Hierarchical clustering of relative abundance data showed that samples obtained using different techniques in an individual clustered in the same CST group. Conclusion These data demonstrate that despite sampling slightly different areas of the lower genital tract, there was no difference in bacterial load or composition between methods. Both are suitable for characterisation of vaginal microbiota in PWWH. The MC offers advantages, including a higher volume of sample available for DNA extraction and complimentary assays.
Collapse
Affiliation(s)
- Charlotte-Eve S. Short
- Section of Virology, Department of Infectious Disease, Imperial College London, London, United Kingdom
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Rachael Quinlan
- Section of Virology, Department of Infectious Disease, Imperial College London, London, United Kingdom
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Yun S. Lee
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Veronica G. Preda
- Section of Virology, Department of Infectious Disease, Imperial College London, London, United Kingdom
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ann Smith
- Faculty of Health and Applied Sciences, University West of England, Bristol, United Kingdom
| | - Julian R. Marchesi
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
- Marchesi Laboratory, Department of Metabolism, Digestion, and Reproduction, Division of Digestive Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Robin Shattock
- Section of Immunology of Infection, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Phillip R. Bennett
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - David A. MacIntyre
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Graham P. Taylor
- Section of Virology, Department of Infectious Disease, Imperial College London, London, United Kingdom
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Williams LD, Shen X, Sawant SS, Akapirat S, Dahora LC, Tay MZ, Stanfield-Oakley S, Wills S, Goodman D, Tenney D, Spreng RL, Zhang L, Yates NL, Montefiori DC, Eller MA, Easterhoff D, Hope TJ, Rerks-Ngarm S, Pittisuttithum P, Nitayaphan S, Excler JL, Kim JH, Michael NL, Robb ML, O’Connell RJ, Karasavvas N, Vasan S, Ferrari G, Tomaras GD, RV305 study team. Viral vector delivered immunogen focuses HIV-1 antibody specificity and increases durability of the circulating antibody recall response. PLoS Pathog 2023; 19:e1011359. [PMID: 37256916 PMCID: PMC10284421 DOI: 10.1371/journal.ppat.1011359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/21/2023] [Accepted: 04/14/2023] [Indexed: 06/02/2023] Open
Abstract
The modestly efficacious HIV-1 vaccine regimen (RV144) conferred 31% vaccine efficacy at 3 years following the four-shot immunization series, coupled with rapid waning of putative immune correlates of decreased infection risk. New strategies to increase magnitude and durability of protective immunity are critically needed. The RV305 HIV-1 clinical trial evaluated the immunological impact of a follow-up boost of HIV-1-uninfected RV144 recipients after 6-8 years with RV144 immunogens (ALVAC-HIV alone, AIDSVAX B/E gp120 alone, or ALVAC-HIV + AIDSVAX B/E gp120). Previous reports demonstrated that this regimen elicited higher binding, antibody Fc function, and cellular responses than the primary RV144 regimen. However, the impact of the canarypox viral vector in driving antibody specificity, breadth, durability and function is unknown. We performed a follow-up analysis of humoral responses elicited in RV305 to determine the impact of the different booster immunogens on HIV-1 epitope specificity, antibody subclass, isotype, and Fc effector functions. Importantly, we observed that the ALVAC vaccine component directly contributed to improved breadth, function, and durability of vaccine-elicited antibody responses. Extended boosts in RV305 increased circulating antibody concentration and coverage of heterologous HIV-1 strains by V1V2-specific antibodies above estimated protective levels observed in RV144. Antibody Fc effector functions, specifically antibody-dependent cellular cytotoxicity and phagocytosis, were boosted to higher levels than was achieved in RV144. V1V2 Env IgG3, a correlate of lower HIV-1 risk, was not increased; plasma Env IgA (specifically IgA1), a correlate of increased HIV-1 risk, was elevated. The quality of the circulating polyclonal antibody response changed with each booster immunization. Remarkably, the ALVAC-HIV booster immunogen induced antibody responses post-second boost, indicating that the viral vector immunogen can be utilized to selectively enhance immune correlates of decreased HIV-1 risk. These results reveal a complex dynamic of HIV-1 immunity post-vaccination that may require careful balancing to achieve protective immunity in the vaccinated population. Trial registration: RV305 clinical trial (ClinicalTrials.gov number, NCT01435135). ClinicalTrials.gov Identifier: NCT00223080.
Collapse
Affiliation(s)
- LaTonya D. Williams
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sheetal S. Sawant
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Siriwat Akapirat
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Lindsay C. Dahora
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Matthew Zirui Tay
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sherry Stanfield-Oakley
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Saintedym Wills
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Derrick Goodman
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - DeAnna Tenney
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Rachel L. Spreng
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lu Zhang
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nicole L. Yates
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Michael A. Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - David Easterhoff
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | | | - Punnee Pittisuttithum
- Royal Thai Army Component, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sorachai Nitayaphan
- Royal Thai Army Component, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Jean-Louis Excler
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jerome H. Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Nelson L. Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Merlin L. Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Robert J. O’Connell
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Nicos Karasavvas
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sandhya Vasan
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Guido Ferrari
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | | |
Collapse
|
7
|
d'Amone L, Sahoo JK, Ostrovsky-Snider N, Kaplan DL, Omenetto FG. Boronic Acid-Tethered Silk Fibroin for pH-Dependent Mucoadhesion. Biomacromolecules 2023; 24:1310-1317. [PMID: 36763594 DOI: 10.1021/acs.biomac.2c01349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Mucus lines all surfaces of the human body not covered by skin and provides lubrication, hydration, and protection. The properties of mucus are influenced by changes in pH that may occur due to physiological conditions and pathological circumstances. Reinforcing the mucus barrier with biopolymers that can adhere to mucus in different conditions can be a useful strategy for protecting the underlying mucosae from damage. In this work, regenerated silk fibroin (silk) was chemically modified with phenyl boronic acid to form reversible covalent complexes with the 1,2- or 1,3-diols. The silk modified with boronic acid pendant groups has an increased affinity for mucins, whose carbohydrate component is rich in diols. These results offer new applications of silk in mucoadhesion, and the ability to bind diols to the silk lays the foundation for the development of silk-based sugar-sensing platforms.
Collapse
Affiliation(s)
- Luciana d'Amone
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | - Fiorenzo G Omenetto
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
- Department of Electrical and Computer Engineering, Tufts University, Medford, Massachusetts 02153, United States
- Department of Physics, Tufts University, Medford, Massachusetts 02153, United States
- Laboratory for Living Devices, Tufts University, Medford, Massachusetts 02153, United States
| |
Collapse
|
8
|
Pendharkar S, Skafte-Holm A, Simsek G, Haahr T. Lactobacilli and Their Probiotic Effects in the Vagina of Reproductive Age Women. Microorganisms 2023; 11:microorganisms11030636. [PMID: 36985210 PMCID: PMC10056154 DOI: 10.3390/microorganisms11030636] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/07/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
In the present narrative review, the probiotic effects of vaginal Lactobacillus spp. are described in detail, covering the importance of the differential production of lactic acid, the lactic acid D/L isoforms, the questionable in vivo effect of hydrogen peroxide, as well as bacteriocins and other core proteins produced by vaginal Lactobacillus spp. Moreover, the microbe–host interaction is explained with emphasis on the vaginal mucosa. To understand the crucial role of Lactobacillus spp. dominance in the vaginal microbiota, different dysbiotic states of the vagina are explained including bacterial vaginosis and aerobic vaginitis. Finally, this review takes on the therapeutic aspect of live lactobacilli in the context of bacterial vaginosis. Until recently, there was very low-quality evidence to suggest that any probiotic might aid in reducing vaginal infections or dysbiosis. Therefore, clinical usage or over the counter usage of probiotics was not recommended. However, recent progress has been made, moving from probiotics that are typically regulated as food supplements to so-called live biotherapeutic products that are regulated as medical drugs. Thus, recently, a phase 2b trial using a Lactobacillus crispatus strain as a therapeutic add-on to standard metronidazole showed significant reduction in the recurrence of bacterial vaginosis by 12 weeks compared to placebo. This may constitute evidence for a brighter future where the therapeutic use of lactobacilli can be harnessed to improve women’s health.
Collapse
Affiliation(s)
| | - Axel Skafte-Holm
- Research Unit for Reproductive Microbiology, Department of Bacteria, Parasites and Fungi, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Gizem Simsek
- Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thor Haahr
- Department of Gynecology and Obstetrics, Aarhus University Hospital, 8200 Aarhus, Denmark
- Correspondence:
| |
Collapse
|
9
|
Caputo V, Libera M, Sisti S, Giuliani B, Diotti RA, Criscuolo E. The initial interplay between HIV and mucosal innate immunity. Front Immunol 2023; 14:1104423. [PMID: 36798134 PMCID: PMC9927018 DOI: 10.3389/fimmu.2023.1104423] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Human Immunodeficiency Virus (HIV) is still one of the major global health issues, and despite significant efforts that have been put into studying the pathogenesis of HIV infection, several aspects need to be clarified, including how innate immunity acts in different anatomical compartments. Given the nature of HIV as a sexually transmitted disease, one of the aspects that demands particular attention is the mucosal innate immune response. Given this scenario, we focused our attention on the interplay between HIV and mucosal innate response: the different mucosae act as a physical barrier, whose integrity can be compromised by the infection, and the virus-cell interaction induces the innate immune response. In addition, we explored the role of the mucosal microbiota in facilitating or preventing HIV infection and highlighted how its changes could influence the development of several opportunistic infections. Although recent progress, a proper characterization of mucosal innate immune response and microbiota is still missing, and further studies are needed to understand how they can be helpful for the formulation of an effective vaccine.
Collapse
|
10
|
Dong M, Dong Y, Bai J, Li H, Ma X, Li B, Wang C, Li H, Qi W, Wang Y, Fan A, Han C, Xue F. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol 2023; 13:1124591. [PMID: 36909729 PMCID: PMC9998931 DOI: 10.3389/fcimb.2023.1124591] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
The female reproductive tract harbours hundreds of bacterial species and produces numerous metabolites. The uterine cervix is located between the upper and lower parts of the female genital tract. It allows sperm and birth passage and hinders the upward movement of microorganisms into a relatively sterile uterus. It is also the predicted site for sexually transmitted infection (STI), such as Chlamydia, human papilloma virus (HPV), and human immunodeficiency virus (HIV). The healthy cervicovaginal microbiota maintains cervical epithelial barrier integrity and modulates the mucosal immune system. Perturbations of the microbiota composition accompany changes in microbial metabolites that induce local inflammation, damage the cervical epithelial and immune barrier, and increase susceptibility to STI infection and relative disease progression. This review examined the intimate interactions between the cervicovaginal microbiota, relative metabolites, and the cervical epithelial-, immune-, and mucus barrier, and the potent effect of the host-microbiota interaction on specific STI infection. An improved understanding of cervicovaginal microbiota regulation on cervical microenvironment homeostasis might promote advances in diagnostic and therapeutic approaches for various STI diseases.
Collapse
Affiliation(s)
- Mengting Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yalan Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Junyi Bai
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Ma
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bijun Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenhui Qi
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiping Fan
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| | - Fengxia Xue
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| |
Collapse
|
11
|
Izadifar Z, Sontheimer-Phelps A, Lubamba BA, Bai H, Fadel C, Stejskalova A, Ozkan A, Dasgupta Q, Bein A, Junaid A, Gulati A, Mahajan G, Kim S, LoGrande NT, Naziripour A, Ingber DE. Modeling mucus physiology and pathophysiology in human organs-on-chips. Adv Drug Deliv Rev 2022; 191:114542. [PMID: 36179916 DOI: 10.1016/j.addr.2022.114542] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/25/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
The surfaces of human internal organs are lined by a mucus layer that ensures symbiotic relationships with commensal microbiome while protecting against potentially injurious environmental chemicals, toxins, and pathogens, and disruption of this layer can contribute to disease development. Studying mucus biology has been challenging due to the lack of physiologically relevant human in vitro models. Here we review recent progress that has been made in the development of human organ-on-a-chip microfluidic culture models that reconstitute epithelial tissue barriers and physiologically relevant mucus layers with a focus on lung, colon, small intestine, cervix and vagina. These organ-on-a-chip models that incorporate dynamic fluid flow, air-liquid interfaces, and physiologically relevant mechanical cues can be used to study mucus composition, mechanics, and structure, as well as investigate its contributions to human health and disease with a level of biomimicry not possible in the past.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | | | - Bob A Lubamba
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Cicely Fadel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Alican Ozkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Queeny Dasgupta
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Amir Bein
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Abidemi Junaid
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Gautam Mahajan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Nina T LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States; Vascular Biology Program, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02115, United States; Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02138, United Kingdom.
| |
Collapse
|
12
|
Kretschmer M, Ceña‐Diez R, Butnarasu C, Silveira V, Dobryden I, Visentin S, Berglund P, Sönnerborg A, Lieleg O, Crouzier T, Yan H. Synthetic Mucin Gels with Self-Healing Properties Augment Lubricity and Inhibit HIV-1 and HSV-2 Transmission. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203898. [PMID: 36104216 PMCID: PMC9661867 DOI: 10.1002/advs.202203898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/14/2022] [Indexed: 05/02/2023]
Abstract
Mucus is a self-healing gel that lubricates the moist epithelium and provides protection against viruses by binding to viruses smaller than the gel's mesh size and removing them from the mucosal surface by active mucus turnover. As the primary nonaqueous components of mucus (≈0.2%-5%, wt/v), mucins are critical to this function because the dense arrangement of mucin glycans allows multivalence of binding. Following nature's example, bovine submaxillary mucins (BSMs) are assembled into "mucus-like" gels (5%, wt/v) by dynamic covalent crosslinking reactions. The gels exhibit transient liquefaction under high shear strain and immediate self-healing behavior. This study shows that these material properties are essential to provide lubricity. The gels efficiently reduce human immunodeficiency virus type 1 (HIV-1) and genital herpes virus type 2 (HSV-2) infectivity for various types of cells. In contrast, simple mucin solutions, which lack the structural makeup, inhibit HIV-1 significantly less and do not inhibit HSV-2. Mechanistically, the prophylaxis of HIV-1 infection by BSM gels is found to be that the gels trap HIV-1 by binding to the envelope glycoprotein gp120 and suppress cytokine production during viral exposure. Therefore, the authors believe the gels are promising for further development as personal lubricants that can limit viral transmission.
Collapse
Affiliation(s)
- Martin Kretschmer
- School of Engineering and Design, Department of Materials EngineeringTechnical University of MunichBoltzmannstrasse 1585748GarchingGermany
- Center for Protein AssembliesTechnical University of MunichErnst‐Otto‐Fischer Str. 885748GarchingGermany
| | - Rafael Ceña‐Diez
- Department of Medicine HuddingeDivision of Infectious DiseasesKarolinska University HospitalKarolinska Institutet, I73Stockholm141 86Sweden
| | - Cosmin Butnarasu
- Department of Molecular Biotechnology and Health ScienceUniversity of TurinTurin10135Italy
| | - Valentin Silveira
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
| | - Illia Dobryden
- Division of Bioeconomy and HealthDepartment of Material and Surface DesignRISE Research Institutes of SwedenMalvinas väg 3StockholmSE‐114 86Sweden
| | - Sonja Visentin
- Department of Molecular Biotechnology and Health ScienceUniversity of TurinTurin10135Italy
| | - Per Berglund
- Department of Industrial BiotechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
| | - Anders Sönnerborg
- Department of Medicine HuddingeDivision of Infectious DiseasesKarolinska University HospitalKarolinska Institutet, I73Stockholm141 86Sweden
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials EngineeringTechnical University of MunichBoltzmannstrasse 1585748GarchingGermany
- Center for Protein AssembliesTechnical University of MunichErnst‐Otto‐Fischer Str. 885748GarchingGermany
| | - Thomas Crouzier
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| | - Hongji Yan
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| |
Collapse
|
13
|
Herrera C, Veazey R, Lemke MM, Arnold K, Kim JH, Shattock RJ. Ex Vivo Evaluation of Mucosal Responses to Vaccination with ALVAC and AIDSVAX of Non-Human Primates. Vaccines (Basel) 2022; 10:187. [PMID: 35214645 PMCID: PMC8879115 DOI: 10.3390/vaccines10020187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/01/2023] Open
Abstract
Non-human primates (NHPs) remain the most relevant challenge model for the evaluation of HIV vaccine candidates; however, discrepancies with clinical trial results have emphasized the need to further refine the NHP model. Furthermore, classical evaluation of vaccine candidates is based on endpoints measured systemically. We assessed the mucosal responses elicited upon vaccination with ALVAC and AIDSVAX using ex vivo Rhesus macaque mucosal tissue explant models. Following booster immunization with ALVAC/AIDSVAX, anti-gp120 HIV-1CM244-specific IgG and IgA were detected in culture supernatant cervicovaginal and colorectal tissue explants, as well as systemically. Despite protection from ex vivo viral challenge, no neutralization was observed with tissue explant culture supernatants. Priming with ALVAC induced distinct cytokine profiles in cervical and rectal tissue. However, ALVAC/AIDSVAX boosts resulted in similar modulations in both mucosal tissues with a statistically significant decrease in cytokines linked to inflammatory responses and lymphocyte differentiation. With ALVAC/AIDSVAX boosts, significant correlations were observed between cytokine levels and specific IgA in cervical explants and specific IgG and IgA in rectal tissue. The cytokine secretome revealed differences between vaccination with ALVAC and ALVAC/AIDSVAX not previously observed in mucosal tissues and distinct from the systemic response, which could represent a biosignature of the vaccine combination.
Collapse
Affiliation(s)
- Carolina Herrera
- Department of Medicine, Imperial College London, London W2 1PG, UK;
| | - Ronald Veazey
- Tulane National Primate Research Center, School of Medicine, Tulane University, Covington, GA 70433, USA;
| | - Melissa M. Lemke
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (M.M.L.); (K.A.)
| | - Kelly Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (M.M.L.); (K.A.)
| | - Jerome H. Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MA 20817, USA;
| | | |
Collapse
|
14
|
Armstrong E, Kaul R. Beyond bacterial vaginosis: vaginal lactobacilli and HIV risk. MICROBIOME 2021; 9:239. [PMID: 34893070 PMCID: PMC8665571 DOI: 10.1186/s40168-021-01183-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 05/17/2023]
Abstract
HIV incidence continues to be unacceptably high in Eastern and Southern Africa, with women disproportionately affected. An increased per-contact risk of HIV acquisition among African, Caribbean, and other Black (ACB) women has been associated with the higher prevalence of bacterial vaginosis (BV) in these communities, wherein the vaginal microbiota is predominated by diverse pro-inflammatory anaerobic bacteria. However, while the vaginal microbiota in BV-free women is typically predominated by one of several different Lactobacillus spp., the degree of HIV protection afforded by a Lactobacillus-predominant vaginal microbiota also varies considerably. Specifically, L. crispatus is associated with an immunoregulatory genital immune environment, exclusion of BV-associated bacteria, and reduced HIV risk. In contrast, less HIV protection or exclusion of BV-associated bacteria and fewer immune benefits have been associated with L. iners-which is unfortunately the most common Lactobacillus species among ACB women. These species-specific clinical differences are underpinned by substantial genomic differences between Lactobacillus species: for instance, the much smaller genome of L. iners lacks the coding sequence for D-lactic acid dehydrogenase and cannot produce the D-lactate isomer that enhances HIV trapping in mucus but encodes for epithelial cell toxins and stress resistance proteins that may enhance bacterial survival in the context of microbiota and environmental fluctuations. While more studies are needed to elucidate whether differences in HIV protection between Lactobacillus species are due to direct genital immune effects or the exclusion of proinflammatory BV-associated bacteria, the current body of work suggests that for BV treatment to succeed as an HIV prevention strategy, it may be necessary to induce a vaginal microbiota that is predominated by specific (non-iners) Lactobacillus species. Video abstract.
Collapse
Affiliation(s)
- Eric Armstrong
- Department of Medicine, University of Toronto, Toronto, Canada.
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Canada
- University Health Network, Toronto General Hospital, Immunodeficiency Clinic, Toronto, Canada
| |
Collapse
|
15
|
Jewanraj J, Ngcapu S, Liebenberg LJP. Semen: A modulator of female genital tract inflammation and a vector for HIV-1 transmission. Am J Reprod Immunol 2021; 86:e13478. [PMID: 34077596 PMCID: PMC9286343 DOI: 10.1111/aji.13478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
In order to establish productive infection in women, HIV must transverse the vaginal epithelium and gain access to local target cells. Genital inflammation contributes to the availability of HIV susceptible cells at the female genital mucosa and is associated with higher HIV transmission rates in women. Factors that contribute to genital inflammation may subsequently increase the risk of HIV infection in women. Semen is a highly immunomodulatory fluid containing several bioactive molecules with the potential to influence inflammation and immune activation at the female genital tract. In addition to its role as a vector for HIV transmission, semen induces profound mucosal changes to prime the female reproductive tract for conception. Still, most studies of mucosal immunity are conducted in the absence of semen or without considering its immune impact on the female genital tract. This review discusses the various mechanisms by which semen exposure may influence female genital inflammation and highlights the importance of routine screening for semen biomarkers in vaginal specimens to account for its impact on genital inflammation.
Collapse
Affiliation(s)
- Janine Jewanraj
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Lenine J. P. Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
16
|
VanBenschoten HM, Woodrow KA. Vaginal delivery of vaccines. Adv Drug Deliv Rev 2021; 178:113956. [PMID: 34481031 PMCID: PMC8722700 DOI: 10.1016/j.addr.2021.113956] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/06/2021] [Accepted: 08/28/2021] [Indexed: 11/22/2022]
Abstract
Recent estimates suggest that one in two sexually active individuals will acquire a sexually transmitted infection by age 25, an alarming statistic that amounts to over 1 million new infections per day worldwide. Vaccination against STIs is highly desirable for alleviating this global burden of disease. Vaginal immunization is a promising strategy to combat transmission via the vaginal mucosa. The vagina is typically considered a poor inductive site for common correlates of adaptive immunity. However, emerging evidence suggests that immune tolerance may be overcome by precisely engineered vaccination schemes that orchestrate cell-mediated immunity and establish tissue resident memory immune cells. In this review, we will discuss the unique immunological milieu of the vaginal mucosa and our current understanding of correlates of pathogenesis and protection for several common STIs. We then present a summary of recent vaginal vaccine studies and explore the role that mucosal adjuvants and delivery systems play in enhancing protection according to requisite features of immunity. Finally, we offer perspectives on the challenges and future directions of vaginal vaccine delivery, discussing remaining physiological barriers and innovative vaccine formulations that may overcome them.
Collapse
Affiliation(s)
- Hannah M VanBenschoten
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
17
|
Abbasi A, Aghebati-Maleki L, Homayouni-Rad A. The promising biological role of postbiotics derived from probiotic Lactobacillus species in reproductive health. Crit Rev Food Sci Nutr 2021; 62:8829-8841. [PMID: 34152234 DOI: 10.1080/10408398.2021.1935701] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent investigations have meaningfully developed our knowledge of the features of the reproductive microbiome/metabolome profile and their relations with host responses to offer an optimal milieu for the development of the embryo during the peri-implantation period and throughout pregnancy. In this context, the establishment of homeostatic circumstances in the Female Reproductive Tract (FRT), in various physiological periods, is a significant challenge, which appears the application of postbiotics can facilitate the achievement of this goal. So, currently, scientific literature confirms that postbiotics due to their antimicrobial, antiviral, and immunomodulatory properties can be considered as a novel biotherapeutic approach. Future investigation in this field will shed more translational mechanistic understanding of the interaction of the postbiotics derived from vaginal Lactobacilli with females' health and reproduction.
Collapse
Affiliation(s)
- Amin Abbasi
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Aziz Homayouni-Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Ayele H, Perner M, McKinnon LR, Birse K, Farr Zuend C, Burgener A. An updated review on the effects of depot medroxyprogesterone acetate on the mucosal biology of the female genital tract. Am J Reprod Immunol 2021; 86:e13455. [PMID: 33991137 PMCID: PMC8459266 DOI: 10.1111/aji.13455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 11/29/2022] Open
Abstract
Background Access to safe, effective, and affordable contraception is important for women’s health and essential to mitigate maternal and fetal mortality rates. The progestin‐based contraceptive depot medroxyprogesterone acetate (DMPA) is a popular contraceptive choice with a low failure rate and convenient administration schedule. Aim In this review, we compiled observational data from human cohorts that examine how DMPA influences the mucosal biology of the female genital tract (FGT) that are essential in maintaining vaginal health, including resident immune cells, pro‐inflammatory cytokines, epithelial barrier function, and the vaginal microbiome Materials and Methods This review focused on the recent published literature published in 2019 and 2020. Results Recent longitudinal studies show that DMPA use associates with an immunosuppressive phenotype, increase in CD4+CCR5+ T cells, and alterations to growth factors. In agreement with previous meta‐analyses, DMPA use is associated with minimal effects of the composition of the vaginal microbiome. Cross‐sectional studies associate a more pro‐inflammatory relationship with DMPA, but these studies are confounded by inherent weaknesses of cross‐sectional studies, including differences in study group sizes, behaviors, and other variables that may affect genital inflammation. Discussion & Conclusion These recent results indicate that the interactions between DMPA and the vaginal mucosa are complex emphasizing the need for comprehensive longitudinal studies that take into consideration the measurement of multiple biological parameters.
Collapse
Affiliation(s)
- Hossaena Ayele
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michelle Perner
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lyle R McKinnon
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kenzie Birse
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christina Farr Zuend
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adam Burgener
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.,Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada.,Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Solna, Sweden
| |
Collapse
|
19
|
Pollara J, Tay MZ, Edwards RW, Goodman D, Crowley AR, Edwards RJ, Easterhoff D, Conley HE, Hoxie T, Gurley T, Jones C, Machiele E, Tuyishime M, Donahue E, Jha S, Spreng RL, Hope TJ, Wiehe K, He MM, Moody MA, Saunders KO, Ackerman ME, Ferrari G, Tomaras GD. Functional Homology for Antibody-Dependent Phagocytosis Across Humans and Rhesus Macaques. Front Immunol 2021; 12:678511. [PMID: 34093580 PMCID: PMC8174565 DOI: 10.3389/fimmu.2021.678511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Analyses of human clinical HIV-1 vaccine trials and preclinical vaccine studies performed in rhesus macaque (RM) models have identified associations between non-neutralizing Fc Receptor (FcR)-dependent antibody effector functions and reduced risk of infection. Specifically, antibody-dependent phagocytosis (ADP) has emerged as a common correlate of reduced infection risk in multiple RM studies and the human HVTN505 trial. This recurrent finding suggests that antibody responses with the capability to mediate ADP are most likely a desirable component of vaccine responses aimed at protecting against HIV-1 acquisition. As use of RM models is essential for development of the next generation of candidate HIV-1 vaccines, there is a need to determine how effectively ADP activity observed in RMs translates to activity in humans. In this study we compared ADP activity of human and RM monocytes and polymorphonuclear leukocytes (PMN) to bridge this gap in knowledge. We observed considerable variability in the magnitude of monocyte and PMN ADP activity across individual humans and RM that was not dependent on FcR alleles, and only modestly impacted by cell-surface levels of FcRs. Importantly, we found that for both human and RM phagocytes, ADP activity of antibodies targeting the CD4 binding site was greatest when mediated by human IgG3, followed by RM and human IgG1. These results demonstrate that there is functional homology between antibody and FcRs from these two species for ADP. We also used novel RM IgG1 monoclonal antibodies engineered with elongated hinge regions to show that hinge elongation augments RM ADP activity. The RM IgGs with engineered hinge regions can achieve ADP activity comparable to that observed with human IgG3. These novel modified antibodies will have utility in passive immunization studies aimed at defining the role of IgG3 and ADP in protection from virus challenge or control of disease in RM models. Our results contribute to a better translation of human and macaque antibody and FcR biology, and may help to improve testing accuracy and evaluations of future active and passive prevention strategies.
Collapse
Affiliation(s)
- Justin Pollara
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Matthew Zirui Tay
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - R Whitney Edwards
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Derrick Goodman
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Andrew R Crowley
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Robert J Edwards
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - David Easterhoff
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Haleigh E Conley
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Taylor Hoxie
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thaddeus Gurley
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Caroline Jones
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Emily Machiele
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Marina Tuyishime
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Elizabeth Donahue
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Shalini Jha
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Rachel L Spreng
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kevin Wiehe
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Max M He
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - M Anthony Moody
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Kevin O Saunders
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | | | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Georgia D Tomaras
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
20
|
Rodriguez‐Garcia M, Patel MV, Shen Z, Wira CR. The impact of aging on innate and adaptive immunity in the human female genital tract. Aging Cell 2021; 20:e13361. [PMID: 33951269 PMCID: PMC8135005 DOI: 10.1111/acel.13361] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023] Open
Abstract
Mucosal tissues in the human female reproductive tract (FRT) are primary sites for both gynecological cancers and infections by a spectrum of sexually transmitted pathogens, including human immunodeficiency virus (HIV), that compromise women's health. While the regulation of innate and adaptive immune protection in the FRT by hormonal cyclic changes across the menstrual cycle and pregnancy are being intensely studied, little to nothing is known about the alterations in mucosal immune protection that occur throughout the FRT as women age following menopause. The immune system in the FRT has two key functions: defense against pathogens and reproduction. After menopause, natural reproductive function ends, and therefore, two overlapping processes contribute to alterations in immune protection in aging women: menopause and immunosenescence. The goal of this review is to summarize the multiple immune changes that occur in the FRT with aging, including the impact on the function of epithelial cells, immune cells, and stromal fibroblasts. These studies indicate that major aspects of innate and adaptive immunity in the FRT are compromised in a site‐specific manner in the FRT as women age. Further, at some FRT sites, immunological compensation occurs. Overall, alterations in mucosal immune protection contribute to the increased risk of sexually transmitted infections (STI), urogenital infections, and gynecological cancers. Further studies are essential to provide a foundation for the development of novel therapeutic interventions to restore immune protection and reverse conditions that threaten women's lives as they age.
Collapse
Affiliation(s)
| | - Mickey V. Patel
- Department of Microbiology and Immunology Geisel School of Medicine at Dartmouth Lebanon NH USA
| | - Zheng Shen
- Department of Microbiology and Immunology Geisel School of Medicine at Dartmouth Lebanon NH USA
| | - Charles R. Wira
- Department of Microbiology and Immunology Geisel School of Medicine at Dartmouth Lebanon NH USA
| |
Collapse
|
21
|
Rodriguez-Garcia M, Connors K, Ghosh M. HIV Pathogenesis in the Human Female Reproductive Tract. Curr HIV/AIDS Rep 2021; 18:139-156. [PMID: 33721260 PMCID: PMC9273024 DOI: 10.1007/s11904-021-00546-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Women remain disproportionately affected by the HIV/AIDS pandemic. The primary mechanism for HIV acquisition in women is sexual transmission, yet the immunobiological factors that contribute to HIV susceptibility remain poorly characterized. Here, we review current knowledge on HIV pathogenesis in women, focusing on infection and immune responses in the female reproductive tract (FRT). RECENT FINDINGS We describe recent findings on innate immune protection and HIV target cell distribution in the FRT. We also review multiple factors that modify susceptibility to infection, including sex hormones, microbiome, trauma, and how HIV risk changes during women's life cycle. Finally, we review current strategies for HIV prevention and identify barriers for research in HIV infection and pathogenesis in women. A complex network of interrelated biological and sociocultural factors contributes to HIV risk in women and impairs prevention and cure strategies. Understanding how HIV establishes infection in the FRT can provide clues to develop novel interventions to prevent HIV acquisition in women.
Collapse
Affiliation(s)
- Marta Rodriguez-Garcia
- Department of Immunology, Tufts University School of Medicine, 150 Harrison Ave, Boston, MA, 02111, USA
| | - Kaleigh Connors
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA, 15261, USA
| | - Mimi Ghosh
- Department of Epidemiology, Milken Institute School of Public Health and Health Services, The George Washington University, 800 22nd St NW, Washington, DC, 20052, USA.
| |
Collapse
|
22
|
Chu TH, Crowley AR, Backes I, Chang C, Tay M, Broge T, Tuyishime M, Ferrari G, Seaman MS, Richardson SI, Tomaras GD, Alter G, Leib D, Ackerman ME. Hinge length contributes to the phagocytic activity of HIV-specific IgG1 and IgG3 antibodies. PLoS Pathog 2020; 16:e1008083. [PMID: 32092122 PMCID: PMC7058349 DOI: 10.1371/journal.ppat.1008083] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 03/05/2020] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
Antibody functions such as neutralization require recognition of antigen by the Fab region, while effector functions are additionally mediated by interactions of the Fc region with soluble factors and cellular receptors. The efficacy of individual antibodies varies based on Fab domain characteristics, such as affinity for antigen and epitope-specificity, and on Fc domain characteristics that include isotype, subclass, and glycosylation profile. Here, a series of HIV-specific antibody subclass and hinge variants were constructed and tested to define those properties associated with differential effector function. In the context of the broadly neutralizing CD4 binding site-specific antibody VRC01 and the variable loop (V3) binding antibody 447-52D, hinge truncation and extension had a considerable impact on the magnitude of phagocytic activity of both IgG1 and IgG3 subclasses. The improvement in phagocytic potency of antibodies with extended hinges could not be attributed to changes in either intrinsic antigen or antibody receptor affinity. This effect was specific to phagocytosis and was generalizable to different phagocytes, at different effector cell to target ratios, for target particles of different size and composition, and occurred across a range of antibody concentrations. Antibody dependent cellular cytotoxicity and neutralization were generally independent of hinge length, and complement deposition displayed variable local optima. In vivo stability testing showed that IgG molecules with altered hinges can exhibit similar biodistribution and pharmacokinetic profiles as IgG1. Overall, these results suggest that when high phagocytic activity is desirable, therapeutic antibodies may benefit from being formatted as human IgG3 or engineered IgG1 forms with elongated hinges.
Collapse
Affiliation(s)
- Thach H. Chu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Andrew R. Crowley
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Iara Backes
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Cheryl Chang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Matthew Tay
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Thomas Broge
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Marina Tuyishime
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael S. Seaman
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Simone I. Richardson
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - David Leib
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| |
Collapse
|
23
|
Easterhoff D, Pollara J, Luo K, Janus B, Gohain N, Williams LD, Tay MZ, Monroe A, Peachman K, Choe M, Min S, Lusso P, Zhang P, Go EP, Desaire H, Bonsignori M, Hwang KK, Beck C, Kakalis M, O’Connell RJ, Vasan S, Kim JH, Michael NL, Excler JL, Robb ML, Rerks-Ngarm S, Kaewkungwal J, Pitisuttithum P, Nitayaphan S, Sinangil F, Tartaglia J, Phogat S, Wiehe K, Saunders KO, Montefiori DC, Tomaras GD, Moody MA, Arthos J, Rao M, Joyce MG, Ofek G, Ferrari G, Haynes BF. HIV vaccine delayed boosting increases Env variable region 2-specific antibody effector functions. JCI Insight 2020; 5:131437. [PMID: 31996483 PMCID: PMC7098725 DOI: 10.1172/jci.insight.131437] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/19/2019] [Indexed: 01/07/2023] Open
Abstract
In the RV144 HIV-1 phase III trial, vaccine efficacy directly correlated with the magnitude of the variable region 2-specific (V2-specific) IgG antibody response, and in the presence of low plasma IgA levels, with the magnitude of plasma antibody-dependent cellular cytotoxicity. Reenrollment of RV144 vaccinees in the RV305 trial offered the opportunity to define the function, maturation, and persistence of vaccine-induced V2-specific and other mAb responses after boosting. We show that the RV144 vaccine regimen induced persistent V2 and other HIV-1 envelope-specific memory B cell clonal lineages that could be identified throughout the approximately 11-year vaccination period. Subsequent boosts increased somatic hypermutation, a critical requirement for antibody affinity maturation. Characterization of 22 vaccine-induced V2-specific mAbs with epitope specificities distinct from previously characterized RV144 V2-specific mAbs CH58 and CH59 found increased in vitro antibody-mediated effector functions. Thus, when inducing non-neutralizing antibodies, one method by which to improve HIV-1 vaccine efficacy may be through late boosting to diversify the V2-specific response to increase the breadth of antibody-mediated anti-HIV-1 effector functions.
Collapse
Affiliation(s)
- David Easterhoff
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Medicine and
| | | | - Kan Luo
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Benjamin Janus
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Neelakshi Gohain
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Matthew Zirui Tay
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Anthony Monroe
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Kristina Peachman
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Misook Choe
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Susie Min
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Paolo Lusso
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Peng Zhang
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Eden P. Go
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Heather Desaire
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mattia Bonsignori
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Medicine and
| | - Kwan-Ki Hwang
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Charles Beck
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Matina Kakalis
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | | | - Sandhya Vasan
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Department of Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Jerome H. Kim
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
| | - Nelson L. Michael
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jean-Louis Excler
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Merlin L. Robb
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Supachai Rerks-Ngarm
- US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | | | - Punnee Pitisuttithum
- Mahidol Bangkok School of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sorachai Nitayaphan
- Mahidol Bangkok School of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - James Tartaglia
- Global Solutions for Infectious Diseases, South San Francisco, California, USA
| | - Sanjay Phogat
- Global Solutions for Infectious Diseases, South San Francisco, California, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Medicine and
| | | | | | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - James Arthos
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Mangala Rao
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
| | - M. Gordon Joyce
- Department of Cell Biology and Molecular Genetics, College of Computational, Biological, and Natural Sciences, and Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Gilad Ofek
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Medicine and
| |
Collapse
|
24
|
Hoang T, Toler E, DeLong K, Mafunda NA, Bloom SM, Zierden HC, Moench TR, Coleman JS, Hanes J, Kwon DS, Lai SK, Cone RA, Ensign LM. The cervicovaginal mucus barrier to HIV-1 is diminished in bacterial vaginosis. PLoS Pathog 2020; 16:e1008236. [PMID: 31971984 PMCID: PMC6999914 DOI: 10.1371/journal.ppat.1008236] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 02/04/2020] [Accepted: 11/25/2019] [Indexed: 11/19/2022] Open
Abstract
Bacterial vaginosis (BV), a condition in which the vaginal microbiota consists of community of obligate and facultative anaerobes rather than dominated by a single species of Lactobacillus, affects ~30% of women in the US. Women with BV are at 60% increased risk for HIV acquisition and are 3-times more likely to transmit HIV to an uninfected partner. As cervicovaginal mucus (CVM) is the first line of defense against mucosal pathogens and the home of the resident vaginal microbiota, we hypothesized the barrier function of CVM to HIV may be diminished in BV. Here, we characterized CVM properties including pH, lactic acid content, and Nugent score to correlate with the microbiota community composition, which was confirmed by 16S rDNA sequencing on a subset of samples. We then quantified the mobility of fluorescently-labeled HIV virions and nanoparticles to characterize the structural and adhesive barrier properties of CVM. Our analyses included women with Nugent scores categorized as intermediate (4–6) and BV (7–10), women that were either symptomatic or asymptomatic, and a small group of women before and after antibiotic treatment for symptomatic BV. Overall, we found that HIV virions had significantly increased mobility in CVM from women with BV compared to CVM from women with Lactobacillus crispatus-dominant microbiota, regardless of whether symptoms were present. We confirmed using nanoparticles and scanning electron microscopy that the impaired barrier function was due to reduced adhesive barrier properties without an obvious degradation of the physical CVM pore structure. We further confirmed a similar increase in HIV mobility in CVM from women with Lactobacillus iners-dominant microbiota, the species most associated with transitions to BV and that persists after antibiotic treatment for BV. Our findings advance the understanding of the protective role of mucus and highlight the interplay between vaginal microbiota and the innate barrier function mucus. Bacterial vaginosis (BV), a condition characterized by the depletion of lactobacillus bacteria in the vagina, is the most common vaginal condition in reproductive age women. BV has been associated with many adverse reproductive and sexual health outcomes, including increased risk of HIV infection. Cervicovaginal mucus is the home to vaginal bacteria and acts as a first line of defense to protect the underlying tissues and cells from infection. Here, we studied the barrier properties of mucus from women with BV compared to women with vaginal bacteria dominated by lactobacilli. We found that mucus from women with BV and women with Lactobacillus iners were permissive to HIV-1, which may allow the virus to more easily reach target cells. These findings are in agreement with the observed increased risk for HIV acquisition seen in women with BV and L. iners bacteria. Furthermore, we found that the barrier against HIV is diminished in women with BV regardless of whether they have symptoms. Our findings highlight the important, yet unexplored interactions between the mucus barrier and the vaginal microbiota and the implications for human health.
Collapse
Affiliation(s)
- Thuy Hoang
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Emily Toler
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kevin DeLong
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nomfuneko A. Mafunda
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
| | - Seth M. Bloom
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hannah C. Zierden
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Thomas R. Moench
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jenell S. Coleman
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Justin Hanes
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Douglas S. Kwon
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC/NCSU Joint Department of Biomedical Engineering, Department of Microbiology & Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard A. Cone
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Laura M. Ensign
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
25
|
Iqbal Z, Dilnawaz F. Nanocarriers For Vaginal Drug Delivery. ACTA ACUST UNITED AC 2020; 13:3-15. [PMID: 30767755 DOI: 10.2174/1872211313666190215141507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Vaginal drug delivery approach represents one of the imperative strategies for local and systemic delivery of drugs. The peculiar dense vascular networks, mucus permeability, and range of physiological characteristics of the vaginal cavity have been exploited for therapeutic benefit. Furthermore, the vaginal drug delivery has been curtailed due to the influence of different physiological factors like acidic pH, constant cervical secretion, microflora, cyclic changes during periods along with turnover of mucus of varying thickness. OBJECTIVE This review highlights advancement of nanomedicine and its prospective progress towards the clinic. METHODS Relevant literature reports and patents related to topics are retrieved and used. RESULT The extensive literature search and patent revealed that nanocarriers are efficacious over conventional treatment approaches. CONCLUSION Recently, nanotechnology based drug delivery approach has promised better therapeutic outcomes by providing enhanced permeation and sustained drug release activity. Different nanoplatforms based on drugs, peptides, proteins, antigens, hormones, nucleic material, and microbicides are gaining momentum for vaginal therapeutics.
Collapse
Affiliation(s)
- Zeenat Iqbal
- Nanomedicine Laboratory, Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi-110062, India
| | - Fahima Dilnawaz
- Laboratory of Nanomedicine, Institute of Life Sciences, Nalco Square, Bhubaneswar -751023, Odisha, India
| |
Collapse
|
26
|
Berkebile AR, Bartlett JA, Abou Alaiwa M, Varga SM, Power UF, McCray PB. Airway Surface Liquid Has Innate Antiviral Activity That Is Reduced in Cystic Fibrosis. Am J Respir Cell Mol Biol 2020; 62:104-111. [PMID: 31242392 PMCID: PMC6938132 DOI: 10.1165/rcmb.2018-0304oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 06/26/2019] [Indexed: 12/30/2022] Open
Abstract
Although chronic bacterial infections and inflammation are associated with progressive lung disease in patients with cystic fibrosis (CF), much less is known regarding the contributions of respiratory viral infections to this process. Clinical studies suggest that antiviral host defenses may be compromised in individuals with CF, and CF airway epithelia exhibit impaired antiviral responses in vitro. Here, we used the CF pig model to test the hypothesis that the antiviral activity of respiratory secretions is reduced in CF. We developed an in vitro assay to measure the innate antiviral activity present in airway surface liquid (ASL) from CF and non-CF pigs. We found that tracheal and nasal ASL from newborn non-CF pigs exhibited dose-dependent inhibitory activity against several enveloped and encapsidated viruses, including Sendai virus, respiratory syncytial virus, influenza A, and adenovirus. Importantly, we found that the anti-Sendai virus activity of nasal ASL from newborn CF pigs was significantly diminished relative to non-CF littermate controls. This diminution of extracellular antiviral defenses appears to be driven, at least in part, by the differences in pH between CF and non-CF ASL. These data highlight the novel antiviral properties of native airway secretions and suggest the possibility that defects in extracellular antiviral defenses contribute to CF pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Steven M. Varga
- Department of Microbiology and Immunology
- Department of Pathology, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Ultan F. Power
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Paul B. McCray
- Department of Microbiology and Immunology
- Department of Pediatrics
| |
Collapse
|
27
|
Natural polymers for vaginal mucoadhesive delivery of vinegar, using design of experiment methods. VOJNOSANIT PREGL 2020. [DOI: 10.2298/vsp200804121p] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background/Aim. Vinegars are of the main international traditional
nutraceuticals which have been used as vaginal health protectant due to
vagina pH balance maintenance and antimicrobial properties. Since the main
used form of vinegar was liquid, it was difficult for vaginal application
with low residence time; in this study a vaginal mucoadhesive gel of vinegar
was designed. Methods. Xanthan gum (XG) and tragacanth (TG) were utilized as
natural gel forming polymers. The effects of Xanthan gum and tragacanth on
mucoadhesion strength and drug release of the gel formulations were
optimized using a 3 level (32) factorial design. Several physico-chemical
properties of the gel formulations including gel viscosity, spreadability,
scanning electron microscopy (SEM) images of hydrogel chains, and release
kinetic were also investigated. Results. demonstrated that tragacanth
possesses a statistically significant effect on release rate control
(p-value=0.0027) while both tragacanth and xanthan gum have significant
effect (p value= 0.0001 and 0.0017, respectively) on mucoadhesion property.
Conclusion. Design of experiment suggested that formulation F7 with 5%
xanthan gum and 1% tragacanth (mucoadhesion = 0.4632 N and release rate =
88.8% in 6 hours) can be considered as the optimum formulation with some
modifications.
Collapse
|
28
|
Dude CM, Saylany A, Brown A, Elovitz M, Anton L. Microbial supernatants from Mobiluncus mulieris, a bacteria strongly associated with spontaneous preterm birth, disrupts the cervical epithelial barrier through inflammatory and miRNA mediated mechanisms. Anaerobe 2019; 61:102127. [PMID: 31760081 DOI: 10.1016/j.anaerobe.2019.102127] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 11/17/2022]
Abstract
Recent human clinical studies have identified Mobiluncus mulieris, a fastidious strict anaerobic bacterium present in the cervicovaginal (CV) space, as being strongly associated with spontaneous preterm birth (sPTB). However, the molecular mechanisms that underlie this association remain unknown. As disruption of the cervical epithelial barrier has been shown to contribute to the premature cervical remodeling that precedes sPTB, we hypothesize that M. mulieris, a microbe strongly associated with sPTB in humans, has the ability to alter cervical epithelial function. We investigated if bacteria-free supernatants of M. mulieris were able to disrupt the cervical epithelial barrier through immunological and epigenetic based mechanisms in an in vitro model system. Ectocervical cells were treated with supernatant from cultured M. mulieris and epithelial cell permeability, immune cytokines and microRNAs (miRNAs) were investigated. M. mulieris supernatant significantly increased cell permeability and the expression of two inflammatory mediators associated with cervical epithelial breakdown, IL-6 and IL-8. Moreover, treatment of the ectocervical cells with the M. mulieris supernatant also increased the expression of miRNAs that have been associated with either sPTB or a shorter gestational length in humans. Collectively, these results suggest that M. mulieris induces molecular and functional changes in the cervical epithelial barrier thought to contribute to the pathogenesis of sPTB, which allows us to hypothesize that targeting CV bacteria such as M. mulieris could provide a therapeutic opportunity to reduce sPTB rates.
Collapse
Affiliation(s)
- Carolynn M Dude
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Anissa Saylany
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Brown
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michal Elovitz
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren Anton
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
Mishra NN, Agarwal A, Moitra T, Polachira SK, Nair R, Gupta SK. Anti-HIV-1 activity and safety profile of a polyherbal gel formulation as a candidate microbicide. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
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
|
31
|
Leyva-Gómez G, Prado-Audelo MLD, Ortega-Peña S, Mendoza-Muñoz N, Urbán-Morlán Z, González-Torres M, González-Del Carmen M, Figueroa-González G, Reyes-Hernández OD, Cortés H. Modifications in Vaginal Microbiota and Their Influence on Drug Release: Challenges and Opportunities. Pharmaceutics 2019; 11:pharmaceutics11050217. [PMID: 31064154 PMCID: PMC6571606 DOI: 10.3390/pharmaceutics11050217] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Vaginal drug delivery represents an attractive alternative to achieve local and systemic effects due to the high contact surface exposed, the mucoadhesion of the epithelium, and the high innervation that facilitates the absorption of drugs into the bloodstream. However, despite the confinement of the vaginal cavity, it is an organ with a highly variable microenvironment. Mechanical alterations such as coitus, or chemical changes such as pH and viscosity, modify the release of drugs. In addition, changes in vaginal microbiota can influence the entire vaginal microenvironment, thus determining the disposition of drugs in the vaginal cavity and decreasing their therapeutic efficacy. Therefore, the influence of microorganisms on vaginal homeostasis can change the pre-established scenario for the application of drugs. This review aims to provide an explanation of normal vaginal microbiota, the factors that modify it, its involvement in the administration of drugs, and new proposals for the design of novel pharmaceutical dosage forms. Finally, challenges and opportunities directed toward the conception of new effective formulations are discussed.
Collapse
Affiliation(s)
- Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - María L Del Prado-Audelo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
- Laboratorio de Posgrado en Tecnología Farmacéutica, FES-Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico.
| | - Silvestre Ortega-Peña
- Laboratorio de Infectología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico.
| | | | - Zaida Urbán-Morlán
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Maykel González-Torres
- CONACyT-Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico.
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Campus Ciudad de México 14380, Mexico.
| | | | - Gabriela Figueroa-González
- CONACyT-Laboratorio de Genómica, Dirección de Investigación, Instituto Nacional de Cancerología. Av. San Fernando 22, Tlalpan, Sección XVI, 14080 Ciudad de México, Mexico.
| | - Octavio D Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico.
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico.
| |
Collapse
|
32
|
Bridging Vaccine-Induced HIV-1 Neutralizing and Effector Antibody Responses in Rabbit and Rhesus Macaque Animal Models. J Virol 2019; 93:JVI.02119-18. [PMID: 30842326 PMCID: PMC6498063 DOI: 10.1128/jvi.02119-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
Nonneutralizing antibody functions have been associated with reduced infection risk, or control of virus replication, for HIV-1 and related viruses. It is therefore critical to evaluate development of these responses throughout all stages of preclinical testing. Rabbits are conventionally used to evaluate the ability of vaccine candidates to safely elicit antibodies that bind and neutralize HIV-1. However, it remained unexplored how effectively rabbits model the development of nonneutralizing antibody responses in primates. We administered identical HIV-1 vaccine regimens to rabbits and rhesus macaques and performed detailed comparisons of vaccine-induced antibody responses. We demonstrated that nonneutralizing HIV-specific antibody responses can be studied in the rabbit model and have identified aspects of these responses that are common, and those that are unique, to rabbits and rhesus macaques. Our findings will help determine how to best utilize preclinical rabbit and rhesus macaque models to accelerate HIV vaccine candidate testing in human trials. Studies in animal models are essential prerequisites for clinical trials of candidate HIV vaccines. Small animals, such as rabbits, are used to evaluate promising strategies prior to further immunogenicity and efficacy testing in nonhuman primates. Our goal was to determine how HIV-specific vaccine-elicited antibody responses, epitope specificity, and Fc-mediated functions in the rabbit model can predict those in the rhesus macaque (RM) model. Detailed comparisons of the HIV-1-specific IgG response were performed on serum from rabbits and RM given identical modified vaccinia virus Ankara-prime/gp120-boost immunization regimens. We found that vaccine-induced neutralizing antibody, gp120-binding antibody levels and immunodominant specificities, antibody-dependent cellular phagocytosis of HIV-1 virions, and antibody-dependent cellular cytotoxicity (ADCC) responses against gp120-coated target cells were similar in rabbits and RM. However, we also identified characteristics of humoral immunity that differed across species. ADCC against HIV-infected target cells was elicited in rabbits but not in RM, and we observed differences among subdominantly targeted epitopes. Human Fc receptor binding assays and analysis of antibody-cell interactions indicated that rabbit vaccine-induced antibodies effectively recruited and activated human natural killer cells, while vaccine-elicited RM antibodies were unable to activate either human or RM NK cells. Thus, our data demonstrate that both Fc-independent and Fc-dependent functions of rabbit antibodies can be measured with commonly used in vitro assays; however, the ability of immunogenicity studies performed in rabbits to predict responses in RM will vary depending on the particular immune parameter of interest. IMPORTANCE Nonneutralizing antibody functions have been associated with reduced infection risk, or control of virus replication, for HIV-1 and related viruses. It is therefore critical to evaluate development of these responses throughout all stages of preclinical testing. Rabbits are conventionally used to evaluate the ability of vaccine candidates to safely elicit antibodies that bind and neutralize HIV-1. However, it remained unexplored how effectively rabbits model the development of nonneutralizing antibody responses in primates. We administered identical HIV-1 vaccine regimens to rabbits and rhesus macaques and performed detailed comparisons of vaccine-induced antibody responses. We demonstrated that nonneutralizing HIV-specific antibody responses can be studied in the rabbit model and have identified aspects of these responses that are common, and those that are unique, to rabbits and rhesus macaques. Our findings will help determine how to best utilize preclinical rabbit and rhesus macaque models to accelerate HIV vaccine candidate testing in human trials.
Collapse
|
33
|
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
|
34
|
Tay MZ, Kunz EL, Deal A, Zhang L, Seaton KE, Rountree W, Eudailey JA, Heptinstall J, McRaven MD, Matias E, McGuire E, Yates NL, Perez LG, Montefiori DC, Overman RG, Hope TJ, Shen X, Kalilani L, Fouda GG, Tomaras GD, Permar SR. Rare Detection of Antiviral Functions of Polyclonal IgA Isolated from Plasma and Breast Milk Compartments in Women Chronically Infected with HIV-1. J Virol 2019; 93:e02084-18. [PMID: 30700599 PMCID: PMC6430545 DOI: 10.1128/jvi.02084-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
The humoral response to invading mucosal pathogens comprises multiple antibody isotypes derived from systemic and mucosal compartments. To understand the contribution of each antibody isotype/source to the mucosal humoral response, parallel investigation of the specificities and functions of antibodies within and across isotypes and compartments is required. The role of IgA against HIV-1 is complex, with studies supporting a protective role as well as a role for serum IgA in blocking effector functions. Thus, we explored the fine specificity and function of IgA in both plasma and mucosal secretions important to infant HIV-1 infection, i.e., breast milk. IgA and IgG were isolated from milk and plasma from 20 HIV-1-infected lactating Malawian women. HIV-1 binding specificities, neutralization potency, inhibition of virus-epithelial cell binding, and antibody-mediated phagocytosis were measured. Fine-specificity mapping showed IgA and IgG responses to multiple HIV-1 Env epitopes, including conformational V1/V2 and linear V2, V3, and constant region 5 (C5). Env IgA was heterogeneous between the milk and systemic compartments (Env IgA, τ = 0.00 to 0.63, P = 0.0046 to 1.00). Furthermore, IgA and IgG appeared compartmentalized as there was a lack of correlation between the specificities of Env-specific IgA and IgG (in milk, τ = -0.07 to 0.26, P = 0.35 to 0.83). IgA and IgG also differed in functions: while neutralization and phagocytosis were consistently mediated by milk and plasma IgG, they were rarely detected in IgA from both milk and plasma. Understanding the ontogeny of the divergent IgG and IgA antigen specificity repertoires and their effects on antibody function will inform vaccination approaches targeted toward mucosal pathogens.IMPORTANCE Antibodies within the mucosa are part of the first line of defense against mucosal pathogens. Evaluating mucosal antibody isotypes, specificities, and antiviral functions in relationship to the systemic antibody profile can provide insights into whether the antibody response is coordinated in response to mucosal pathogens. In a natural immunity cohort of HIV-infected lactating women, we mapped the fine specificity and function of IgA in breast milk and plasma and compared these with the autologous IgG responses. Antigen specificities and functions differed between IgG and IgA, with antiviral functions (neutralization and phagocytosis) predominantly mediated by the IgG fraction in both milk and plasma. Furthermore, the specificity of milk IgA differed from that of systemic IgA. Our data suggest that milk IgA and systemic IgA should be separately examined as potential correlates of risk. Preventive vaccines may need to employ different strategies to elicit functional antiviral immunity by both antibody isotypes in the mucosa.
Collapse
Affiliation(s)
- Matthew Zirui Tay
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Erika L Kunz
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Aaron Deal
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Lu Zhang
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Kelly E Seaton
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Joshua A Eudailey
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jack Heptinstall
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Michael D McRaven
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edgar Matias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Erin McGuire
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Nicole L Yates
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Lautaro G Perez
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - R Glenn Overman
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Thomas J Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Linda Kalilani
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Genevieve G Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Immunology, Duke University, Durham, North Carolina, USA
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| |
Collapse
|
35
|
Calenda G, Villegas G, Reis A, Millen L, Barnable P, Mamkina L, Kumar N, Roberts K, Kalir T, Martinelli E, Sperling R, Teleshova N. Mucosal Susceptibility to Human Immunodeficiency Virus Infection in the Proliferative and Secretory Phases of the Menstrual Cycle. AIDS Res Hum Retroviruses 2019; 35:335-347. [PMID: 30600686 DOI: 10.1089/aid.2018.0154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Factors underlying HIV acquisition in women remain incompletely understood. This study evaluated ex vivo mucosal HIV-1BaL infection (ectocervix, endocervix), T cell frequencies and phenotype (ectocervix, endocervix, peripheral blood), and HIV-1BaL-induced tissue immune responses (ectocervix) in the proliferative and secretory phases of the menstrual cycle using samples obtained from women undergoing hysterectomies. Tissue infectivity (number of productively infected explants) and infection level following 500 and/or fifty 50% tissue culture infectious dose (TCID50) HIV-1BaL challenge were similar in the proliferative and secretory phases. Although not associated with infection outcomes, higher frequencies of HIV target CD4+α4β7+ T cells, and stronger HIV-1BaL-induced proinflammatory responses were detected in ectocervix in the secretory versus proliferative phase. Independently of the cycle phase, serum E2 concentrations were inversely associated with ectocervical and endocervical tissue infection levels following high-dose 500 TCID50 HIV-1BaL challenge, with frequencies of CD4+α4β7+ T cells in endocervix, and with HIV-induced interleukin (IL)2R and IL4 in ectocervix. Although serum P4 concentrations and P4/E2 ratios were neither associated with tissue infection level nor infectivity, high P4 concentrations and/or P4/E2 ratios correlated with high frequencies of CD4+α4β7+ T cells in ectocervix, low frequencies of CD4+CD103+ blood T cells, low CD4+LFA-1+ T cells in endocervix, and high proinflammatory (IL1β, IL17, tumor necrosis factor α) ectocervical tissue responses to HIV-1BaL. The data suggest an inhibitory effect of E2 on mucosal HIV infection, provide insights into potential mechanisms of E2-mediated anti-HIV activity, and highlight P4-associated immune changes in the mucosa.
Collapse
Affiliation(s)
- Giulia Calenda
- Center for Biomedical Research, Population Council, New York, New York
| | | | - Alexandra Reis
- Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Lily Millen
- Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Patrick Barnable
- Center for Biomedical Research, Population Council, New York, New York
| | - Lisa Mamkina
- Center for Biomedical Research, Population Council, New York, New York
| | - Narender Kumar
- Center for Biomedical Research, Population Council, New York, New York
| | - Kevin Roberts
- Center for Biomedical Research, Population Council, New York, New York
| | - Tamara Kalir
- Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, New York, New York
| | - Rhoda Sperling
- Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Natalia Teleshova
- Center for Biomedical Research, Population Council, New York, New York
| |
Collapse
|
36
|
Tay MZ, Wiehe K, Pollara J. Antibody-Dependent Cellular Phagocytosis in Antiviral Immune Responses. Front Immunol 2019; 10:332. [PMID: 30873178 PMCID: PMC6404786 DOI: 10.3389/fimmu.2019.00332] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
Antiviral activities of antibodies may either be dependent only on interactions between the antibody and cognate antigen, as in binding and neutralization of an infectious virion, or instead may require interactions between antibody-antigen immune complexes and immunoproteins or Fc receptor expressing immune effector cells. These Fc receptor-dependent antibody functions provide a direct link between the innate and adaptive immune systems by combining the potent antiviral activity of innate effector cells with the diversity and specificity of the adaptive humoral response. The Fc receptor-dependent function of antibody-dependent cellular phagocytosis (ADCP) provides mechanisms for clearance of virus and virus-infected cells, as well as for stimulation of downstream adaptive immune responses by facilitating antigen presentation, or by stimulating the secretion of inflammatory mediators. In this review, we discuss the properties of Fc receptors, antibodies, and effector cells that influence ADCP. We also provide and interpret evidence from studies that support a potential role for ADCP in either inhibiting or enhancing viral infection. Finally, we describe current approaches used to measure antiviral ADCP and discuss considerations for the translation of studies performed in animal models. We propose that additional investigation into the role of ADCP in protective viral responses, the specific virus epitopes targeted by ADCP antibodies, and the types of phagocytes and Fc receptors involved in ADCP at sites of virus infection will provide insight into strategies to successfully leverage this important immune response for improved antiviral immunity through rational vaccine design.
Collapse
Affiliation(s)
- Matthew Zirui Tay
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin Wiehe
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Justin Pollara
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
37
|
Koppolu S, Wang L, Mathur A, Nigam JA, Dezzutti CS, Isaacs C, Meyn L, Bunge KE, Moncla BJ, Hillier SL, Rohan LC, Mahal LK. Vaginal Product Formulation Alters the Innate Antiviral Activity and Glycome of Cervicovaginal Fluids with Implications for Viral Susceptibility. ACS Infect Dis 2018; 4:1613-1622. [PMID: 30183260 DOI: 10.1021/acsinfecdis.8b00157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glycosylated proteins (i.e., mucins, IgG) are important mediators of innate antiviral immunity in the vagina; however, our current knowledge of the role that glycan themselves play in genital immunity is relatively low. Herein, we evaluate the relationship between innate antiviral immunity and glycomic composition in cervicovaginal lavage fluid (CVL) collected as part of a Phase I clinical trial testing the impact of two distinct formulations of the antiretroviral drug dapivirine. Using lectin microarray technology, we discovered that formulation (hydrogel- versus film-based delivery) impacted the CVL glycome, with hydrogel formulations inducing more changes, including a loss of high-mannose. The loss of this epitope correlated to a loss of anti-HIV-1 activity. Glycoproteomic identification of high-mannose proteins revealed a cohort of antiproteases shown to be important in HIV-1 resistance, whose expression covaried with the high-mannose signature. Our data strongly suggests high-mannose as a marker for secreted proteins mediating innate antiviral immunity in vaginal fluids and that drug formulation may impact this activity as reflected in the glycome.
Collapse
Affiliation(s)
- Sujeethraj Koppolu
- Biomedical Chemistry Institute, Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| | - Linlin Wang
- Biomedical Chemistry Institute, Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| | - Ayushi Mathur
- Biomedical Chemistry Institute, Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| | - Jayeshwar A. Nigam
- Biomedical Chemistry Institute, Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| | - Charlene S. Dezzutti
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, Pennsylvania 15213, United States
| | - Charles Isaacs
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, New York, New York 10314, United States
| | - Leslie Meyn
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, Pennsylvania 15213, United States
| | - Katherine E. Bunge
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, Pennsylvania 15213, United States
| | - Bernard J. Moncla
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, Pennsylvania 15213, United States
| | - Sharon L. Hillier
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, Pennsylvania 15213, United States
| | - Lisa C. Rohan
- Magee-Womens Research Institute, 204 Craft Avenue, B511, Pittsburgh, Pennsylvania 15213, United States
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Lara K. Mahal
- Biomedical Chemistry Institute, Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003, United States
| |
Collapse
|
38
|
Schroeder HA, Nunn KL, Schaefer A, Henry CE, Lam F, Pauly MH, Whaley KJ, Zeitlin L, Humphrys MS, Ravel J, Lai SK. Herpes simplex virus-binding IgG traps HSV in human cervicovaginal mucus across the menstrual cycle and diverse vaginal microbial composition. Mucosal Immunol 2018; 11:1477-1486. [PMID: 29988116 PMCID: PMC6485947 DOI: 10.1038/s41385-018-0054-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 02/04/2023]
Abstract
IgG possesses an important yet little recognized effector function in mucus. IgG bound to viral surface can immobilize otherwise readily diffusive viruses to the mucin matrix, excluding them from contacting target cells and facilitating their elimination by natural mucus clearance mechanisms. Cervicovaginal mucus (CVM) is populated by a microbial community, and its viscoelastic and barrier properties can vary substantially not only across the menstrual cycle, but also in women with distinct microbiota. How these variations impact the "muco-trapping" effector function of IgGs remains poorly understood. Here we obtained multiple fresh, undiluted CVM specimens (n = 82 unique specimens) from six women over time, and employed high-resolution multiple particle tracking to quantify the mobility of fluorescent Herpes Simplex Viruses (HSV-1) in CVM treated with different HSV-1-binding IgG. The IgG trapping potency was then correlated to the menstrual cycle, and the vaginal microbial composition was determined by 16 s rRNA. In the specimens studied, both polyclonal and monoclonal HSV-1-binding IgG appeared to consistently and effectively trap HSV-1 in CVM obtained at different times of the menstrual cycle and containing a diverse spectrum of commensals, including G. vaginalis-dominant microbiota. Our findings underscore the potential broad utility of this "muco-trapping" effector function of IgG to reinforce the vaginal mucosal defense, and motivates further investigation of passive immunization of the vagina as a strategy to protect against vaginally transmitted infections.
Collapse
Affiliation(s)
- Holly A. Schroeder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA
| | - Kenetta L. Nunn
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA.,UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27519, USA
| | - Alison Schaefer
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA
| | - Christine E. Henry
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA
| | - Felix Lam
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA
| | | | | | - Larry Zeitlin
- Mapp Biopharmaceutical Inc., San Diego, CA, 92121, USA
| | - Mike S. Humphrys
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jacques Ravel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27519, USA.,UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27519, USA
| |
Collapse
|
39
|
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: 64] [Impact Index Per Article: 9.1] [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
|
40
|
Tyssen D, Wang YY, Hayward JA, Agius PA, DeLong K, Aldunate M, Ravel J, Moench TR, Cone RA, Tachedjian G. Anti-HIV-1 Activity of Lactic Acid in Human Cervicovaginal Fluid. mSphere 2018; 3:e00055-18. [PMID: 29976641 PMCID: PMC6034077 DOI: 10.1128/msphere.00055-18] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/05/2018] [Indexed: 12/14/2022] Open
Abstract
Women of reproductive age with a Lactobacillus-dominated vaginal microbiota have a reduced risk of acquiring and transmitting HIV and a vaginal pH of ~4 due to the presence of ~1% (wt/vol) lactic acid. While lactic acid has potent HIV virucidal activity in vitro, whether lactic acid present in the vaginal lumen inactivates HIV has not been investigated. Here we evaluated the anti-HIV-1 activity of native, minimally diluted cervicovaginal fluid obtained from women of reproductive age (n = 20) with vaginal microbiota dominated by Lactobacillus spp. Inhibition of HIVBa-L was significantly associated with the protonated form of lactic acid in cervicovaginal fluid. The HIVBa-L inhibitory activity observed in the <3-kDa acidic filtrate was similar to that of the corresponding untreated native cervicovaginal fluid as well as that of clarified neat cervicovaginal fluid subjected to protease digestion. These ex vivo studies indicate that protonated lactic acid is a major anti-HIV-1 metabolite present in acidic cervicovaginal fluid, suggesting a potential role in reducing HIV transmission by inactivating virus introduced or shed into the cervicovaginal lumen.IMPORTANCE The Lactobacillus-dominated vaginal microbiota is associated with a reduced risk of acquiring and transmitting HIV and other sexually transmitted infections (STIs). Lactic acid is a major organic acid metabolite produced by lactobacilli that acidifies the vagina and has been reported to have inhibitory activity in vitro against bacterial, protozoan, and viral STIs, including HIV infections. However, the anti-HIV properties of lactic acid in native vaginal lumen fluids of women colonized with Lactobacillus spp. have not yet been established. Our study, using native cervicovaginal fluid from women, found that potent and irreversible anti-HIV-1 activity is significantly associated with the concentration of the protonated (acidic, uncharged) form of lactic acid. This work advances our understanding of the mechanisms by which vaginal microbiota modulate HIV susceptibility and could lead to novel strategies to prevent women from acquiring HIV or transmitting the virus during vaginal intercourse and vaginal birth.
Collapse
Affiliation(s)
- David Tyssen
- Disease Elimination Program, Life Sciences Discipline, Burnet Institute, Melbourne, Victoria, Australia
| | - Ying-Ying Wang
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joshua A Hayward
- Disease Elimination Program, Life Sciences Discipline, Burnet Institute, Melbourne, Victoria, Australia
| | - Paul A Agius
- Maternal and Child Health Program, Public Health Discipline, Burnet Institute, Melbourne, Victoria, Australia
- Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Victoria, Australia
| | - Kevin DeLong
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Muriel Aldunate
- Disease Elimination Program, Life Sciences Discipline, Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Richard A Cone
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gilda Tachedjian
- Disease Elimination Program, Life Sciences Discipline, Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
41
|
Berard AR, Perner M, Mutch S, Farr Zuend C, McQueen P, Burgener AD. Understanding mucosal and microbial functionality of the female reproductive tract by metaproteomics: Implications for HIV transmission. Am J Reprod Immunol 2018; 80:e12977. [PMID: 29790240 DOI: 10.1111/aji.12977] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 04/18/2018] [Indexed: 12/25/2022] Open
Abstract
The mucosal surface of the female genital tract contains physiological, immunological, and microbial components that collectively comprise a functioning "mucosal system" that is critical for reproductive health. Alterations or imbalances to any of these components can have significant consequences for susceptibility to sexually transmitted infections, such as HIV. In recent years the advent of advanced systems biology technologies, such as metaproteomics, has provided new toolsets to studying mucosal systems. Studies have linked an altered mucosal proteome to many HIV risk factors including mucosal inflammation, bacterial vaginosis, hormonal contraceptives, and reduced efficacy of antiretroviral drugs for HIV prevention. Herein we will discuss how metaproteomics has been used to study mucosal system components, including epithelial barriers, inflammation, and the microbiome, with a focus on what alterations may contribute to increased HIV transmission risk in women.
Collapse
Affiliation(s)
- Alicia R Berard
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada
| | - Michelle Perner
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada
| | - Sarah Mutch
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada
| | - Christina Farr Zuend
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada
| | - Peter McQueen
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada
| | - Adam D Burgener
- National HIV and Retrovirology Labs, JCWilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,University of Manitoba, Winnipeg, MB, Canada.,Karolinska Institutet, Solna, Sweden
| |
Collapse
|
42
|
Optimising the collection of female genital tract fluid for cytokine analysis in pregnant women. J Immunol Methods 2018; 458:15-20. [PMID: 29625077 PMCID: PMC5981004 DOI: 10.1016/j.jim.2018.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/19/2018] [Accepted: 03/28/2018] [Indexed: 02/08/2023]
Abstract
Introduction To better understand the immunology of pregnancy, study of female genital tract fluid (FGF) is desirable. However the optimum method of collection of FGF in pregnant women for immunological methods, specifically cytokine measurement, is unknown. Methods A prospective study of HIV-uninfected pregnant women comparing two methods of FGF collection: polyvinyl acetal sponge collection of cervical fluid (CF) and menstrual cup collection of cervicovaginal fluid (CVF). Samples were collected at 3 time points across the second and third trimesters: 14–21, 22–25 and 26–31 weeks. Multiplex chemi-luminescent assays were used to measure: IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-13 and TNF-α. Optimal methodology for cytokine normalisation (sample weight, volume and total protein) was explored. Results All cytokines were measurable in both fluid types. IL-1β, IL-8 and IL-6 were detected at the highest concentrations (ranking order CF > CVF > plasma). CVF collection was simpler, provided the largest volume of sample (median 0.5 g) with the potential for undiluted usage, and allowed for self-insertion. CF cytokine concentrations were intrinsically associated with sample weight and protein concentration however CVF cytokines were independent of these. Conclusion Both methods of collection are robust for measurement of FGF cytokines during pregnancy. We recommend CVF collection using a menstrual cup as a viable option in pregnant women for high dimensional biological techniques. PVA sponges and MCs are robust methods for measuring FGF cytokines in pregnancy. MCs enable collection of large undiluted CVF volumes for high dimensional assays. CVF (not CF) cytokine concentrations are largely independent of sample weight or protein. Self-insertion and short collection time of MCs is an attractive option to women. Cytokine concentrations are higher in sponge samples reflecting the CF immune site.
Collapse
|
43
|
HIV-1-Specific IgA Monoclonal Antibodies from an HIV-1 Vaccinee Mediate Galactosylceramide Blocking and Phagocytosis. J Virol 2018; 92:JVI.01552-17. [PMID: 29321320 PMCID: PMC5972890 DOI: 10.1128/jvi.01552-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/03/2017] [Indexed: 02/01/2023] Open
Abstract
Vaccine-elicited humoral immune responses comprise an array of antibody forms and specificities, with only a fraction contributing to protective host immunity. Elucidation of antibody effector functions responsible for protective immunity against human immunodeficiency virus type 1 (HIV-1) acquisition is a major goal for the HIV-1 vaccine field. Immunoglobulin A (IgA) is an important part of the host defense against pathogens; however, little is known about the role of vaccine-elicited IgA and its capacity to mediate antiviral functions. To identify the antiviral functions of HIV-1-specific IgA elicited by vaccination, we cloned HIV-1 envelope-specific IgA monoclonal antibodies (MAbs) by memory B cell cultures from peripheral blood mononuclear cells from an RV144 vaccinee and produced two IgA clonal cell lines (HG129 and HG130) producing native, nonrecombinant IgA MAbs. The HG129 and HG130 MAbs mediated phagocytosis by monocytes, and HG129 blocked HIV-1 Env glycoprotein binding to galactosylceramide, an alternative HIV-1 receptor. These findings elucidate potential antiviral functions of vaccine-elicited HIV-1 envelope-specific IgA that may act to block HIV-1 acquisition at the portal of entry by preventing HIV-1 binding to galactosylceramide and mediating antibody Fc receptor-mediated virion phagocytosis. Furthermore, these findings highlight the complex and diverse interactions of vaccine-elicited IgA with pathogens that depend on IgA fine specificity and form (e.g., multimeric or monomeric) in the systemic circulation and mucosal compartments. IMPORTANCE Host-pathogen interactions in vivo involve numerous immune mechanisms that can lead to pathogen clearance. Understanding the nature of antiviral immune mechanisms can inform the design of efficacious HIV-1 vaccine strategies. Evidence suggests that both neutralizing and nonneutralizing antibodies can mediate some protection against HIV in animal models. Although numerous studies have characterized the functional properties of HIV-1-specific IgG, more studies are needed on the functional attributes of HIV-1-specific IgA, specifically for vaccine-elicited IgA. Characterization of the functional properties of HIV-1 Env-specific IgA monoclonal antibodies from human vaccine clinical trials are critical toward understanding the capacity of the host immune response to block HIV-1 acquisition.
Collapse
|
44
|
Passmore JAS, Jaspan HB. Vaginal microbes, inflammation, and HIV risk in African women. THE LANCET. INFECTIOUS DISEASES 2018; 18:483-484. [PMID: 29396005 DOI: 10.1016/s1473-3099(18)30061-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/09/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Jo-Ann S Passmore
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town Medical School, Cape Town 7925, South Africa; NRF-DST CAPRISA Centre of Excellence, Cape Town, South Africa; National Health Laboratory Service, Cape Town, South Africa.
| | - Heather B Jaspan
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town Medical School, Cape Town 7925, South Africa; Seattle Children's Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
45
|
Angstmann CN, Henry BI, McGann AV. Generalized fractional diffusion equations for subdiffusion in arbitrarily growing domains. Phys Rev E 2018; 96:042153. [PMID: 29347596 DOI: 10.1103/physreve.96.042153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Indexed: 11/07/2022]
Abstract
The ubiquity of subdiffusive transport in physical and biological systems has led to intensive efforts to provide robust theoretical models for this phenomena. These models often involve fractional derivatives. The important physical extension of this work to processes occurring in growing materials has proven highly nontrivial. Here we derive evolution equations for modeling subdiffusive transport in a growing medium. The derivation is based on a continuous-time random walk. The concise formulation of these evolution equations requires the introduction of a new, comoving, fractional derivative. The implementation of the evolution equation is illustrated with a simple model of subdiffusing proteins in a growing membrane.
Collapse
Affiliation(s)
- C N Angstmann
- School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - B I Henry
- School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - A V McGann
- School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
46
|
Klein K, Nickel G, Nankya I, Kyeyune F, Demers K, Ndashimye E, Kwok C, Chen PL, Rwambuya S, Poon A, Munjoma M, Chipato T, Byamugisha J, Mugyenyi P, Salata RA, Morrison CS, Arts EJ. Higher sequence diversity in the vaginal tract than in blood at early HIV-1 infection. PLoS Pathog 2018; 14:e1006754. [PMID: 29346424 PMCID: PMC5773221 DOI: 10.1371/journal.ppat.1006754] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
In the majority of cases, human immunodeficiency virus type 1 (HIV-1) infection is transmitted through sexual intercourse. A single founder virus in the blood of the newly infected donor emerges from a genetic bottleneck, while in rarer instances multiple viruses are responsible for systemic infection. We sought to characterize the sequence diversity at early infection, between two distinct anatomical sites; the female reproductive tract vs. systemic compartment. We recruited 72 women from Uganda and Zimbabwe within seven months of HIV-1 infection. Using next generation deep sequencing, we analyzed the total genetic diversity within the C2-V3-C3 envelope region of HIV-1 isolated from the female genital tract at early infection and compared this to the diversity of HIV-1 in plasma. We then compared intra-patient viral diversity in matched cervical and blood samples with three or seven months post infection. Genetic analysis of the C2-V3-C3 region of HIV-1 env revealed that early HIV-1 isolates within blood displayed a more homogeneous genotype (mean 1.67 clones, range 1–5 clones) than clones in the female genital tract (mean 5.7 clones, range 3–10 clones) (p<0.0001). The higher env diversity observed within the genital tract compared to plasma was independent of HIV-1 subtype (A, C and D). Our analysis of early mucosal infections in women revealed high HIV-1 diversity in the vaginal tract but few transmitted clones in the blood. These novel in vivo finding suggest a possible mucosal sieve effect, leading to the establishment of a homogenous systemic infection. During chronic HIV-1 infection, high viral diversity can be found in the blood and semen of donors. However, a single HIV-1 clone establishes productive infection in the recipient following heterosexual transmission. To investigate the genetic bottleneck occurring at the earliest stages of heterosexual HIV-1 transmission, we characterized the HIV-1 envelope sequence diversity at very early and early stages of infection in the female reproductive tract and matched plasma samples from a cohort of Ugandan and Zimbabwean women. A more diverse viral population was observed in the endocervical swab samples compared to plasma. Endocervical samples harbored a larger number of viral clones, while in the majority of plasma samples only a single clone was present early in infection. Interestingly, these observations were independent of HIV-1 subtype, hormonal contraceptive use or the number of sex acts and partners. Furthermore, in the cases of higher HIV-1 diversity in the blood during early infection, faster CD4 T cell decline were observed during chronic disease suggesting faster disease progression. Our findings provide novel in vivo evidence for the existence of an intra-patient genetic bottleneck restricting the HIV-1 from the vaginal tract to the blood during early heterosexual HIV-1 transmission.
Collapse
Affiliation(s)
- Katja Klein
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Gabrielle Nickel
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | | | | | - Korey Demers
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Joint Clinical Research Centre, Kampala, Uganda
| | - Emmanuel Ndashimye
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Joint Clinical Research Centre, Kampala, Uganda
| | - Cynthia Kwok
- FHI 360, Durham, North Carolina, United States of America
| | - Pai-Lien Chen
- FHI 360, Durham, North Carolina, United States of America
| | - Sandra Rwambuya
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Joint Clinical Research Centre, Kampala, Uganda
| | - Art Poon
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Marshall Munjoma
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
| | - Tsungai Chipato
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
| | | | | | - Robert A. Salata
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | | | - Eric J. Arts
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Joint Clinical Research Centre, Kampala, Uganda
- * E-mail:
| |
Collapse
|
47
|
Lechanteur A, das Neves J, Sarmento B. The role of mucus in cell-based models used to screen mucosal drug delivery. Adv Drug Deliv Rev 2018; 124:50-63. [PMID: 28751201 DOI: 10.1016/j.addr.2017.07.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/12/2017] [Accepted: 07/22/2017] [Indexed: 12/23/2022]
Abstract
The increasing interest in developing tools to predict drug absorption through mucosal surfaces is fostering the establishment of epithelial cell-based models. Cell-based in vitro techniques for drug permeability assessment are less laborious, cheaper and address the concerns of using laboratory animals. Simultaneously, in vitro barrier models that thoroughly simulate human epithelia or mucosae may provide useful data to speed up the entrance of new drugs and new drug products into the clinics. Nevertheless, standard cell-based in vitro models that intend to reproduce epithelial surfaces often discard the role of mucus in influencing drug permeation/absorption. Biomimetic models of mucosae in which mucus production has been considered may not be able to fully reproduce the amount and architecture of mucus, resulting in biased characterization of permeability/absorption. In these cases, artificial mucus may be used to supplement cell-based models but still proper identification and quantification are required. In this review, considerations regarding the relevance of mucus in the development of cell-based epithelial and mucosal models mimicking the gastro-intestinal tract, the cervico-vaginal tract and the respiratory tract, and the impact of mucus on the permeability mechanisms are addressed. From simple epithelial monolayers to more complex 3D structures, the impact of the presence of mucus for the extrapolation to the in vivo scenario is critically analyzed. Finally, an overview is provided on several techniques and methods to characterize the mucus layer over cell-based barriers, in order to intimately reproduce human mucosal layer and thereby, improve in vitro/in vivo correlation.
Collapse
|
48
|
A slippery slope: On the origin, role and physiology of mucus. Adv Drug Deliv Rev 2018; 124:16-33. [PMID: 29108861 DOI: 10.1016/j.addr.2017.10.014] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/17/2017] [Accepted: 10/29/2017] [Indexed: 02/07/2023]
Abstract
The mucosa of the gastrointestinal tract, eyes, nose, lungs, cervix and vagina is lined by epithelium interspersed with mucus-secreting goblet cells, all of which contribute to their unique functions. This mucus provides an integral defence to the epithelium against noxious agents and pathogens. However, it can equally act as a barrier to drugs and delivery systems targeting epithelial passive and active transport mechanisms. This review highlights the various mucins expressed at different mucosal surfaces on the human body, and their role in creating a mucoid architecture to protect epithelia with specialized functions. Various factors compromising the barrier properties of mucus have been discussed, with an emphasis on how disease states and microbiota can alter the physical properties of mucus. For instance, Akkermansia muciniphila, a bacterium found in higher levels in the gut of lean individuals induces the production of a thickened gut mucus layer. The aims of this article are to elucidate the different physiological, biochemical and physical properties of bodily mucus, a keen appreciation of which will help circumvent the slippery slope of challenges faced in achieving effective mucosal drug and gene delivery.
Collapse
|
49
|
Ren T, Wang Q, Xu Y, Cong L, Gou J, Tao X, Zhang Y, He H, Yin T, Zhang H, Zhang Y, Tang X. Enhanced oral absorption and anticancer efficacy of cabazitaxel by overcoming intestinal mucus and epithelium barriers using surface polyethylene oxide (PEO) decorated positively charged polymer-lipid hybrid nanoparticles. J Control Release 2018; 269:423-438. [DOI: 10.1016/j.jconrel.2017.11.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/21/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022]
|
50
|
Han L, Taub R, Jensen JT. Cervical mucus and contraception: what we know and what we don't. Contraception 2017; 96:310-321. [DOI: 10.1016/j.contraception.2017.07.168] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/17/2017] [Accepted: 07/28/2017] [Indexed: 01/09/2023]
|