1
|
Werner A, Holmes A, Moldovan G, Rodriguez-Garcia M. Innate lymphoid cells in HIV pathogenesis and in the human female genital tract. Curr Opin HIV AIDS 2025; 20:117-123. [PMID: 39774425 DOI: 10.1097/coh.0000000000000908] [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: 01/11/2025]
Abstract
PURPOSE OF REVIEW Women are underrepresented in HIV infection and prevention research despite making up half of people living with HIV. The female genital tract (FGT) serves as a primary site of HIV acquisition, but gaps in knowledge remain regarding protective innate immune mechanisms. Innate lymphoid cells are tissue-resident cells involved in mucosal barrier maintenance and protection, and innate lymphoid cells (ILCs) are altered during chronic HIV infection. However, ILCs role in mucosal HIV pathogenesis is unclear and they are poorly characterized in the FGT. RECENT FINDINGS Human ILCs differ from their mouse counterparts and plastically adjust to their tissue of residency. Human ILC characterization is difficult due to tissue-specific adaptations and transition between subsets. While evidence for ILC involvement in antiviral activity and barrier maintenance is provided in mouse models, human ILC role in mucosal immunity remain understudied, particularly in the FGT. In chronic HIV/simian immunodeficiency virus (SIV) infection, ILCs are altered in a tissue-specific manner, and SIV models indicate potential for antiviral responses. SUMMARY ILCs are tissue-resident plastic cells that provide barrier protection at mucosal surfaces and display antiviral capacity. Considering that HIV is primarily transmitted through mucosal exposure, more research is needed to understand ILC contribution to HIV pathogenesis in human mucosal surfaces relevant for HIV acquisition.
Collapse
Affiliation(s)
- Alexandra Werner
- C.S. Mott Center for Human Growth and Development
- Immunology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Aleah Holmes
- C.S. Mott Center for Human Growth and Development
- Immunology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Genna Moldovan
- C.S. Mott Center for Human Growth and Development
- Department of Obstetrics and Gynecology
| | - Marta Rodriguez-Garcia
- C.S. Mott Center for Human Growth and Development
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan
- Immunology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Parthasarathy S, Moreno de Lara L, Carrillo-Salinas FJ, Werner A, Borchers A, Iyer V, Vogell A, Fortier JM, Wira CR, Rodriguez-Garcia M. Human genital dendritic cell heterogeneity confers differential rapid response to HIV-1 exposure. Front Immunol 2024; 15:1472656. [PMID: 39524443 PMCID: PMC11543421 DOI: 10.3389/fimmu.2024.1472656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Dendritic cells (DCs) play critical roles in HIV pathogenesis and require further investigation in the female genital tract, a main portal of entry for HIV infection. Here we characterized genital DC populations at the single cell level and how DC subsets respond to HIV immediately following exposure. We found that the genital CD11c+HLA-DR+ myeloid population contains three DC subsets (CD1c+ DC2s, CD14+ monocyte-derived DCs and CD14+CD1c+ DC3s) and two monocyte/macrophage populations with distinct functional and phenotypic properties during homeostasis. Following HIV exposure, the antiviral response was dominated by DCs' rapid secretory response, activation of non-classical inflammatory pathways and host restriction factors. Further, we uncovered subset-specific differences in anti-HIV responses. CD14+ DCs were the main population activated by HIV and mediated the secretory antimicrobial response, while CD1c+ DC2s activated inflammasome pathways and IFN responses. Identification of subset-specific responses to HIV immediately after exposure could aid targeted strategies to prevent HIV infection.
Collapse
Affiliation(s)
- Siddharth Parthasarathy
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Laura Moreno de Lara
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | | | - Alexandra Werner
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Anna Borchers
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Vidya Iyer
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
- Mass General Research Institute (MGRI), Division of Clinical Research, Massachusetts General Hospital, Boston, MA, United States
| | - Alison Vogell
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
| | - Jared M. Fortier
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Charles R. Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Marta Rodriguez-Garcia
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
3
|
Moodley M, Moodley J, Naicker T. Placental neutrophil reverse trans-migration and maternal serum neutrophil extracellular trap expression in HIV infection co-morbid pre-eclampsia in women of African ancestry. Histochem Cell Biol 2024; 162:273-286. [PMID: 38913117 PMCID: PMC11364576 DOI: 10.1007/s00418-024-02298-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 06/25/2024]
Abstract
Neutrophil extracellular traps (NETs) and placental neutrophil reverse transmigration (r-TM) are implicated in the pathogenesis of pre-eclampsia (PE). However, the role of the comorbidity of PE and human immunodeficiency virus (HIV) infection in placental neutrophil r-TM and serum NETs remains unknown. Human placental tissue (n = 160) and serum (n = 80) samples were obtained post-ethical approval and divided by pregnancy type and HIV status and across the study population. Immunohistochemistry and morphometry were performed to localize and quantify junctional adhesion molecule-C (JAM-C) expression as an inverse marker of neutrophil r-TM within placental villi. An enzyme-linked immunosorbent assay (ELISA) was performed to quantify the concentration of citrullinated histone H3 (cit-H3) as a marker of NETs. GraphPad Prism (version 8.0.2) was used to compare the results, and a p value of p < 0.05 was considered statistically significant. The localization of JAM-C was observed on the syncytiotrophoblasts (STBs) and endothelial cells of placental villi. The immunoexpression of JAM-C was elevated in PE vs. normotensive (N) placentae. In the exchange villi, JAM-C immunoexpression was higher in the N+ve vs. N-ve group. However, in PE comorbid HIV infection, JAM-C expression was lower in the PE+ve vs. PE-ve group. Citrullinated histone-H3 concentration was lower in the N+ve vs. N-ve group but elevated in early-onset PE (EOPE)+ve vs. late-onset PE (LOPE)+ve group. These results indicate that PE and HIV-infected placentae individually express elevated JAM-C, manifesting in less neutrophil r-TM. However, in exchange villi of PE comorbid with HIV infection reduced JAM-C enhances neutrophil r-TM, thus supporting the synergistic effect of PE comorbid with HIV.
Collapse
Affiliation(s)
- Merantha Moodley
- Department of Obstetrics and Gynecology, School of Clinical Medicine, Women's Health and HIV Research Group, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa.
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa.
| | - Jagidesa Moodley
- Department of Obstetrics and Gynecology, School of Clinical Medicine, Women's Health and HIV Research Group, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa.
| |
Collapse
|
4
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
5
|
Shen Z, vom Steeg LG, Patel MV, Rodriguez-Garcia M, Wira CR. Impact of aging on the frequency, phenotype, and function of CD4+ T cells in the human female reproductive tract. Front Immunol 2024; 15:1465124. [PMID: 39328419 PMCID: PMC11424415 DOI: 10.3389/fimmu.2024.1465124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Since CD4+ T cells are essential for regulating adaptive immune responses and for long lasting mucosal protection, changes in CD4+ T cell numbers and function are likely to affect protective immunity. What remains unclear is whether CD4+ T cell composition and function in the female reproductive tract (FRT) changes as women age. Here we investigated the changes in the composition and function of CD4+ T cells in the endometrium (EM), endocervix (CX), and ectocervix (ECX) with aging. We observed a significant decrease in both the total number and percentage of CD4+ T cells in the EM with increasing age, particularly in the years following menopause. CD4+ T cells within the FRT predominantly expressed CD69. The proportion of CD69+CD4+ T cells increased significantly with increasing age in the EM, CX and ECX. The composition of T helper cell subsets within the EM CD4+ T cell population also showed age-related changes. Specifically, there was a significant increase in the proportion of Th1 cells and a significant decrease in Th17 and Treg cells with increasing age. Furthermore, the production of IFNγ by CD4+ T cells in the EM, CX, and ECX significantly decreased with increasing age upon activation. Our findings highlight the complex changes occurring in CD4+ T cell frequency, phenotype, and function within the FRT as women age. Understanding these age-related immune changes in the FRT is crucial for enhancing our knowledge of reproductive health and immune responses in women.
Collapse
Affiliation(s)
- Zheng Shen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Landon G. vom Steeg
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Mickey V. Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Marta Rodriguez-Garcia
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Charles R. Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
6
|
Khazaeipour Z, Gholamzadeh M, Behnoush AH, Pestei K. The relationship of COVID-19 severity with laboratory findings and neutrophil-to-lymphocyte ratio in patients admitted to a large teaching hospital in Iran: A cross-sectional study. Health Sci Rep 2024; 7:e70045. [PMID: 39246725 PMCID: PMC11377493 DOI: 10.1002/hsr2.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/05/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Background and Aims COVID-19 patients might be admitted to the hospital based on their clinical manifestations or to the intensive care unit (ICU) due to the severity of their symptoms or critical situation. Our main objective was to investigate clinical and demographic factors influencing COVID-19 patients' admission to the ICU and length of stay (LOS) using extracted data from the hospital information systems in Iran. Methods The data of hospitalized patients with confirmed COVID-19 were retrieved from the health information system of Imam Khomeini Hospital Complex, Tehran, Iran between March 2020 and February 2022. The primary outcome was the ICU admission, and the secondary outcome was the LOS. The correlation analysis between laboratory findings and demographic data with ICU admission and LOS was done using SPSS 21.0, and p < 0.05 was considered significant. Results Of all the 4156 patients, 2391 (57.5%) were male and the mean age was 58.69 ± 8.19 years. Of these, 9.5% of patients were admitted to ICU at any time point during their hospital stay. Age and laboratory variables such as neutrophil-to-lymphocyte ratio (NLR), ALT (U/L), albumin (g/dL), plasma glucose (mg/dL), ferritin levels (ng/mL), and phosphorous levels (mg/dL) shown the significant relationship with ICU admission. Also, being a smoker and having hypoxemia had a significant relationship with longer stays in the hospital. In this study, we validated a cut-off value of 4.819 for NLR, calculated at hospitalization, as a useful predictor of disease progression and occurrence of serious clinical outcomes, such as ICU admission. Conclusion The study examined various clinical factors associated with ICU admission in COVID-19 patients. The findings suggest that certain factors can increase the risk of ICU admission and influence the length of hospital stay which should be focused in future studies.
Collapse
Affiliation(s)
- Zahra Khazaeipour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute Tehran University of Medical Sciences Tehran Iran
| | - Marsa Gholamzadeh
- Health Information Management and Medical Informatics Department, School of Allied Medical Sciences Tehran University of Medical Sciences Tehran Iran
| | | | - Khalil Pestei
- Department of Anesthesiology, Imam Khomeini Hospital Complex Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
7
|
Nasr MA, Aldous A, Daniels J, Joy C, Capozzi E, Yang M, Moriarty P, Emmanuel-Baker V, Malcolm S, Green SJ, Gomez-Lobo V, Ghosh M. Effect of progestin-based contraceptives on HIV-associated vaginal immune biomarkers and microbiome in adolescent girls. PLoS One 2024; 19:e0306237. [PMID: 39008499 PMCID: PMC11249223 DOI: 10.1371/journal.pone.0306237] [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: 07/04/2023] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Adolescent girls bear a disproportionate burden of both the HIV epidemic and unintended pregnancies; yet important questions remain unanswered regarding the effects of hormonal contraceptives on the vaginal immune microenvironment, which can impact HIV susceptibility in this group. Multiple studies report genital immune alterations associated with the progestin-based contraceptive Depot medroxyprogesterone acetate (DMPA) in adult women, but there is little available data in adolescents. The objective of this longitudinal cohort study was to evaluate the effects of short-term use of three progestin-based contraceptives, levonorgestrel intrauterine device (LNG-IUD), subdermal etonogestrel (ETNG), and injectable DMPA, on HIV-associated vaginal immune biomarkers and microbiome in adolescent girls. Fifty-nine sexually active, HIV-uninfected girls aged 15-19, were recruited from the Washington DC metro area and self-selected into Control (condoms only), combined oral contraceptive pills, LNG-IUD, ETNG and DMPA groups. Vaginal swabs were collected at baseline prior to contraceptive use and at 3-month follow-up visit. Vaginal secretions were tested for pro-inflammatory (IL-1α, IL-1β, TNF-α, IL-6, IL-8, MIP-3α, IP-10, RANTES, MIP-1α, MIP-1β) and anti-inflammatory/anti-HIV (Serpin-A1, Elafin, Beta-Defensin-2, SLPI) immune biomarkers using ELISA and for anti-HIV activity using TZM-bl assay. Vaginal microbiome was evaluated using 16S rRNA gene sequencing. Data were analyzed using SAS Version 9. Among the 34 participants who completed both visits, no significant changes in median biomarker concentrations, HIV inhibition and microbiome composition were observed between baseline and follow-up visits for any of the contraceptive groups. IL-8 (p<0.01), MIP-3α (0.02), Elafin (p = 0.03) and RANTES (p<0.01) differed significantly by race whereas IL-6 was significantly different by age (p = 0.03). We conclude that 3-month use of LNG-IUD, ETNG and DMPA have minimal effects on adolescent vaginal immune microenvironment, and therefore unlikely to impact HIV risk. Future studies with larger sample size and longer follow-up are recommended to continue to evaluate effects of contraceptives on the lower genital tract immunity and susceptibility to sexually transmitted infections.
Collapse
Affiliation(s)
- Mélodie A. Nasr
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| | - Annette Aldous
- Department of Biostatistics and Bioinformatics, George Washington University, Washington, DC, United States of America
| | - Jason Daniels
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| | - Christopher Joy
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| | - Eleanor Capozzi
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| | - Michelle Yang
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| | - Patricia Moriarty
- MedStar Washington Hospital Center, Washington, DC, United States of America
| | | | - Sharyn Malcolm
- Children’s National Hospital, Washington, DC, United States of America
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, United States of America
| | - Veronica Gomez-Lobo
- MedStar Washington Hospital Center, Washington, DC, United States of America
- Children’s National Hospital, Washington, DC, United States of America
- National Institute of Child Health and Human Development, National Institutes of Health, Washington, DC, United States of America
| | - Mimi Ghosh
- Department of Epidemiology, George Washington University, Washington, DC, United States of America
| |
Collapse
|
8
|
Li Q, Gan Q, Chi H, Meng X, Dalmo RA, Sheng X, Tang X, Xing J, Zhan W. Extracellular traps in skin lesions infected with lymphocystis disease virus in black rockfish (Sebastes schlegelii). FISH & SHELLFISH IMMUNOLOGY 2024; 150:109643. [PMID: 38763177 DOI: 10.1016/j.fsi.2024.109643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
The lymphocystis disease (LCD), caused by Lymphocystis disease virus (LCDV), is a benign and self-limiting disease described in a many freshwater and marine fish species. Hypertrophic fibroblasts and extensive aggregation of inflammatory cells are characteristics of LCD. In the present study, small animal imaging and ultrastructural investigations were carried out on the lymphocystis nodules of black rockfish (Sebastes schlegelii) naturally infected with lymphocystis iridovirus, to assess pathology, and the exudate with particular attention to the formation of extracellular traps (ETs) in vivo. Ex vivo were examined by nodules sections and primary cells stimulation. By histopathological analysis, the nodules contained infiltrated inflammatory cells and extensive basophilic fibrillar filaments at the periphery of the hypertrophied fibroblasts. ETs were assessed in nodules samples using indirect immunofluorescence to detect DNA and myeloperoxidase. Moreover, LCDV was able to infect peritoneal cells of black rockfish in vitro and induce the formation of ETs within 4 h. In summary, this study proved that ETs are involved in the response to LCDV infection and may be involved in formation of lymphoid nodules. Taken together, the findings provide a new perspective to determine the impact factors on the growth of nodules.
Collapse
Affiliation(s)
- Qian Li
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Qiujie Gan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Xianghu Meng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Roy Ambli Dalmo
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, University of Tromsø - The Artic University of Norway, Tromsø, Norway
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
9
|
Choi MW, Isidoro CA, Gillgrass A. Mechanisms of mucosal immunity at the female reproductive tract involved in defense against HIV infection. Curr Opin Virol 2024; 66:101398. [PMID: 38484474 DOI: 10.1016/j.coviro.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 06/07/2024]
Abstract
Human immunodeficiency virus-1 remains a major global health threat. Since the virus is often transmitted through sexual intercourse and women account for the majority of new infections within the most endemic regions, research on mucosal immunity at the female reproductive tract (FRT) is of paramount importance. At the FRT, there are intrinsic barriers to HIV-1 infection, such as epithelial cells and the microbiome, and immune cells of both the innate and adaptive arms are prepared to respond in case the virus overcomes the first line of defense. In this review, we discuss recent findings on FRT mucosal mechanisms of HIV-1 defense and highlight research gaps. While defense from HIV-1 infection at the FRT has been understudied, current and future research is essential to develop new therapeutics and vaccines that can protect this unique mucosal site from HIV-1.
Collapse
Affiliation(s)
- Margaret Wy Choi
- McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Carmina A Isidoro
- McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Amy Gillgrass
- McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
10
|
Dos Ramos Almeida CJL, Veras FP, Paiva IM, Schneider AH, da Costa Silva J, Gomes GF, Costa VF, Silva BMS, Caetite DB, Silva CMS, Salina ACG, Martins R, Bonilha CS, Cunha LD, Jamur MC, da Silva LLP, Arruda E, Zamboni DS, Louzada-Junior P, de Oliveira RDR, Alves-Filho JC, Cunha TM, de Queiroz Cunha F. Neutrophil Virucidal Activity Against SARS-CoV-2 Is Mediated by Neutrophil Extracellular Traps. J Infect Dis 2024; 229:1352-1365. [PMID: 38015657 DOI: 10.1093/infdis/jiad526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Inflammation in the lungs and other vital organs in COVID-19 is characterized by the presence of neutrophils and a high concentration of neutrophil extracellular traps (NETs), which seems to mediate host tissue damage. However, it is not known whether NETs could have virucidal activity against SARS-CoV-2. METHODS We investigated whether NETs could prevent SARS-CoV-2 replication in neutrophils and epithelial cells and what the consequence of NETs degradation would be in K18-humanized ACE2 transgenic mice infected with SARS-CoV-2. RESULTS Here, by immunofluorescence microscopy, we observed that viral particles colocalize with NETs in neutrophils isolated from patients with COVID-19 or healthy individuals and infected in vitro. The inhibition of NETs production increased virus replication in neutrophils. In parallel, we observed that NETs inhibited virus abilities to infect and replicate in epithelial cells after 24 hours of infection. Degradation of NETs with DNase I prevented their virucidal effect in vitro. Using K18-humanized ACE2 transgenic mice, we observed a higher viral load in animals treated with DNase I. However, the virucidal effect of NETs was not dependent on neutrophil elastase or myeloperoxidase activity. CONCLUSIONS Our results provide evidence of the role of NETosis as a mechanism of SARS-CoV-2 viral capture and inhibition.
Collapse
Affiliation(s)
| | - Flávio Protásio Veras
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto
| | - Isadora Marques Paiva
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Ayda Henriques Schneider
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Juliana da Costa Silva
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Giovanni Freitas Gomes
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Victor Ferreira Costa
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - Diego Brito Caetite
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | | | - Ronaldo Martins
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Caio Santos Bonilha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - Maria Célia Jamur
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
| | - Luís Lamberti Pinto da Silva
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Eurico Arruda
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | | | - Paulo Louzada-Junior
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| |
Collapse
|
11
|
da Silva NM, Leite NPDM, Carvalho AE, Almeida VDD, Santos ÍKD, Cavalcanti JRLDP, Fernandes TAADM, Nascimento EGCD, Andrade MFD. The Role of Extracellular Traps in HIV Infection. AIDS Res Hum Retroviruses 2024; 40:308-316. [PMID: 37772695 DOI: 10.1089/aid.2022.0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection is still an important public health problem, which justifies the research of new therapies to combat it. Recent studies show that Extracellular Traps (ETs) are cellular mechanisms useful in the capture and destruction of some viruses, such as the HIV. Here, we show that neutrophils from peripheral blood, genital tissues, and placenta are activated when exposed to human immunodeficiency virus type 1 (HIV-1) and release Neutrophil Extracellular Traps (NETs). The NETs can capture, neutralize, and inactivate the virus and, also, protect other target cells from HIV infection, as long as the DNA and other constituents of the NETs remain intact. Further, the review indicates that the immunoprotective role of NETs in the context of HIV-1 infection is a promising finding for the development of new antiviral therapies. It is necessary, however, the development of studies that evaluate the tissue injury that NETs can cause and the biological relationships with other cells to improve them as therapeutic targets.
Collapse
Affiliation(s)
- Natanias Macson da Silva
- Graduate Program in Health and Society, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Department of Biomedical Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | | | - Amanda Estevam Carvalho
- Multicenter Graduate Program in Biochemistry and Molecular Biology, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | - Valéria Duarte de Almeida
- Multicenter Graduate Program in Physiological Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | - Ísis Kelly Dos Santos
- Department of Physical Education, School of Physical Education, University of Rio Grande do Norte State, Mossoro, Brazil
| | - José Rodolfo Lopes de Paiva Cavalcanti
- Graduate Program in Health and Society, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Department of Biomedical Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Multicenter Graduate Program in Biochemistry and Molecular Biology, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Multicenter Graduate Program in Physiological Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | - Thales Allyrio Araújo de Medeiros Fernandes
- Graduate Program in Health and Society, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Department of Biomedical Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Multicenter Graduate Program in Biochemistry and Molecular Biology, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Multicenter Graduate Program in Physiological Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | - Ellany Gurgel Cosme do Nascimento
- Graduate Program in Health and Society, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Department of Biomedical Sciences, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
| | - Micássio Fernandes de Andrade
- Graduate Program in Health and Society, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Multicenter Graduate Program in Biochemistry and Molecular Biology, School of Health Sciences, University of Rio Grande do Norte State, Mossoro, Brazil
- Department of Health Sciences, School of Biological and Health Sciences, Federal Rural University of the Semi-arid, Mossoro, Brazil
| |
Collapse
|
12
|
Yan Q, Jia S, Li D, Yang J. The role and mechanism of action of microbiota-derived short-chain fatty acids in neutrophils: From the activation to becoming potential biomarkers. Biomed Pharmacother 2023; 169:115821. [PMID: 37952355 DOI: 10.1016/j.biopha.2023.115821] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate, have emerged as critical mediators in the communication between the human microbiota and its host. As the first responder to the inflammatory site, neutrophils play an important role in protecting the host against bacterial infections. Recent investigations revealed that SCFAs generated from microbiota influence various neutrophil activities, including activation, migration, and generation of mediators of inflammatory processes. SCFAs have also been demonstrated to exhibit potential therapeutic benefits in a variety of disorders related to neutrophil dysfunction, including inflammatory bowel disease, viral infectious disorders, and cancer. This study aims to examine the molecular processes behind the complicated link between SCFAs and neutrophils, as well as their influence on neutrophil-driven inflammatory disorders. In addition, we will also provide an in-depth review of current research on the diagnostic and therapeutic value of SCFAs as possible biomarkers for neutrophil-related diseases.
Collapse
Affiliation(s)
- Qingzhu Yan
- Department of Ultrasound Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Shengnan Jia
- Digestive Diseases Center, Department of Hepatopancreatobiliary Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Dongfu Li
- Digestive Diseases Center, Department of Hepatopancreatobiliary Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Junling Yang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
13
|
Wang L, Liu T, Yue H, Zhang J, Sheng Q, Wu L, Wang X, Zhang M, Wang J, Wang J, Yu W. Clinical characteristics and high risk factors of patients with Omicron variant strain infection in Hebei, China. Front Cell Infect Microbiol 2023; 13:1294904. [PMID: 38145047 PMCID: PMC10744887 DOI: 10.3389/fcimb.2023.1294904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/26/2023] Open
Abstract
Objective The Omicron variant has a weaker pathogenicity compared to the Delta variant but is highly transmissible and elderly critically ill patients account for the majority. This study has significant implications for guiding clinical personalized treatment and effectively utilizing healthcare resources. Methods The study focuses on 157 patients infected with the novel coronavirus Omicron variant, from December, 2022, to February, 2023. The objective is to analyze the baseline data, test results, imaging findings and identify risk factors associated with severe illness. Results Among the 157 included patients, there were 55 cases in the non-severe group (all were moderate cases) and 102 cases in the severe group (including severe and critical cases). Infection with the Omicron variant exhibits significant differences between non-severe and severe cases (baseline data, blood routine, coagulation, inflammatory markers, cardiac, liver, kidney functions, Chest CT, VTE score, etc.). A multifactorial logistic regression analysis showed that neutrophil percentage >75%, eosinophil percentage <0.4%, D-dimer >0.55 mg/L, PCT >0.25 ng/mL, LDH >250 U/L, albumin <40 g/L, A/G ratio <1.2, cholinesterase<5100 U/L, uric acid >357 mole/L and blood calcium<2.11 mmol/L were the most likely independent risk factors for severe novel coronavirus infection. Conclusion Advanced age, low oxygenation index, elevated neutrophil percentage, decreased eosinophil percentage, elevated PCT, elevated LDH, decreased albumin, decreased A/G ratio, elevated uric acid, decreased blood calcium, and elevated D-dimer are independent prognostic risk factors for non-severe patients progressing to severe illness. These factors should be closely monitored and actively treated to prevent or minimize the occurrence of severe illness.
Collapse
Affiliation(s)
- Lihong Wang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ting Liu
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongjuan Yue
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaojiao Zhang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qihong Sheng
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ling Wu
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoyu Wang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mei Zhang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Wang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jia Wang
- Department of Infectious Diseases, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weifang Yu
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Moreno de Lara L, Werner A, Borchers A, Carrillo-Salinas FJ, Marmol W, Parthasarathy S, Iyer V, Vogell A, Illanes D, Abadía-Molina AC, Ochsenbauer C, Wira CR, Rodriguez-Garcia M. Aging dysregulates neutrophil extracellular trap formation in response to HIV in blood and genital tissues. Front Immunol 2023; 14:1256182. [PMID: 38035114 PMCID: PMC10684664 DOI: 10.3389/fimmu.2023.1256182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Women acquire HIV through sexual transmission, with increasing incidence in women >50 years old. Identifying protective mechanisms in the female genital tract (FGT) is important to prevent HIV-acquisition in women as they age. Human genital and blood neutrophils inactivate HIV by releasing neutrophil extracellular traps (NETs), an innate protective mechanism against HIV-infection. However, how NET formation is triggered by HIV in different tissues and whether this mechanism is affected by aging remain unknown. We demonstrate that the mechanisms that trigger NET release in response to HIV are different in blood and genital tissues, and that NET release decreases with aging. In blood neutrophils, HIV stimulation independently activated calcium pathways and endosomal TLR8, but aging reduced calcium responses, resulting in delayed NET release. In contrast, calcium responses were absent in genital neutrophils and NET release was triggered preferentially through TLR8 activation, but aging impaired this pathway. HIV induced NET formation through non-lytic pathways in blood and FGT neutrophils, except for a small subset of NETs that incorporated annexin V and lactoferrin predominantly in blood, suggesting proinflammatory and lytic NET release. Our findings demonstrate that blood neutrophils cannot model genital neutrophil responses which has important implications to understanding protection against HIV acquisition.
Collapse
Affiliation(s)
- Laura Moreno de Lara
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Alexandra Werner
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Anna Borchers
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | | | - Wendelin Marmol
- Program in Genetics, Molecular, and Cellular Biology, Tufts University School of Medicine, Boston, MA, United States
| | | | - Vidya Iyer
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
| | - Alison Vogell
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
| | - Diego Illanes
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
| | - Ana C. Abadía-Molina
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular 3 e Inmunología, Universidad de Granada, Granada, Spain
| | - Christina Ochsenbauer
- Department of Medicine and UAB Center for AIDS Research, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charles R. Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Marta Rodriguez-Garcia
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
15
|
Fa P, Ke BG, Dupre A, Tsung A, Zhang H. The implication of neutrophil extracellular traps in nonalcoholic fatty liver disease. Front Immunol 2023; 14:1292679. [PMID: 38022519 PMCID: PMC10652891 DOI: 10.3389/fimmu.2023.1292679] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an expanding worldwide health concern, and the underlying mechanisms contributing to its progression still need further exploration. Neutrophil extracellular traps (NETs) are intricate formations comprised of nuclear constituents and diverse antimicrobial granules that are released into the extracellular milieu by activated neutrophils upon various triggers, which play a pivotal part in the onset and advancement of NAFLD. NETs actively participate in the genesis of NAFLD by fostering oxidative stress and inflammation, ultimately resulting in hepatic fat accumulation and the escalation of liver injury. Recent insights into the interaction with other hepatic immune populations and mediators, such as macrophages and T regulatory cells, have revealed several important mechanisms that can trigger further liver injury. In conclusion, the formation of NETs emerged as an important factor in the development of NAFLD, offering a promising target for innovative therapeutic approaches against this debilitating condition. This comprehensive review seeks to compile existing studies exploring the involvement of NETs in the genesis of NAFLD and their influence on the immune response throughout the progression of NAFLD.
Collapse
Affiliation(s)
- Pengyan Fa
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Benjamin G. Ke
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Abigail Dupre
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
16
|
Park J, Dean LS, Heckl J, Gangcuangco LM, Pedro TK, Tallquist MD, Seto TB, Shiramizu B, Chow DC, Shikuma CM. Low-density granulocytes display immature cells with enhanced NET formation in people living with HIV. Sci Rep 2023; 13:13282. [PMID: 37587169 PMCID: PMC10432506 DOI: 10.1038/s41598-023-40475-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
While the protective role of neutrophil extracellular traps (NETs) in limiting human immunodeficiency virus (HIV) spread to susceptible cells has been documented, there is comparatively little insight into whether NET formation is harmful in people living with HIV (PLWH). To gain insight into neutrophil dysregulation and the pathological role of NETs in HIV, we examined expressions of NET-associated markers [cell-free DNA (cfDNA) and citrullinated histone H3 (CitH3)] in the plasmas from a cohort of the Hawaii Aging with HIV-cardiovascular and HIV-seronegative (HIV-) individuals. In a subset of participants, circulating low-density granulocyte (LDG) levels and their maturation and activation status were analyzed via flow cytometry. We demonstrated higher plasma levels of CitH3 in PLWH compared to HIV- individuals. LDGs from PLWH had heightened CD66b, but reduced CD16 expression. The percentages and counts of CD10+ LDGs were significantly decreased in PLWH. In addition, the CD16Lo LDG subsets were enriched in PLWH, compared to HIV- group, indicating that immature LDGs are increased in PLWH. Moreover, LDGs from PLWH exhibited significantly higher NET forming capacity. In summary, our study presents evidence that LDGs from PLWH on ART display an immature and altered phenotype with increased NET formation. Among PLWH, plasma NET levels as well as LDG parameters correlated with blood markers for inflammation and coagulation, suggesting that neutrophil activation and NETs may exert proinflammatory and coagulation effects. Our data provide insights into the pathologic role of LDGs at least in part mediated through NET formation in PLWH.
Collapse
Affiliation(s)
- Juwon Park
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA.
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA.
| | - Logan S Dean
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Jack Heckl
- Department of Cell and Molecular Biology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Louie Mar Gangcuangco
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Te-Kie Pedro
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, 96813, USA
| | - Todd B Seto
- The Queen's Medical Center, Honolulu, HI, 96813, USA
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Bruce Shiramizu
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Dominic C Chow
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Cecilia M Shikuma
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| |
Collapse
|
17
|
Costa-Fujishima M, Yazdanpanah A, Horne S, Lamont A, Lopez P, Farr Zuend C, Birse K, Taverner M, Greenslade R, Abou M, Noel-Romas L, Abrenica B, Ajibola O, Ikeogu N, Su RC, McKinnon LR, Pymar H, Poliquin V, Berard AR, Burgener AD, Murooka TT. Nonoptimal bacteria species induce neutrophil-driven inflammation and barrier disruption in the female genital tract. Mucosal Immunol 2023; 16:341-356. [PMID: 37121385 DOI: 10.1016/j.mucimm.2023.04.001] [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: 02/09/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023]
Abstract
Neutrophil recruitment and activation within the female genital tract are often associated with tissue inflammation, loss of vaginal epithelial barrier integrity, and increased risk for sexually transmitted infections, such as HIV-1. However, the direct role of neutrophils on vaginal epithelial barrier function during genital inflammation in vivo remains unclear. Using complementary proteome and immunological analyses, we show high neutrophil influx into the lower female genital tract in response to physiological surges in progesterone, stimulating distinct stromal, immunological, and metabolic signaling pathways. However, despite the release of extracellular matrix-modifying proteases and inflammatory mediators, neutrophils contributed little to physiological mucosal remodeling events such as epithelial shedding or re-epithelialization during transition from diestrus to estrus phase. In contrast, the presence of bacterial vaginosis-associated bacteria resulted in a rapid and sustained neutrophil recruitment, resulting in vaginal epithelial barrier leakage and decreased cell-cell junction protein expression in vivo. Thus, neutrophils are important mucosal sentinels that rapidly respond to various biological cues within the female genital tract, dictating the magnitude and duration of the ensuing inflammatory response at steady state and during disease processes.
Collapse
Affiliation(s)
- Marina Costa-Fujishima
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Atta Yazdanpanah
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Samantha Horne
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Alana Lamont
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Paul Lopez
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Christina Farr Zuend
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA
| | - Kenzie Birse
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Morgan Taverner
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada
| | - Riley Greenslade
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Max Abou
- National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Laura Noel-Romas
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Bernard Abrenica
- National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Oluwaseun Ajibola
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Nnamdi Ikeogu
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Ruey-Chyi Su
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Lyle R McKinnon
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa; Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Helen Pymar
- University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Vanessa Poliquin
- University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Alicia R Berard
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Adam D Burgener
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada; Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas T Murooka
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada; University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada.
| |
Collapse
|
18
|
Mo J, Yang Y, Feng J, Lei Y, Huang S, Cen W, Wei S, Huang H, Lu J, Zhang J. Single-cell analysis reveals dysregulated inflammatory response in peripheral blood immunity in patients with acute respiratory distress syndrome. Front Cell Dev Biol 2023; 11:1199122. [PMID: 37283946 PMCID: PMC10239863 DOI: 10.3389/fcell.2023.1199122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: Acute respiratory distress syndrome (ARDS) remains a major clinical challenge for patients in intensive care units. Determining the differential mechanisms underlying ARDS with different etiologies is a key goal to improve the effectiveness of ARDS therapy. Despite growing evidence that different immune cell types are involved in ARDS, the role of altered immune cell subpopulations in disease progression is unelucidated. Methods: In this study, we combined scRNA-seq and bulk-level sequencing to analyze the transcriptomes of peripheral blood mononuclear cells from healthy volunteers and patients with septic ARDS (sep-ARDS) and pneumonic ARDS (PNE-ARDS). Results: Our data revealed differential alterations at the cellular and molecular levels and within biological signaling pathways in ARDS with different etiologies. The dynamics of neutrophils, macrophages (Macs), classical dendritic cells (cDCs), myeloid-derived suppressive cells (MDSCs), and CD8+ T cells varied significantly among groups of different samples, with neutrophils and cDCs at higher, and Macs at significantly lower, amounts in the patients with sep-ARDS. Furthermore, MDSCs were highly enriched only in the sep-ARDS patients, whereas a higher abundance of CD8+ T cells was observed in patients with PNE-ARDS. In addition, these cell subpopulations were found to be significantly involved in apoptosis, inflammatory, and immune-related pathways. In particular, a significant enhancement of the oxidative stress response was observed in the neutrophil subpopulation. Conclusion: Our study shows that the composition of cells involved in the main peripheral circulation differs in patients with ARDS with different etiologies. Studying the role and mechanism of action of these cells during ARDS will provide new opportunities for the treatment of this condition.
Collapse
Affiliation(s)
- Jingjia Mo
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanli Yang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jihua Feng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanhua Lei
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Suhong Huang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weiluan Cen
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shanshan Wei
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Huang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianfeng Zhang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
19
|
Feng C, Jin C, Liu K, Yang Z. Microbiota-derived short chain fatty acids: Their role and mechanisms in viral infections. Biomed Pharmacother 2023. [DOI: 10.1016/j.biopha.2023.114414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023] Open
|
20
|
Schaid TR, LaCroix I, Hansen KC, D'Alessandro A, Moore EE, Sauaia A, Dzieciatkowska M, DeBot M, Cralley AL, Thielen O, Hallas W, Erickson C, Mitra S, Banerjee A, Jones K, Silliman CC, Cohen MJ. A proteomic analysis of NETosis in trauma: Emergence of serpinB1 as a key player. J Trauma Acute Care Surg 2023; 94:361-370. [PMID: 36730076 PMCID: PMC9974543 DOI: 10.1097/ta.0000000000003849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Release of neutrophil extracellular traps (NETosis) may mediate postinjury organ dysfunction, but mechanisms remain unclear. The intracellular serine protease inhibitor (serpin) B1 is vital to neutrophil function and has been shown to restrict NETosis in inflammatory settings. In this study, we used discovery proteomics to identify the proteomic signature of trauma-induced NETosis. We hypothesized that serpinB1 would be a major component of this NET protein profile and associated with adverse outcomes. METHODS This was a post hoc analysis of data collected as part of the COMBAT randomized clinical trial. Blood was collected from injured patients at a single Level I Trauma Center. Proteomic analyses were performed through targeted liquid chromatography coupled with mass spectrometry. Abundances of serpinB1 and known NETosis markers were analyzed with patient and injury characteristics, clinical data, and outcomes. RESULTS SerpinB1 levels on emergency department (ED) arrival were significantly correlated with proteomic markers of NETosis, including core histones, transketolase, and S100A8/A9 proteins. More severely injured patients had elevated serpinB1 and NETosis markers on ED arrival. Levels of serpinB1 and top NETosis markers were significantly elevated on ED arrival in nonsurvivors and patients with fewer ventilator- and ICU-free days. In proteome-wide receiver operating characteristic analysis, serpinB1 was consistently among the top proteins associated with adverse outcomes. Among NETosis markers, levels of serpinB1 early in the patient's course exhibited the greatest separation between patients with fewer and greater ventilator- and ICU-free days. Gene Ontology analysis of top predictors of adverse outcomes further supports NETosis as a potential mediator of postinjury organ dysfunction. CONCLUSION We have identified a proteomic signature of trauma-induced NETosis, and NETosis is an early process following severe injury that may mediate organ dysfunction. In addition, serpinB1 is a major component of this NET protein profile that may serve as an early marker of excessive NETosis after injury.
Collapse
Affiliation(s)
- Terry R Schaid
- From the Department of Surgery/Trauma Research Center (T.R.S.Jr, E.E.M., A.S., M.D.B., O.T., W.H., S.M., A.B., K.J., C.C.S., M.J.C.), Department of Biochemistry and Molecular Genetics (I.L.C., K.C.H., A.D'A., M.D., C.E.), University of Colorado Denver, School of Medicine, Aurora; Department of Surgery (E.E.M., A.L.C.), Denver Health Medical Center, Denver; Department of Health Systems, Management, and Policy (A.S.), University of Colorado Denver, School of Medicine, Aurora; Vitalant Research Institute (C.C.S.), Denver; and Department of Pediatrics (C.C.S.), University of Colorado Denver, School of Medicine, Aurora, CO
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
Adapen C, Réot L, Menu E. Role of the human vaginal microbiota in the regulation of inflammation and sexually transmitted infection acquisition: Contribution of the non-human primate model to a better understanding? FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:992176. [PMID: 36560972 PMCID: PMC9763629 DOI: 10.3389/frph.2022.992176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
The human vaginal microbiota has a central role in the regulation of the female reproductive tract (FRT) inflammation. Indeed, on one hand an optimal environment leading to a protection against sexually transmitted infections (STI) is associated with a high proportion of Lactobacillus spp. (eubiosis). On the other hand, a more diverse microbiota with a high amount of non-Lactobacillus spp. (dysbiosis) is linked to a higher local inflammation and an increased STI susceptibility. The composition of the vaginal microbiota is influenced by numerous factors that may lead to a dysbiotic environment. In this review, we first discuss how the vaginal microbiota composition affects the local inflammation with a focus on the cytokine profiles, the immune cell recruitment/phenotype and a large part devoted on the interactions between the vaginal microbiota and the neutrophils. Secondly, we analyze the interplay between STI and the vaginal microbiota and describe several mechanisms of action of the vaginal microbiota. Finally, the input of the NHP model in research focusing on the FRT health including vaginal microbiota or STI acquisition/control and treatment is discussed.
Collapse
Affiliation(s)
- Cindy Adapen
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Louis Réot
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
23
|
de Jesus Gonzalez-Contreras F, Zarate X. Neutrophil extracellular traps: Modulation mechanisms by pathogens. Cell Immunol 2022; 382:104640. [PMID: 36413806 DOI: 10.1016/j.cellimm.2022.104640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022]
Abstract
Neutrophils, as innate effector cells, play an essential role in the containment and elimination of pathogens. Among the main neutrophil mechanisms use for these processes is the release of neutrophil extracellular traps (NETs), which consist of decondensed DNA decorated with various cytoplasmic proteins. NETs' principal role is the trapping and elimination of infectious agents; therefore, the formation of NETs is regulated by bacteria, fungi, parasites, and viruses through different mechanisms: the presence of virulence factors (adhered or secreted), microbial load, size of the microorganism, and even due to other immune cells activation (mainly platelets). This review summarizes the significant aspects that contribute to NETs modulation by pathogens and their components, and the effect NETs have on these pathogens as a cellular defense mechanism.
Collapse
Affiliation(s)
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, San Nicolas de los Garza 66455, NL, Mexico
| |
Collapse
|
24
|
Yang K, Liu J, Gong Y, Li Y, Liu Q. Bioinformatics and systems biology approaches to identify molecular targeting mechanism influenced by COVID-19 on heart failure. Front Immunol 2022; 13:1052850. [DOI: 10.3389/fimmu.2022.1052850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has emerged as a contemporary hazard to people. It has been known that COVID-19 can both induce heart failure (HF) and raise the risk of patient mortality. However, the mechanism underlying the association between COVID-19 and HF remains unclear. The common molecular pathways between COVID-19 and HF were identified using bioinformatic and systems biology techniques. Transcriptome analysis was performed to identify differentially expressed genes (DEGs). To identify gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways, common DEGs were used for enrichment analysis. The results showed that COVID-19 and HF have several common immune mechanisms, including differentiation of T helper (Th) 1, Th 2, Th 17 cells; activation of lymphocytes; and binding of major histocompatibility complex class I and II protein complexes. Furthermore, a protein-protein interaction network was constructed to identify hub genes, and immune cell infiltration analysis was performed. Six hub genes (FCGR3A, CD69, IFNG, CCR7, CCL5, and CCL4) were closely associated with COVID-19 and HF. These targets were associated with immune cells (central memory CD8 T cells, T follicular helper cells, regulatory T cells, myeloid-derived suppressor cells, plasmacytoid dendritic cells, macrophages, eosinophils, and neutrophils). Additionally, transcription factors, microRNAs, drugs, and chemicals that are closely associated with COVID-19 and HF were identified through the interaction network.
Collapse
|
25
|
Chen X, Ma H, Mo S, Yu S, Lu Z, Chen J. Intratumoral neutrophil extracellular traps are associated with unfavorable clinical outcomes and immunogenic context in pancreatic ductal adenocarcinoma. Front Immunol 2022; 13:1027459. [PMID: 36325339 PMCID: PMC9618733 DOI: 10.3389/fimmu.2022.1027459] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/15/2022] Open
Abstract
Extracellular traps (ETs) and tumor-infiltrating immune cells play crucial roles in tumor progression. However, little is known about the clinical significance of tumor-infiltrating neutrophils and macrophages and the related ETs in pancreatic ductal adenocarcinoma (PDAC). This study investigates the associations between neutrophil or macrophage infiltration or ET formation and the clinicopathological features, molecular characteristics, immune checkpoint molecules, clinical outcomes, and response to adjuvant chemotherapy (ACT) in PDAC. We performed multiplex immunofluorescence staining to detect ET formation by neutrophils or macrophages using tissue microarrays obtained from 205 patients, and analyzed the immunohistochemistry data for PD-L1, PD-L2, B7-H3, and B7-H4. The ET expression rates in macrophages and neutrophils were 23.9% and 45.4%, respectively. Patients with a high density of neutrophils or positive expression of neutrophil ETs exhibited poorer progression-free survival (PFS) and disease-specific survival (DSS), whereas macrophage ETs were not related to PFS and DSS. Neutrophil infiltration and ET formation were identified as independent prognostic predictors of DSS using univariate and multivariate Cox analyses. Patients with PDAC with lower neutrophil infiltration or negative staining for neutrophil ETs are more likely to benefit from ACT. Patients with PDAC were more accurately stratified based on the infiltration of neutrophils and presence of neutrophil ETs, and patients with low neutrophil infiltration and negative staining for neutrophil ETs showed the best survival. Patients with positive neutrophil ETs demonstrated inferior DSS compared to those with negative neutrophil ETs in the PD-L1 tumor proportion score (TPS) < 1% and PD-L1 IC < 1% subgroups. However, the positive expression of neutrophil ETs was not related to DSS in the PD-L1 TPS ≥ 1% or PD-L1 IC ≥ 1% subgroup. Our findings emphasize the potential of neutrophil infiltration and ETs as prognostic markers that could guide the formulation of more effective personalized treatments for PDAC.
Collapse
|
26
|
Margiana R, Sharma SK, Khan BI, Alameri AA, Opulencia MJC, Hammid AT, Hamza TA, Babakulov SK, Abdelbasset WK, Jawhar ZH. RETRACTED: The pathogenicity of COVID-19 and the role of pentraxin-3: An updated review study. Pathol Res Pract 2022; 238:154128. [PMID: 36137396 PMCID: PMC9476367 DOI: 10.1016/j.prp.2022.154128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 01/08/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. In investigating concerns regarding the contributions of the authors to this article, the editors reached out to the authors for an explanation. In addition to the concerns regarding the contribution of each author, the editors discovered suspicious changes in authorship between the original submission and the revised version of this paper. The names of the authors Ameer A Alameri and Zanko Hassan Jawhar were added to the revised version of the article without explanation and without the exceptional approval by the handling Editor, which is contrary to the journal policy on changes to authorship. The authors were unable to provide a reasonable explanation for either of the issues raised. The editor therefore feels that the findings of the manuscript cannot be relied upon and that the article needs to be retracted.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Satish Kumar Sharma
- Department of Pharmacology, Glocal School of Pharmacy, The Glocal University, Saharanpur, India.
| | | | | | | | - Ali Thaeer Hammid
- Computer Engineering Techniques Department, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - Thulfeqar Ahmed Hamza
- Medical laboratory techniques department, Al-Mustaqbal University College, Babylon, Iraq
| | - Sharaf Khamrakulovich Babakulov
- Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan; Research scholar, Department of Scientific affairs, Samarkand State Medical Institute, Amir Temur Street 18, Samarkand, Uzbekistan
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia; Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Science, Lebanese French University, Kurdistan Region, Iraq
| |
Collapse
|
27
|
Lu L, Liu LP, Gui R, Dong H, Su YR, Zhou XH, Liu FX. Discovering common pathogenetic processes between COVID-19 and sepsis by bioinformatics and system biology approach. Front Immunol 2022; 13:975848. [PMID: 36119022 PMCID: PMC9471316 DOI: 10.3389/fimmu.2022.975848] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
Corona Virus Disease 2019 (COVID-19), an acute respiratory infectious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has spread rapidly worldwide, resulting in a pandemic with a high mortality rate. In clinical practice, we have noted that many critically ill or critically ill patients with COVID-19 present with typical sepsis-related clinical manifestations, including multiple organ dysfunction syndrome, coagulopathy, and septic shock. In addition, it has been demonstrated that severe COVID-19 has some pathological similarities with sepsis, such as cytokine storm, hypercoagulable state after blood balance is disrupted and neutrophil dysfunction. Considering the parallels between COVID-19 and non-SARS-CoV-2 induced sepsis (hereafter referred to as sepsis), the aim of this study was to analyze the underlying molecular mechanisms between these two diseases by bioinformatics and a systems biology approach, providing new insights into the pathogenesis of COVID-19 and the development of new treatments. Specifically, the gene expression profiles of COVID-19 and sepsis patients were obtained from the Gene Expression Omnibus (GEO) database and compared to extract common differentially expressed genes (DEGs). Subsequently, common DEGs were used to investigate the genetic links between COVID-19 and sepsis. Based on enrichment analysis of common DEGs, many pathways closely related to inflammatory response were observed, such as Cytokine-cytokine receptor interaction pathway and NF-kappa B signaling pathway. In addition, protein-protein interaction networks and gene regulatory networks of common DEGs were constructed, and the analysis results showed that ITGAM may be a potential key biomarker base on regulatory analysis. Furthermore, a disease diagnostic model and risk prediction nomogram for COVID-19 were constructed using machine learning methods. Finally, potential therapeutic agents, including progesterone and emetine, were screened through drug-protein interaction networks and molecular docking simulations. We hope to provide new strategies for future research and treatment related to COVID-19 by elucidating the pathogenesis and genetic mechanisms between COVID-19 and sepsis.
Collapse
Affiliation(s)
- Lu Lu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Le-Ping Liu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yan-Rong Su
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiong-Hui Zhou
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Feng-Xia Liu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Feng-Xia Liu,
| |
Collapse
|
28
|
Carrillo-Salinas FJ, Parthasarathy S, Moreno de Lara L, Borchers A, Ochsenbauer C, Panda A, Rodriguez-Garcia M. Short-Chain Fatty Acids Impair Neutrophil Antiviral Function in an Age-Dependent Manner. Cells 2022; 11:2515. [PMID: 36010593 PMCID: PMC9406757 DOI: 10.3390/cells11162515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Half of the people living with HIV are women. Younger women remain disproportionally affected in endemic areas, but infection rates in older women are rising worldwide. The vaginal microbiome influences genital inflammation and HIV infection risk. Multiple factors, including age, induce vaginal microbial alterations, characterized by high microbial diversity that generate high concentrations of short-chain fatty acids (SCFAs), known to modulate neutrophil function. However, how SCFAs may modulate innate anti-HIV protection by neutrophils is unknown. To investigate SCFA-mediated alterations of neutrophil function, blood neutrophils from younger and older women were treated with SCFAs (acetate, butyrate and propionate) at concentrations within the range reported during bacterial vaginosis, and phenotype, migration and anti-HIV responses were evaluated. SCFA induced phenotypical changes preferentially in neutrophils from older women. Butyrate decreased CD66b and increased CD16 and CD62L expression, indicating low activation and prolonged survival, while propionate increased CD54 and CXCR4 expression, indicating a mature aged phenotype. Furthermore, acetate and butyrate significantly inhibited neutrophil migration in vitro and specifically reduced α-defensin release in older women, molecules with anti-HIV activity. Following HIV stimulation, SCFA treatment delayed NET release and dampened chemokine secretion compared to untreated neutrophils in younger and older women. Our results demonstrate that SCFAs can impair neutrophil-mediated anti-HIV responses.
Collapse
Affiliation(s)
| | - Siddharth Parthasarathy
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
- Immunology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Laura Moreno de Lara
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
- Immunology Unit, Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
| | - Anna Borchers
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Christina Ochsenbauer
- Department of Medicine, Hem/Onc & CFAR, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Alexander Panda
- Tufts Medical Center/Division of Pulmonary and Critical Care (PCCM), Boston, MA 02111, USA
- Tufts Clinical and Translational Science Institute (CTSI), Boston, MA 02111, USA
| | - Marta Rodriguez-Garcia
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
- Immunology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| |
Collapse
|
29
|
Liana P, Liberty IA, Murti K, Hafy Z, Salim EM, Zulkarnain M, Umar TP. A systematic review on neutrophil extracellular traps and its prognostication role in COVID-19 patients. Immunol Res 2022; 70:449-460. [PMID: 35604493 PMCID: PMC9125547 DOI: 10.1007/s12026-022-09293-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Neutrophil extracellular traps (NETs) are extracellular webs composed of neutrophil granular and nuclear elements. Because of the potentially dangerous amplification circuit between inflammation and tissue damage, NETs are becoming one of the investigated components in the current Coronavirus Disease 2019 (COVID-19) pandemic. The purpose of this systematic review is to summarize studies on the role of NETs in determining the prognosis of COVID-19 patients. The study used six databases: PubMed, Science Direct, EBSCOHost, Europe PMC, ProQuest, and Scopus. This literature search was implemented until October 31, 2021. The search terms were determined specifically for each databases, generally included the Neutrophil Extracellular Traps, COVID-19, and prognosis. The Newcastle Ottawa Scale (NOS) was then used to assess the risk of bias. Ten studies with a total of 810 participants were chosen based on the attainment of the prerequisite. Two were of high quality, seven were of moderate quality, and the rest were of low quality. The majority of studies compared COVID-19 to healthy control. Thrombosis was observed in three studies, while four studies recorded the need for mechanical ventilation. In COVID-19 patients, the early NETs concentration or the evolving NETs degradations can predict patient mortality. Based on their interactions with inflammatory and organ dysfunction markers, it is concluded that NETs play a significant role in navigating the severity of COVID-19 patients and thus impacting their prognosis.
Collapse
Affiliation(s)
- Phey Liana
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sriwijaya/Dr Mohammad Hoesin General Hospital, Palembang, Indonesia
- Biomedicine Doctoral Program, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Iche Andriyani Liberty
- Department of Public Health and Community Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Krisna Murti
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Sriwijaya, Dr. Moh. Ali Street RSMH complex, Palembang, South Sumatera Indonesia
| | - Zen Hafy
- Biomedical Department, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Eddy Mart Salim
- Division of Allergy and Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Sriwijaya/Dr, Mohammad Hoesin General Hospital, Palembang, Indonesia
| | - Mohammad Zulkarnain
- Department of Public Health and Community Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Tungki Pratama Umar
- Medical Profession Program, Faculty of Medicine, Sriwijaya University, Palembang, Indonesia
| |
Collapse
|
30
|
Ogweno G. Challenges in Platelet Functions in HIV/AIDS Management. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The interest in platelet functions in HIV/AIDS is due to the high incidence of microvascular thrombosis in these individuals. A lot of laboratory data have been generated regarding platelet functions in this population. The tests demonstrate platelet hyperactivity but decreased aggregation, though results are inconsistent depending on the study design. Antiretroviral treatments currently in use display complex interactions. Many studies on platelet functions in these patients have been for research purposes, but none have found utility in guiding drug treatment of thrombosis.
Collapse
|
31
|
Dave TV, Nair AG, Joseph J, Freitag SK. Immunopathology of COVID-19 and its implications in the development of rhino-orbital-cerebral mucormycosis: a major review. Orbit 2022; 41:670-679. [PMID: 35856238 DOI: 10.1080/01676830.2022.2099428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE To present a literature review on various immunopathologic dysfunctions following COVID-19 infection and their potential implications in development of rhino-orbital-cerebral mucormycosis (ROCM). METHODS A literature search was performed via Google Scholar and PubMed with subsequent review of the accompanying references. Analogies were drawn between the immune and physiologic deviations caused by COVID-19 and the tendency of the same to predispose to ROCM. RESULTS Sixty-two articles were reviewed. SARS-CoV-2 virus infection leads to disruption of epithelial integrity in the respiratory passages, which may be a potential entry point for the ubiquitous Mucorales to become invasive. COVID-19 related GRP78 protein upregulation may aid in spore germination and hyphal invasion by Mucorales. COVID-19 causes interference in macrophage functioning by direct infection, a tendency for hyperglycemia, and creation of neutrophil extracellular traps. This affects innate immunity against Mucorales. Thrombocytopenia and reduction in the number of natural killer (NK) cells and infected dendritic cells is seen in COVID-19. This reduces the host immune response to pathogenic invasion by Mucorales. Cytokines released in COVID-19 cause mitochondrial dysfunction and accumulation of reactive oxygen species, which cause oxidative damage to the leucocytes. Hyperferritinemia also occurs in COVID-19 resulting in suppression of the hematopoietic proliferation of B- and T-lymphocytes. CONCLUSIONS COVID-19 has a role in the occurrence of ROCM due to its effects at the entry point of the fungus in the respiratory mucosa, effects of the innate immune system, creation of an environment of iron overload, propagation of hyperglycemia, and effects on the adaptive immune system.
Collapse
Affiliation(s)
- Tarjani Vivek Dave
- Ophthalmic Plastic Surgery Service, LV Prasad Eye Institute, Hyderabad, India
| | - Akshay Gopinathan Nair
- Aditya Jyot Eye Hospital, Mumbai, India.,Advanced Eye hospital and Institute, Navi Mumbai, India
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Kallam Anji Reddy Campus, LV Prasad Eye Institute, Hyderabad, India
| | - Suzanne K Freitag
- Ophthalmic Plastic Surgery Service, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Penaloza Arias LC, Huynh DN, Babity S, Marleau S, Brambilla D. Optimization of a Liposomal DNase I Formulation with an Extended Circulating Half-Life. Mol Pharm 2022; 19:1906-1916. [PMID: 35543327 DOI: 10.1021/acs.molpharmaceut.2c00086] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Drug delivery systems such as liposomes are widely used to stabilize and increase the plasma half-life of therapeutics. In this article, we have investigated two strategies to increase the half-life of deoxyribonuclease I, an FDA-approved enzyme used for the treatment of cystic fibrosis, and a potential candidate for the reduction of uncontrolled inflammation induced by neutrophil extracellular traps. We demonstrate that our optimized preparation procedure resulted in nanoparticles with improved plasma half-life and total exposure relative to native protein, while maintaining enzymatic activity.
Collapse
Affiliation(s)
| | - David N Huynh
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec Canada H3T 1J4
| | - Samuel Babity
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec Canada H3T 1J4
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec Canada H3T 1J4
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec Canada H3T 1J4
| |
Collapse
|
33
|
Pastorek M, Dúbrava M, Celec P. On the Origin of Neutrophil Extracellular Traps in COVID-19. Front Immunol 2022; 13:821007. [PMID: 35359960 PMCID: PMC8961727 DOI: 10.3389/fimmu.2022.821007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Despite ongoing vaccination COVID-19 is a global healthcare problem because of the lack of an effective targeted therapy. In severe COVID-19 manifesting as acute respiratory distress syndrome, uncontrolled innate immune system activation results in cytokine deregulation, damage-associated molecular patterns release upon tissue damage and high occurrence of thrombotic events. These pathomechanisms are linked to neutrophil function and dysfunction, particularly increased formation of neutrophil extracellular traps (NETs). While the association of NETs and severity of COVID-19 has been shown and proved, the causes of NETs formation are unclear. The aim of this review is to summarize potential inducers of NETs formation in severe COVID-19 and to discuss potential treatment options targeting NETs formation of removal.
Collapse
Affiliation(s)
- Michal Pastorek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Dúbrava
- Department of Geriatric Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| |
Collapse
|
34
|
Girsch JH, Mejia Plazas MC, Olivier A, Farah M, Littlefield D, Behl S, Punia S, Sakemura R, Hemsath JR, Norgan A, Enninga EAL, Johnson EL, Chakraborty R. Host-Viral Interactions at the Maternal-Fetal Interface. What We Know and What We Need to Know. FRONTIERS-A JOURNAL OF WOMEN STUDIES 2022; 2:833106. [PMID: 36742289 PMCID: PMC9894500 DOI: 10.3389/fviro.2022.833106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In humans, the hemochorial placenta is a unique temporary organ that forms during pregnancy to support fetal development, gaseous exchange, delivery of nutrition, removal of waste products, and provides immune protection, while maintaining tolerance to the HLA-haploidentical fetus. In this review, we characterize decidual and placental immunity during maternal viral (co)-infection with HIV-1, human cytomegalovirus (HCMV), and Zika virus. We discuss placental immunology, clinical presentation, and epidemiology, before characterizing host susceptibility and cellular tropism, and how the three viruses gain access into specific placental target cells. We describe current knowledge on host-viral interactions with decidual and stromal human placental macrophages or Hofbauer cells, trophoblasts including extra villous trophoblasts, T cells, and decidual natural killer (dNK) cells. These clinically significant viral infections elicit both innate and adaptive immune responses to control replication. However, the three viruses either during mono- or co-infection (HIV-1 and HCMV) escape detection to initiate placental inflammation associated with viral transmission to the developing fetus. Aside from congenital or perinatal infection, other adverse pregnancy outcomes include preterm labor and spontaneous abortion. In addition, maternal HIV-1 and HCMV co-infection are associated with impaired fetal and infant immunity in postnatal life and poor clinical outcomes during childhood in exposed infants, even in the absence of vertical transmission of HIV-1. Given the rapidly expanding numbers of HIV-1-exposed uninfected infants and children globally, further research is urgently needed on neonatal immune programming during maternal mono-and co-infection. This review therefore includes sections on current knowledge gaps that may prompt future research directions. These gaps reflect an emerging but poorly characterized field. Their significance and potential investigation is underscored by the fact that although viral infections result in adverse consequences in both mother and developing fetus/newborn, antiviral and immunomodulatory therapies can improve clinical outcomes in the dyad.
Collapse
Affiliation(s)
- James H. Girsch
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Mayo Clinic Graduate School of Biomedical Science, Rochester, MN, United States
| | - Maria C. Mejia Plazas
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Amanda Olivier
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Mohamed Farah
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Dawn Littlefield
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Supriya Behl
- Department of Pediatric Research, Mayo Clinic, Rochester, MN, United States
| | - Sohan Punia
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Reona Sakemura
- Department of Hematology Research, Mayo Clinic, Rochester, MN, United States
| | - Jack R. Hemsath
- Department of Infectious Diseases Research, Mayo Clinic, Rochester, MN, United States
| | - Andrew Norgan
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Elizabeth A. L. Enninga
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Erica L. Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States,Correspondence: Rana Chakraborty
| |
Collapse
|
35
|
Reno TA, Tarnus L, Tracy R, Landay AL, Sereti I, Apetrei C, Pandrea I. The Youngbloods. Get Together. Hypercoagulation, Complement, and NET Formation in HIV/SIV Pathogenesis. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2021.795373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic, systemic T-cell immune activation and inflammation (IA/INFL) have been reported to be associated with disease progression in persons with HIV (PWH) since the inception of the AIDS pandemic. IA/INFL persist in PWH on antiretroviral therapy (ART), despite complete viral suppression and increases their susceptibility to serious non-AIDS events (SNAEs). Increased IA/INFL also occur during pathogenic SIV infections of macaques, while natural hosts of SIVs that control chronic IA/INFL do not progress to AIDS, despite having persistent high viral replication and severe acute CD4+ T-cell loss. Moreover, natural hosts of SIVs do not present with SNAEs. Multiple mechanisms drive HIV-associated IA/INFL, including the virus itself, persistent gut dysfunction, coinfections (CMV, HCV, HBV), proinflammatory lipids, ART toxicity, comorbidities, and behavioral factors (diet, smoking, and alcohol). Other mechanisms could also significantly contribute to IA/INFL during HIV/SIV infection, notably, a hypercoagulable state, characterized by elevated coagulation biomarkers, including D-dimer and tissue factor, which can accurately identify patients at risk for thromboembolic events and death. Coagulation biomarkers strongly correlate with INFL and predict the risk of SNAE-induced end-organ damage. Meanwhile, the complement system is also involved in the pathogenesis of HIV comorbidities. Despite prolonged viral suppression, PWH on ART have high plasma levels of C3a. HIV/SIV infections also trigger neutrophil extracellular traps (NETs) formation that contribute to the elimination of viral particles and infected CD4+ T-cells. However, as SIV infection progresses, generation of NETs can become excessive, fueling IA/INFL, destruction of multiple immune cells subsets, and microthrombotic events, contributing to further tissue damages and SNAEs. Tackling residual IA/INFL has the potential to improve the clinical course of HIV infection. Therefore, therapeutics targeting new pathways that can fuel IA/INFL such as hypercoagulation, complement activation and excessive formation of NETs might be beneficial for PWH and should be considered and evaluated.
Collapse
|
36
|
Al-Kuraishy HM, Al-Gareeb AI, Al-Hussaniy HA, Al-Harcan NAH, Alexiou A, Batiha GES. Neutrophil Extracellular Traps (NETs) and Covid-19: A new frontiers for therapeutic modality. Int Immunopharmacol 2022; 104:108516. [PMID: 35032828 PMCID: PMC8733219 DOI: 10.1016/j.intimp.2021.108516] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (Covid-19) is a worldwide infectious disease caused by severe acute respiratory coronavirus 2 (SARS-CoV-2). In severe SARS-CoV-2 infection, there is severe inflammatory reactions due to neutrophil recruitments and infiltration in the different organs with the formation of neutrophil extracellular traps (NETs), which involved various complications of SARS-CoV-2 infection. Therefore, the objective of the present review was to explore the potential role of NETs in the pathogenesis of SARS-CoV-2 infection and to identify the targeting drugs against NETs in Covid-19 patients. Different enzyme types are involved in the formation of NETs, such as neutrophil elastase (NE), which degrades nuclear protein and release histones, peptidyl arginine deiminase type 4 (PADA4), which releases chromosomal DNA and gasdermin D, which creates pores in the NTs cell membrane that facilitating expulsion of NT contents. Despite of the beneficial effects of NETs in controlling of invading pathogens, sustained formations of NETs during respiratory viral infections are associated with collateral tissue injury. Excessive development of NETs in SARS-CoV-2 infection is linked with the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) due to creation of the NETs-IL-1β loop. Also, aberrant NTs activation alone or through NETs formation may augment SARS-CoV-2-induced cytokine storm (CS) and macrophage activation syndrome (MAS) in patients with severe Covid-19. Furthermore, NETs formation in SARS-CoV-2 infection is associated with immuno-thrombosis and the development of ALI/ARDS. Therefore, anti-NETs therapy of natural or synthetic sources may mitigate SARS-CoV-2 infection-induced exaggerated immune response, hyperinflammation, immuno-thrombosis, and other complications.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyiah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyiah University, Baghdad, Iraq
| | | | - Nasser A Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Bagdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia; AFNP Med Austria, Wien, Austria.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Al Beheira, Egypt.
| |
Collapse
|
37
|
Efficacy and specificity of different methods for human neutrophil extracellular trap isolation and handling. Cent Eur J Immunol 2021; 46:384-387. [PMID: 34764811 PMCID: PMC8574115 DOI: 10.5114/ceji.2021.108771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/23/2021] [Indexed: 12/01/2022] Open
Abstract
Introduction Although in vitro incubation of various cell types with neutrophil extracellular traps (NETs) is commonly used to investigate the influence of NETs on cellular function, it is unclear which human NET isolation and handling protocol is superior. The present study sought to assess the efficacy (yield and purity) and efficiency (time taken) of different available human NET isolation and handling protocols. Material and methods Neutrophils isolated from human blood were stimulated using phorbol 12-myristate 13-acetate. Four distinct protocols were used to isolate NETs, and the yield was quantified using fluorimetry. Results Addition of the restriction enzyme AluI prior to centrifugation is unique to the most effective NET isolation method, yielding a NET concentration of 1077.22 ±229.04 ng/ml (at 523 nm) measured with PicoGreen. Immediate centrifugation to pellet neutrophils is unique to the most efficient method. Conclusions Balancing protocol efficacy and efficiency, the method incorporating centrifugation for 5 min at 450 × γ to pellet neutrophils is more than adequate.
Collapse
|
38
|
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
|
39
|
Jones R, Manickam C, Ram DR, Kroll K, Hueber B, Woolley G, Shah SV, Smith S, Varner V, Reeves RK. Systemic and mucosal mobilization of granulocyte subsets during lentiviral infection. Immunology 2021; 164:348-357. [PMID: 34037988 PMCID: PMC8442246 DOI: 10.1111/imm.13376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 11/30/2022] Open
Abstract
Granulocytes mediate broad immunoprotection through phagocytosis, extracellular traps, release of cytotoxic granules, antibody effector functions and recruitment of other immune cells against pathogens. However, descriptions of granulocytes in HIV infection and mucosal tissues are limited. Our goal was to characterize granulocyte subsets in systemic, mucosal and lymphoid tissues during lentiviral infection using the rhesus macaque (RM) model. Mononuclear cells from jejunum, colon, cervix, vagina, lymph nodes, spleen, liver and whole blood from experimentally naïve and chronically SHIVsf162p3-infected RM were analysed by microscopy and polychromatic flow cytometry. Granulocytes were identified using phenotypes designed specifically for RM: eosinophils-CD45+ CD66+ CD49d+ ; neutrophils-CD45+ CD66+ CD14+ ; and basophils-CD45+ CD123+ FcRε+ . Nuclear visualization with DAPI staining and surface marker images by ImageStream (cytometry/microscopy) further confirmed granulocytic phenotypes. Flow cytometric data showed that all RM granulocytes expressed CD32 (FcRγII) but did not express CD16 (FcRγIII). Additionally, constitutive expression of CD64 (FcRγI) on neutrophils and FcRε on basophils indicates the differential expression of Fc receptors on granulocyte subsets. Granulocytic subsets in naïve whole blood ranged from 25·4% to 81·5% neutrophils, 0·59% to 13·3% eosinophils and 0·059% to 1·8% basophils. Interestingly, elevated frequencies of circulating neutrophils, colorectal neutrophils and colorectal eosinophils were all observed in chronic lentiviral disease. Conversely, circulating basophils, jejunal eosinophils, vaginal neutrophils and vaginal eosinophils of SHIVsf162p3-infected RM declined in frequency. Overall, our data suggest modulation of granulocytes in chronic lentiviral infection, most notably in the gastrointestinal mucosae where a significant inflammation and disruption occurs in lentivirus-induced disease. Furthermore, granulocytes may migrate to inflamed tissues during infection and could serve as targets of immunotherapeutic intervention.
Collapse
Affiliation(s)
- Rhianna Jones
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Cordelia Manickam
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Daniel R. Ram
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Kyle Kroll
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Brady Hueber
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Griffin Woolley
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Spandan V. Shah
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Scott Smith
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - Valerie Varner
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| | - R. Keith Reeves
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
- Ragon Institute of Massachusetts General Hospital, MIT, and HarvardCambridgeMAUSA
- Division of Innate and Comparative Immunology, Center for Human Systems ImmunologyDuke University School of MedicineDurhamNCUSA
| |
Collapse
|
40
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
41
|
Abstract
Abstract
The innate immune system is mandatory for the activation of antiviral host defense and eradication of the infection. In this regard, dendritic cells, natural killer cells, macrophages, neutrophils representing the cellular component, and cytokines, interferons, complement or Toll-Like Receptors, representing the mediators of unspecific response act together for both activation of the adaptive immune response and viral clearance. Of great importance is the proper functioning of the innate immune response from the very beginning. For instance, in the early stages of viral infection, the defective interferon response leads to uncontrolled viral replication and pathogen evasion, while hypersecretion during the later stages of infection generates hyperinflammation. This cascade activation of systemic inflammation culminates with cytokine storm syndrome and hypercoagulability state, due to a close interconnection between them. Thus an unbalanced reaction, either under- or over- stimulation of the innate immune system will lead to an uncoordinated response and unfavorable disease outcomes. Since both cellular and humoral factors are involved in the time-course of the innate immune response, in this review we aimed to address their gradual involvement in the antiviral response with emphasis on key steps in SARS-CoV-2 infection.
Collapse
|
42
|
Fontoura MA, Rocha RF, Marques RE. Neutrophil Recruitment and Participation in Severe Diseases Caused by Flavivirus Infection. Life (Basel) 2021; 11:717. [PMID: 34357089 PMCID: PMC8304117 DOI: 10.3390/life11070717] [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: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are first-line responders to infections and are recruited to target tissues through the action of chemoattractant molecules, such as chemokines. Neutrophils are crucial for the control of bacterial and fungal infections, but their role in the context of viral infections has been understudied. Flaviviruses are important human viral pathogens transmitted by arthropods. Infection with a flavivirus may result in a variety of complex disease manifestations, including hemorrhagic fever, encephalitis or congenital malformations. Our understanding of flaviviral diseases is incomplete, and so is the role of neutrophils in such diseases. Here we present a comprehensive overview on the participation of neutrophils in severe disease forms evolving from flavivirus infection, focusing on the role of chemokines and their receptors as main drivers of neutrophil function. Neutrophil activation during viral infection was shown to interfere in viral replication through effector functions, but the resulting inflammation is significant and may be detrimental to the host. For congenital infections in humans, neutrophil recruitment mediated by CXCL8 would be catastrophic. Evidence suggests that control of neutrophil recruitment to flavivirus-infected tissues may reduce immunopathology in experimental models and patients, with minimal loss to viral clearance. Further investigation on the roles of neutrophils in flaviviral infections may reveal unappreciated functions of this leukocyte population while increasing our understanding of flaviviral disease pathogenesis in its multiple forms.
Collapse
Affiliation(s)
- Marina Alves Fontoura
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Cellular and Structural Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-865, Brazil
| | - Rebeca Fróes Rocha
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Genetics and Molecular Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
| |
Collapse
|
43
|
Tabrizi ZA, Khosrojerdi A, Aslani S, Hemmatzadeh M, Babaie F, Bairami A, Shomali N, Hosseinzadeh R, Safari R, Mohammadi H. Multi-facets of neutrophil extracellular trap in infectious diseases: Moving beyond immunity. Microb Pathog 2021; 158:105066. [PMID: 34174356 DOI: 10.1016/j.micpath.2021.105066] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Neutrophil extracellular traps (NETs) are networks of extracellular chromosomal DNA fibers, histones, and cytoplasmic granule proteins. The release of NET components from neutrophils is involved in the suppression of pathogen diffusion. Development of NETs around target microbes leads to disruption of the cell membrane, eventuating in kind of cell death that is called as NETosis. The very first step in the process of NETosis is activation of Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase upon signaling by innate immune receptors. Afterwards, produced Reactive oxygen species (ROS) trigger protein-arginine deiminase type 4, neutrophil elastase, and myeloperoxidase to generate decondensed chromatin and disrupted integrity of nuclear membrane. Subsequently, decondensed chromatin is mixed with several enzymes in the cytoplasm released from granules, leading to release of DNA and histones, and finally formation of NET. Several reports have indicated that NETosis might contribute to the immune responses through limiting the dissemination of microbial organisms. In this review, we discuss recent advances on the role of neutrophils, NETs, and their implications in the pathogenesis of microbial infections. Additionally, the prospective of the NET modulation as a therapeutic strategy to treat infectious diseases are clarified.
Collapse
Affiliation(s)
- Zahra Azimzadeh Tabrizi
- Department of Laboratory Sciences, School of Allied Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Babaie
- Department of Immunology and Genetic, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Bairami
- Department of Medical Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Roghaiyeh Safari
- Molecular and Cellular Epigenetics, GIGA, University of Liege, Sart-Tilman Liège, Belgium; Molecular and Cellular Biology, TERRA, Gembloux Agro-Bio Tech, University of Liege, Gembloux, Belgium
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
44
|
Reif T, Dyckhoff G, Hohenberger R, Kolbe CC, Gruell H, Klein F, Latz E, Stolp B, Fackler OT. Contact-dependent inhibition of HIV-1 replication in ex vivo human tonsil cultures by polymorphonuclear neutrophils. CELL REPORTS MEDICINE 2021; 2:100317. [PMID: 34195682 PMCID: PMC8233696 DOI: 10.1016/j.xcrm.2021.100317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/02/2021] [Accepted: 05/20/2021] [Indexed: 12/01/2022]
Abstract
Polymorphonuclear neutrophils (PMNs), the most abundant white blood cells, are recruited rapidly to sites of infection to exert potent anti-microbial activity. Information regarding their role in infection with human immunodeficiency virus (HIV) is limited. Here we report that addition of PMNs to HIV-infected cultures of human tonsil tissue or peripheral blood mononuclear cells causes immediate and long-lasting suppression of HIV-1 spread and virus-induced depletion of CD4 T cells. This inhibition of HIV-1 spread strictly requires PMN contact with infected cells and is not mediated by soluble factors. 2-Photon (2PM) imaging visualized contacts of PMNs with HIV-1-infected CD4 T cells in tonsil tissue that do not result in lysis or uptake of infected cells. The anti-HIV activity of PMNs also does not involve degranulation, formation of neutrophil extracellular traps, or integrin-dependent cell communication. These results reveal that PMNs efficiently blunt HIV-1 replication in primary target cells and tissue by an unconventional mechanism. PMNs blunt HIV-1 spread and CD4 T cell depletion in HIV-infected human tonsils Suppression of HIV-1 replication by PMNs requires cell-cell contacts PMNs do not affect HIV via effector functions such as NETosis or degranulation PMNs exert unconventional antiviral activity
Collapse
Affiliation(s)
- Tatjana Reif
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ralph Hohenberger
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Carl-Christian Kolbe
- Institute of Innate Immunity, Department of Innate Immunity and Metaflammation, University Hospital Bonn, 53127 Bonn, Germany
| | - Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Department of Innate Immunity and Metaflammation, University Hospital Bonn, 53127 Bonn, Germany
| | - Bettina Stolp
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
45
|
Garishah FM, Rother N, Riswari SF, Alisjahbana B, Overheul GJ, van Rij RP, van der Ven A, van der Vlag J, de Mast Q. Neutrophil Extracellular Traps in Dengue Are Mainly Generated NOX-Independently. Front Immunol 2021; 12:629167. [PMID: 34122402 PMCID: PMC8187769 DOI: 10.3389/fimmu.2021.629167] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/05/2021] [Indexed: 01/12/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are increasingly recognized to play a role in the pathogenesis of viral infections, including dengue. NETs can be formed NADPH oxidase (NOX)-dependently or NOX-independently. NOX-independent NETs can be induced by activated platelets and are very potent in activating the endothelium. Platelet activation with thrombocytopenia and endothelial dysfunction are prominent features of dengue virus infection. We postulated that dengue infection is associated with NOX-independent NET formation, which is related to platelet activation, endothelial perturbation and increased vascular permeability. Using our specific NET assays, we investigated the time course of NET formation in a cohort of Indonesian dengue patients. We found that plasma levels of NETs were profoundly elevated and that these NETs were predominantly NOX-independent NETs. During early recovery phase (7-13 days from fever onset), total NETs correlated negatively with platelet number and positively with platelet P-selectin expression, the binding of von Willebrand factor to platelets and levels of Syndecan-1. Patients with gall bladder wall thickening, an early marker of plasma leakage, had a higher median level of total NETs. Ex vivo, platelets induced NOX-independent NET formation in a dengue virus non-structural protein 1 (NS1)-dependent manner. We conclude that NOX-independent NET formation is enhanced in dengue, which is most likely mediated by NS1 and activated platelets.
Collapse
Affiliation(s)
- Fadel Muhammad Garishah
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Nils Rother
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Silvita Fitri Riswari
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia.,Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Bachti Alisjahbana
- Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia.,Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - André van der Ven
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
46
|
Dauletova M, Hafsan H, Mahhengam N, Zekiy AO, Ahmadi M, Siahmansouri H. Mesenchymal stem cell alongside exosomes as a novel cell-based therapy for COVID-19: A review study. Clin Immunol 2021; 226:108712. [PMID: 33684527 PMCID: PMC7935675 DOI: 10.1016/j.clim.2021.108712] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
In the past year, an emerging disease called Coronavirus disease 2019 (COVID-19), caused by Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been discovered in Wuhan, China, which has become a worrying pandemic and has challenged the world health system and economy. SARS-CoV-2 enters the host cell through a specific receptor (Angiotensin-converting enzyme 2) expressed on epithelial cells of various tissues. The virus, by inducing cell apoptosis and production of pro-inflammatory cytokines, generates as cytokine storm, which is the major cause of mortality in the patients. This type of response, along with responses by other immune cell, such as alveolar macrophages and neutrophils causes extensive damage to infected tissue. Newly, a novel cell-based therapy by Mesenchymal stem cell (MSC) as well as by their exosomes has been developed for treatment of COVID-19 that yielded promising outcomes. In this review study, we discuss the characteristics and benefits of MSCs therapy as well as MSC-secreted exosome therapy in treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Meruyert Dauletova
- Department of Propaedeutics and Internal Medicine, Akhmet Yassawi Internationl Kazakh-Turkish University, Turkistan, Kazakhstan
| | - Hafsan Hafsan
- Department of Biology, Faculty of Science and Technology, Universitas Islam Negeri Alauddin Makassar, South Sulawesi, Indonesia
| | - Negah Mahhengam
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Olivera-Valle I, Latorre MC, Calvo M, Gaspar B, Gómez-Oro C, Collazos A, Breton A, Caballero-Campo P, Ardoy M, Asensio F, Sánchez-Mateos P, Pérez-Millan F, Relloso M. Vaginal neutrophils eliminate sperm by trogocytosis. Hum Reprod 2021; 35:2567-2578. [PMID: 33011783 DOI: 10.1093/humrep/deaa198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION What is the vaginal polymorphonuclear (PMN) spermicidal mechanism to reduce the excess of sperm? SUMMARY ANSWER We show that PMNs are very efficient at killing sperm by a trogocytosis-dependent spermicidal activity independent of neutrophil extracellular traps (NETs). WHAT IS KNOWN ALREADY Trogocytosis has been described as an active membrane exchange between immune cells with a regulatory purpose. Recently, trogocytosis has been reported as a mechanism which PMNs use to kill tumour cells or Trichomonas vaginalis. STUDY DESIGN, SIZE, DURATION We used in vivo murine models and human ex vivo sperm and PMNs to investigate the early PMN-sperm response. PARTICIPANTS/MATERIALS, SETTING, METHODS We set up a live/dead sperm detection system in the presence of PMNs to investigate in vivo and ex vivo PMN-spermicidal activity by confocal microscopy, flow cytometry and computer-assisted sperm analysis (SCA). MAIN RESULTS AND THE ROLE OF CHANCE We revealed that PMNs are highly efficient at killing sperm by way of a NETs-independent, contact-dependent and serine proteases-dependent engulfment mechanism. PMNs 'bite' sperm and quickly reduce sperm motility (within 5 min) and viability (within 20 min) after contact. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study was conducted using murine models and healthy human blood PMNs; whether it is relevant to human vaginal PMNs or to cases of infertility is unknown. WIDER IMPLICATIONS OF THE FINDINGS Vaginal PMNs attack and immobilize excess sperm in the vagina by trogocytosis because sperm are exogenous and may carry pathogens. Furthermore, this mechanism of sperm regulation has low mucosal impact and avoids an exacerbated inflammatory response that could lead to mucosal damage or infertility. STUDY FUNDING/COMPETING INTEREST(S) This work was partially supported by Ministry of Economy and Competitiveness ISCIII-FIS grants, PI16/00050, and PI19/00078, co-financed by ERDF (FEDER) Funds from the European Commission, 'A way of making Europe' and IiSGM intramural grant II-PI-MRC-2017. M.R. holds a Miguel Servet II contract (CPII14/00009). M.C.L. holds IiSGM intramural contract. There are no competing interests.
Collapse
Affiliation(s)
- I Olivera-Valle
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - M C Latorre
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - M Calvo
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - B Gaspar
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Servicio de Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - C Gómez-Oro
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - A Collazos
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - A Breton
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Servicio de Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - P Caballero-Campo
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - M Ardoy
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Servicio de Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - F Asensio
- Animalario, Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - P Sánchez-Mateos
- Laboratorio de Inmuno-oncología, Servicio de Inmunologia, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - F Pérez-Millan
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Servicio de Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - M Relloso
- Laboratorio de InmunoFisiología, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
48
|
Yang HC, Ma TH, Tjong WY, Stern A, Chiu DTY. G6PD deficiency, redox homeostasis, and viral infections: implications for SARS-CoV-2 (COVID-19). Free Radic Res 2021; 55:364-374. [PMID: 33401987 PMCID: PMC7799378 DOI: 10.1080/10715762.2020.1866757] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/08/2023]
Abstract
The COVID-19 pandemic has so far affected more than 45 million people and has caused over 1 million deaths worldwide. Infection with SARS-CoV-2, the pathogenic agent, which is associated with an imbalanced redox status, causes hyperinflammation and a cytokine storm, leading to cell death. Glucose-6-phosphate dehydrogenase (G6PD) deficient individuals may experience a hemolytic crisis after being exposed to oxidants or infection. Individuals with G6PD deficiency are more susceptible to coronavirus infection than individuals with normally functioning G6PD. An altered immune response to viral infections is found in individuals with G6PD deficiency. Evidence indicates that G6PD deficiency is a predisposing factor of COVID-19.
Collapse
Affiliation(s)
- Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Tian-Hsiang Ma
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Wen-Ye Tjong
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, NY, USA
| | - Daniel Tsun-Yee Chiu
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
49
|
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
|
50
|
Blondin-Ladrie L, Aranguren M, Doyon-Laliberté K, Poudrier J, Roger M. The Importance of Regulation in Natural Immunity to HIV. Vaccines (Basel) 2021; 9:vaccines9030271. [PMID: 33803543 PMCID: PMC8003059 DOI: 10.3390/vaccines9030271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Worldwide, most Human Immunodeficiency Virus (HIV) infections are acquired through heterosexual intercourse, and in sub-Saharan Africa, 59% of new HIV infections affect women. Vaccines and microbicides hold promise for preventing the acquisition of HIV. To this end, the study of HIV highly exposed seronegative (HESN) female commercial sex workers (CSWs), who constitute a model of natural immunity to HIV, provides an exceptional opportunity to determine important clues for the development of preventive strategies. Studies using both female genital tract (FGT) and peripheral blood samples of HESN CSWs, have allowed identifying distinct features, notably low-inflammatory patterns associated with resistance to infection. How this seemingly regulated response is achieved at the initial site of HIV infection remains unknown. One hypothesis is that populations presenting regulatory profiles contribute to the orchestration of potent anti-viral and low-inflammatory responses at the initial site of HIV transmission. Here, we view to update our knowledge regarding this issue.
Collapse
Affiliation(s)
- Laurence Blondin-Ladrie
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Matheus Aranguren
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Kim Doyon-Laliberté
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Johanne Poudrier
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
- Correspondence: (J.P.); (M.R.)
| | - Michel Roger
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
- Institut National de Santé Publique du Québec, Montréal, QC H2P1E2, Canada
- Correspondence: (J.P.); (M.R.)
| |
Collapse
|