1
|
Hamamah S, Barry F, Vannier S, Anahory T, Haahtela T, Antó JM, Chapron C, Ayoubi JM, Czarlewski W, Bousquet J. Infertility, IL-17, IL-33 and Microbiome Cross-Talk: The Extended ARIA-MeDALL Hypothesis. Int J Mol Sci 2024; 25:11981. [PMID: 39596052 PMCID: PMC11594021 DOI: 10.3390/ijms252211981] [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] [Received: 09/24/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Infertility, defined as the inability to obtain pregnancy after 12 months of regular unprotected sexual intercourse, has increased in prevalence over the past decades, similarly to chronic, allergic, autoimmune, or neurodegenerative diseases. A recent ARIA-MeDALL hypothesis has proposed that all these diseases are linked to dysbiosis and to some cytokines such as interleukin 17 (IL-17) and interleukin 33 (IL-33). Our paper suggests that endometriosis, a leading cause of infertility, is linked to endometrial dysbiosis and two key cytokines, IL-17 and IL-33, which interact with intestinal dysbiosis. Intestinal dysbiosis contributes to elevated estrogen levels, a primary factor in endometriosis. Estrogens strongly activate IL-17 and IL-33, supporting the existence of a gut-endometrial axis as a significant contributor to infertility.
Collapse
Affiliation(s)
- Samir Hamamah
- Biologie de la Reproduction, Hôpital Arnaud de Villeneuve, 34295 Montpellier, France; (F.B.); (T.A.)
- INSERM DEFE, Université de Montpellier, 34090 Montpellier, France
| | - Fatima Barry
- Biologie de la Reproduction, Hôpital Arnaud de Villeneuve, 34295 Montpellier, France; (F.B.); (T.A.)
- INSERM DEFE, Université de Montpellier, 34090 Montpellier, France
| | - Sarah Vannier
- Gynécologie Médicale, Hôpital Arnaud de Villeneuve, 34295 Montpellier, France;
| | - Tal Anahory
- Biologie de la Reproduction, Hôpital Arnaud de Villeneuve, 34295 Montpellier, France; (F.B.); (T.A.)
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, 00250 Helsinki, Finland;
| | - Josep M. Antó
- ISGlobal, Barcelona Institute for Global Health, 08036 Barcelona, Spain;
- IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
- Departamento de Ciencias Experimentales y de la Salud, Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Charles Chapron
- Service de Gynécologie-Obs., Hôpital Cochin, 75014 Paris, France;
| | - Jean-Marc Ayoubi
- Gynécologie et médecine de la Reproduction, Hôpital Foch, 92150 Suresnes, France;
| | | | - Jean Bousquet
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| |
Collapse
|
2
|
Mian MF, Pa S, Rahman N, Gillgrass A, Kaushic C. NET-EN treatment leads to delayed HSV-2 infection, enhanced mucin and T cell functions in the female genital tract when compared to DMPA in a preclinical mouse model. Front Immunol 2024; 15:1427842. [PMID: 39569191 PMCID: PMC11576457 DOI: 10.3389/fimmu.2024.1427842] [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: 05/04/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Depot-medroxyprogesterone acetate (DMPA) and Norethisterone Enanthate (NET-EN) are progestin-only injectable contraceptives widely used by women in sub-Sharan Africa, where incidence of HIV-1 and HSV-2 infection remains high. Studies indicate that DMPA usage can increase the risk of HSV-2 infection, but limited data indicate no increased risk with use of NET-EN. We therefore investigated the effects of NET-EN and DMPA on susceptibility to vaginal HSV-2 infection in ovariectomized (OVX) mice and effects on immune responses, particularly in the vaginal tract (VT). OVX mice, when treated with NET-EN and infected intravaginally, had delayed genital pathology, decreased viral shedding, and extended survival compared to DMPA- or untreated OVX mice. CD4+ T cells isolated from VT showed no significant change in frequency with either contraceptive. However, DMPA significantly decreased the total number of VT CD4+ and CD8+ T cells and the number of IFN-γ producing CD4 and CD8 T cells and increased the percentage of CD4 and CD8 T cells producing TNF-α compared to untreated mice. In contrast, NET-EN significantly enhanced percentages of CD8+ T cells compared to DMPA treated mice, and frequencies of IFN-γ+ CD4 and CD8 T cells in the VT compared to untreated mice. Comparative analysis of splenic lymphocytes indicated that DMPA treatment resulted in reduction of CD4+ T cell frequency, but enhanced TNF-α+ CD4 T cells compared to untreated mice. NET-EN enhanced the frequency of CD8 T cells, as well as IFN-γ+ and TNF-α+ CD4, and IFN-γ+ CD8 T cells in the spleen compared to untreated mice. Importantly, we found DMPA treatment that significantly reduced mucin production, whereas NET-EN enhanced expression of cell-associated mucin in VT. High levels of mucin in NET-EN mice were associated with lower levels of HSV-2 virus detected in the vaginal tract. This study provides the first evidence that NET-EN treatment can delay HSV-2 infection compared to DMPA.
Collapse
Affiliation(s)
- M Firoz Mian
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sidney Pa
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Nuzhat Rahman
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amy Gillgrass
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
3
|
Hoffmann JP, Srivastava A, Yang H, Iwanaga N, Remcho TP, Hewes JL, Sharoff R, Song K, Norton EB, Kolls JK, McCombs JE. Vaccine-elicited IL-1R signaling results in Th17 TRM-mediated immunity. Commun Biol 2024; 7:433. [PMID: 38594380 PMCID: PMC11003962 DOI: 10.1038/s42003-024-06138-0] [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: 05/18/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Lung tissue resident memory (TRM) cells are thought to play crucial roles in lung host defense. We have recently shown that immunization with the adjuvant LTA1 (derived from the A1 domain of E. coli heat labile toxin) admixed with OmpX from K. pneumoniae can elicit antigen specific lung Th17 TRM cells that provide serotype independent immunity to members of the Enterobacteriaceae family. However, the upstream requirements to generate these cells are unclear. Single-cell RNA-seq showed that vaccine-elicited Th17 TRM cells expressed high levels of IL-1R1, suggesting that IL-1 family members may be critical to generate these cells. Using a combination of genetic and antibody neutralization approaches, we show that Th17 TRM cells can be generated independent of caspase-1 but are compromised when IL-1α is neutralized. Moreover IL-1α could serve as a molecular adjuvant to generate lung Th17 TRM cells independent of LTA1. Taken together, these data suggest that IL-1α plays a major role in vaccine-mediated lung Th17 TRM generation.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Akhilesh Srivastava
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Haoran Yang
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Naoki Iwanaga
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - T Parks Remcho
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jenny L Hewes
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Rayshma Sharoff
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kejing Song
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth B Norton
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Janet E McCombs
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
4
|
Wan M, Yang X, Chen Z, Su W, Cai L, Hou A, Sun B, Zhang Y, Kong W, Jiang C, Zhou Y. Comparison of Effects of Multiple Adjuvants and Immunization Routes on the Immunogenicity and Protection of HSV-2 gD Subunit Vaccine. Immunol Lett 2023:S0165-2478(23)00097-4. [PMID: 37290556 DOI: 10.1016/j.imlet.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/22/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
Genital herpes caused by herpes simplex virus type 2 (HSV-2) poses a global health issue. HSV-2 infection increases the risk of acquiring HIV infection. Studies have demonstrated that HSV-2 subunit vaccines have potential benefits, but require adjuvants to induce a balanced Th1/Th2 response. To develop a novel, effective vaccine, in this study, a truncated glycoprotein D (aa 1-285) of HSV-2 was formulated with an Al(OH)3 adjuvant, three squalene adjuvants, MF59, AS03, and AS02, or a mucosal adjuvant, bacterium-like particles (BLPs). The immunogenicity of these subunit vaccines was evaluated in mice. After three immunizations, vaccines formulated with Al(OH)3, MF59, AS03, and AS02 (intramuscularly) induced higher titers of neutralizing antibody than that formulated without adjuvant, and in particular, mice immunized with the vaccine plus AS02 had the highest neutralizing antibody titers and tended to produce a more balanced immune reaction than others. Intranasal gD2-PA-BLPs also induced excellent IgA levels and a more balanced Th1 and Th2 responses than intranasal gD2. After challenge with a lethal dose of HSV-2, all five adjuvants exhibited a positive effect in improving the survival rate. AS02 and gD2-PA-BLPs enhanced survival by 50% and 25%, respectively, when compared with the vaccine without adjuvant. AS02 was the only adjuvant that resulted in complete vaginal virus clearance and genital lesion healing within eight days. These results demonstrate the potential of using AS02 as a subunit vaccine adjuvant, and BLPs as a mucosal vaccine adjuvant.
Collapse
Affiliation(s)
- Mingming Wan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xiao Yang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zhijun Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Weiheng Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ali Hou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bo Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yan Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
5
|
Yi EJ, Kim YI, Song JH, Ko HJ, Chang SY. Intranasal immunization with curdlan induce Th17 responses and enhance protection against enterovirus 71. Vaccine 2023; 41:2243-2252. [PMID: 36863926 DOI: 10.1016/j.vaccine.2023.01.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/28/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023]
Abstract
Mucosal surfaces are in contact with the external environment and protect the body from infection by various microbes. To prevent infectious diseases at the first line of defense, the establishment of pathogen-specific mucosal immunity by mucosal vaccine delivery is needed. Curdlan, a 1,3-β-glucan has a strong immunostimulatory effect when delivered as a vaccine adjuvant. Here, we investigated whether intranasal administration of curdlan and antigen (Ag) could induce sufficient mucosal immune responses and protect against viral infections. Intranasal co-administration of curdlan and OVA increased OVA-specific IgG and IgA Abs in both serum and mucosal secretions. In addition, intranasal co-administration of curdlan and OVA induced the differentiation of OVA-specific Th1/Th17 cells in the draining lymph nodes. To investigate the protective immunity of curdlan against viral infection, intranasal co-administration of curdlan with recombinant VP1 of EV71 C4a was administered and showed enhanced protection against enterovirus 71 in a passive serum transfer model using neonatal hSCARB2 mice, although intranasal administration of VP1 plus curdlan increased VP1-specific helper T cells responses but not mucosal IgA. Next, Mongolian gerbils were intranasally immunized with curdlan plus VP1, and they had effective protection against EV71 C4a infection, while decreasing viral infection and tissue damage by inducing Th17 responses. These results indicated that intranasal curdlan with Ag improved Ag-specific protective immunity by enhancing mucosal IgA and Th17 against viral infection. Our results suggest that curdlan is an advantageous candidate as a mucosal adjuvant and delivery vehicle for the development of mucosal vaccines.
Collapse
Affiliation(s)
- Eun-Je Yi
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Young-In Kim
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea; AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea.
| |
Collapse
|
6
|
Lee C, Lee H, Park JC, Im SH. Microbial Components and Effector Molecules in T Helper Cell Differentiation and Function. Immune Netw 2023; 23:e7. [PMID: 36911805 PMCID: PMC9995987 DOI: 10.4110/in.2023.23.e7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/12/2023] [Accepted: 02/12/2023] [Indexed: 03/07/2023] Open
Abstract
The mammalian intestines harbor trillions of commensal microorganisms composed of thousands of species that are collectively called gut microbiota. Among the microbiota, bacteria are the predominant microorganism, with viruses, protozoa, and fungi (mycobiota) making up a relatively smaller population. The microbial communities play fundamental roles in the maturation and orchestration of the immune landscape in health and disease. Primarily, the gut microbiota modulates the immune system to maintain homeostasis and plays a crucial role in regulating the pathogenesis and pathophysiology of inflammatory, neuronal, and metabolic disorders. The microbiota modulates the host immune system through direct interactions with immune cells or indirect mechanisms such as producing short-chain acids and diverse metabolites. Numerous researchers have put extensive efforts into investigating the role of microbes in immune regulation, discovering novel immunomodulatory microbial species, identifying key effector molecules, and demonstrating how microbes and their key effector molecules mechanistically impact the host immune system. Consequently, recent studies suggest that several microbial species and their immunomodulatory molecules have therapeutic applicability in preclinical settings of multiple disorders. Nonetheless, it is still unclear why and how a handful of microorganisms and their key molecules affect the host immunity in diverse diseases. This review mainly discusses the role of microbes and their metabolites in T helper cell differentiation, immunomodulatory function, and their modes of action.
Collapse
Affiliation(s)
- Changhon Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Haena Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - John Chulhoon Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education, Yonsei University, Seoul 03722, Korea
- ImmunoBiome Inc., Pohang 37673, Korea
| |
Collapse
|
7
|
Paroli M, Caccavale R, Fiorillo MT, Spadea L, Gumina S, Candela V, Paroli MP. The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology. Pathogens 2022; 11:pathogens11121547. [PMID: 36558881 PMCID: PMC9781511 DOI: 10.3390/pathogens11121547] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
T-helper 17 (Th17) cells represent a subpopulation of CD4+ T lymphocytes that play an essential role in defense against pathogens. Th17 cells are distinguished from Th1 and Th2 cells by their ability to produce members of the interleukin-17 (IL-17) family, namely IL-17A and IL-17F. IL-17 in turn induces several target cells to synthesize and release cytokines, chemokines, and metalloproteinases, thereby amplifying the inflammatory cascade. Th17 cells reside predominantly in the lamina propria of the mucosa. Their main physiological function is to maintain the integrity of the mucosal barrier against the aggression of infectious agents. However, in an appropriate inflammatory microenvironment, Th17 cells can transform into immunopathogenic cells, giving rise to inflammatory and autoimmune diseases. This review aims to analyze the complex mechanisms through which the interaction between Th17 and pathogens can be on the one hand favorable to the host by protecting it from infectious agents, and on the other hand harmful, potentially generating autoimmune reactions and tissue damage.
Collapse
Affiliation(s)
- Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Stefano Gumina
- Department of Anatomy, Histology, Legal Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| | - Vittorio Candela
- Department of Anatomy, Histology, Legal Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
8
|
Network Pharmacology and Molecular Docking Analysis Reveal Insights into the Molecular Mechanism of Shengma-Gegen Decoction on Monkeypox. Pathogens 2022; 11:pathogens11111342. [DOI: 10.3390/pathogens11111342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Background: A new viral outbreak caused by monkeypox has appeared after COVID-19. As of yet, no specific drug has been found for its treatment. Shengma-Gegen decoction (SMGGD), a pathogen-eliminating and detoxifying agent composed of four kinds of Chinese herbs, has been demonstrated to be effective against several viruses in China, suggesting that it may be effective in treating monkeypox, however, the precise role and mechanisms are still unknown. Methods: Network pharmacology was used to investigate the monkeypox-specific SMGGD targets. These targets were analyzed via String for protein-to-protein interaction (PPI), followed by identification of hub genes with Cytoscape software. Function enrichment analysis of the hub targets was performed. The interactions between hub targets and corresponding ligands were validated via molecular docking. Results: Through screening and analysis, a total of 94 active components and 8 hub targets were identified in the TCM-bioactive compound-hub gene network. Molecular docking results showed that the active components of SMGGD have strong binding affinity for their corresponding targets. According to functional analysis, these hub genes are mainly involved in the TNF, AGE-RAGE, IL-17, and MAPK pathways, which are linked to the host inflammatory response to infection and viral replication. Therefore, SMGGD might suppress the replication of monkeypox virus through the MAPK signaling pathway while also reducing inflammatory damage caused by viral infection. Conclusion: SMGGD may have positive therapeutic effects on monkeypox by reducing inflammatory damage and limiting virus replication.
Collapse
|
9
|
Collins MK, McCutcheon CR, Petroff MG. Impact of Estrogen and Progesterone on Immune Cells and Host–Pathogen Interactions in the Lower Female Reproductive Tract. THE JOURNAL OF IMMUNOLOGY 2022; 209:1437-1449. [DOI: 10.4049/jimmunol.2200454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/05/2022] [Indexed: 11/05/2022]
|
10
|
Adachi A, Honda T. Regulatory Roles of Estrogens in Psoriasis. J Clin Med 2022; 11:jcm11164890. [PMID: 36013129 PMCID: PMC9409683 DOI: 10.3390/jcm11164890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/06/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease of the interleukin (IL)-23/IL-17 axis. The severity of psoriasis has been reported as higher in men than in women. The immunoregulatory role of female sex hormones has been proposed to be one of the factors responsible for sex differences. Among female sex hormones, estrogens have been suggested to be significantly involved in the development of psoriasis by various epidemiological and in vitro studies. For example, the severity of psoriasis is inversely correlated with serum estrogen levels. In vitro, estrogens suppress the production of psoriasis-related cytokines such as IL-1β and IL-23 from neutrophils and dendritic cells, respectively. Furthermore, a recent study using a mouse psoriasis model indicated the inhibitory role of estrogens in psoriatic dermatitis by suppressing IL-1β production from neutrophils and macrophages. Understanding the role and molecular mechanisms of female sex hormones in psoriasis may lead to better control of the disease.
Collapse
Affiliation(s)
- Akimasa Adachi
- Department of Dermatology, Tokyo Metropolitan Bokutoh Hospital, Tokyo 130-8575, Japan
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Tetsuya Honda
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Correspondence:
| |
Collapse
|
11
|
Cervantes O, Talavera IC, Every E, Coler B, Li M, Li A, Li H, Adams Waldorf K. Role of hormones in the pregnancy and sex-specific outcomes to infections with respiratory viruses. Immunol Rev 2022; 308:123-148. [PMID: 35373371 PMCID: PMC9189035 DOI: 10.1111/imr.13078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023]
Abstract
Pregnant women infected with pathogenic respiratory viruses, such as influenza A viruses (IAV) and coronaviruses, are at higher risk for mortality, hospitalization, preterm birth, and stillbirth. Several factors are likely to contribute to the susceptibility of pregnant individuals to severe lung disease including changes in pulmonary physiology, immune defenses, and effector functions of some immune cells. Pregnancy is also a physiologic state characterized by higher levels of multiple hormones that may impact the effector functions of immune cells, such as progesterone, estrogen, human chorionic gonadotropin, prolactin, and relaxin. Each of these hormones acts to support a tolerogenic immune state of pregnancy, which helps prevent fetal rejection, but may also contribute to an impaired antiviral response. In this review, we address the unique role of adaptive and innate immune cells in the control of pathogenic respiratory viruses and how pregnancy and specific hormones can impact their effector actions. We highlight viruses with sex-specific differences in infection outcomes and why pregnancy hormones may contribute to fetal protection but aid the virus at the expense of the mother's health.
Collapse
Affiliation(s)
- Orlando Cervantes
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Irene Cruz Talavera
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emma Every
- University of Washington School of Medicine, Spokane, Washington, United States of America
| | - Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, United States of America
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Biological Sciences, Columbia University, New York City, New York, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Case Western Reserve, Cleveland, Ohio, United States of America
| | - Hanning Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
12
|
Chandrasekar SS, Phanse Y, Riel M, Hildebrand RE, Hanafy M, Osorio JE, Abdelgayed SS, Talaat AM. Systemic Neutralizing Antibodies and Local Immune Responses Are Critical for the Control of SARS-CoV-2. Viruses 2022; 14:v14061262. [PMID: 35746733 PMCID: PMC9227431 DOI: 10.3390/v14061262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Antibody measurements are primarily used to evaluate experimental and approved COVID-19 vaccines, which is unilateral considering our immune responses’ complex nature. Previously, we showed that nanoparticle plasmid DNA adjuvant system, QAC, and MVA based vaccines were immunogenic against SARS-CoV-2. Here, we report on the protective efficacy of systemic humoral and mucosal cell-mediated immune responses in transgenic mice models against SARS-CoV-2 following nanoparticle immunization. Parenteral, intramuscular administration of QAC-based plasmid DNA vaccine-encoding SARS-CoV-2 S and N led to the induction of significant serum neutralizing humoral responses, which reduced viral burden in the lungs and prevented viral dissemination to the brain. In contrast, the mucosal, intranasal administration of a heterologous vaccine elicited significant mucosal cell-mediated immune responses in the lungs that limited lung viral replication. The presented results demonstrate that serum neutralizing humoral and local lung T-cell immune responses are critical for the control of SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Shaswath S. Chandrasekar
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | | | - Mariah Riel
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | - Rachel E. Hildebrand
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | - Mostafa Hanafy
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Colombia Wisconsin One Health Consortium, Universidad Nacional Medellín, Calle 75#79a 5, Colombia
| | - Sherein S. Abdelgayed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Adel M. Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Pan Genome Systems, Madison, WI 53719, USA;
- Correspondence: ; Tel.: +1-608-262-2861
| |
Collapse
|
13
|
Bagri P, Anipindi VC, Kaushic C. The Role of IL-17 During Infections in the Female Reproductive Tract. Front Immunol 2022; 13:861444. [PMID: 35493460 PMCID: PMC9046847 DOI: 10.3389/fimmu.2022.861444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/22/2022] [Indexed: 11/15/2022] Open
Abstract
Interleukin-17 (IL-17A) is a cytokine involved in a complex array of both protective and detrimental processes. Although early biological studies focused on the pro-inflammatory function of IL-17 in the context of autoimmune and inflammatory disorders, it has become increasingly evident that the roles of IL-17 are far more nuanced. Recent work has demonstrated that the functions of IL-17 are highly context- and tissue-dependent, and there is a fine balance between the pathogenic and protective functions of IL-17. This is especially evident in mucosal tissues such as the female reproductive tract, where IL-17 has been shown to play an important role in the immune response generated during fungal, bacterial and viral infections associated with protection, but also with inflammation. In this review, we discuss the evolving landscape of IL-17 biology within the context of the vaginal mucosa, focusing on key findings that highlight the importance of this cytokine in genital mucosal immunity.
Collapse
Affiliation(s)
- Puja Bagri
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Varun C. Anipindi
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- *Correspondence: Charu Kaushic,
| |
Collapse
|
14
|
Paiva IA, Familiar-Macedo D, Badolato-Corrêa J, Carvalho FR, Dias HG, Pauvolid-Corrêa A, dos Santos CF, Silva AA, de Azeredo EL, Vianna RADO, Cardoso CAA, Grifoni A, Sette A, Weiskopf D, de-Oliveira-Pinto LM. Involvement of Th1Th17 Cell Subpopulations in the Immune Responses of Mothers Who Gave Birth to Children with Congenital Zika Syndrome (CZS). Viruses 2022; 14:v14020250. [PMID: 35215843 PMCID: PMC8879837 DOI: 10.3390/v14020250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
High levels of T helper 17 cell (Th17)-related cytokines have been shown in acute Zika virus (ZIKV) infection. We hypothesized that the high levels of Th17-related cytokines, associated with a regulatory environment during pregnancy, create a favorable milieu for the differentiation of CD4+Th17 cells. We present data from a cross-sectional study on mothers who confirmed ZIKV infection by qRT-PCR and their children. We also recruited non-pregnant women infected with ZIKV in the same period. ZIKV infection occurred between 2015 and 2017. We collected samples for this study between 2018 and 2019, years after the initial infection. We highlight that, after in vitro stimulation with ZIKV CD4 megapool (ZIKV MP), we found a lower frequency of IL-17-producing CD4+ T cells (Th17), especially in the mothers, confirmed by the decrease in IL-17 production in the supernatant. However, a higher frequency of CD4+ IL-17+ IFN-γ+ T cells (Th1Th17) responding to the ZIKV MP was observed in the cells of the mothers and children but not in those of the non-pregnant women. Our data indicate that the priming of CD4 T cells of the Th1Th17 phenotype occurred preferentially in the mothers who gave birth to children with CZS and in the children.
Collapse
Affiliation(s)
- Iury Amancio Paiva
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Débora Familiar-Macedo
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Jéssica Badolato-Corrêa
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Fabiana Rabe Carvalho
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
| | - Helver Gonçalves Dias
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Alex Pauvolid-Corrêa
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA;
- Laboratory of Respiratory Viruses and Measles, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Caroline Fernandes dos Santos
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Andréa Alice Silva
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
| | - Elzinandes Leal de Azeredo
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | | | - Claudete Aparecida Araújo Cardoso
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
- Department of Maternal and Child, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil;
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
| | - Luzia Maria de-Oliveira-Pinto
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
- Correspondence:
| |
Collapse
|
15
|
Del Rio L, Murcia-Belmonte A, Buendía AJ, Navarro JA, Ortega N, Alvarez D, Salinas J, Caro MR. Effect of Female Sex Hormones on the Immune Response against Chlamydia abortus and on Protection Conferred by an Inactivated Experimental Vaccine in a Mouse Model. Pathogens 2022; 11:pathogens11010093. [PMID: 35056041 PMCID: PMC8781621 DOI: 10.3390/pathogens11010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
Mice are valuable models extensively used to test vaccine candidates against Chlamydia abortus and to clarify immunopathological mechanisms of the bacteria. As this pathogen has the ability to reactivate during pregnancy, it is important to deepen the knowledge and understanding of some of the effects of female hormones on immunity and vaccination. This study is aimed at describing the role of sex hormones in the pathology of OEA during chlamydial clearance using ovariectomised mice and also gaining an understanding of how 17β-oestradiol or progesterone may impact the effectiveness of vaccination. Animals were treated with sex hormones and infected with C. abortus, and the kinetics of infection and immune response were analysed by means of bacterial isolation, histopathology, and immunohistochemistry. In a second phase of the study, protection conferred by an experimental vaccine after hormone treatment was assessed. Oestradiol showed a stimulatory effect on the immune response during infection, with a more efficient recruitment of macrophages and T-cells at the infection site. Furthermore, after vaccination, oestradiol-treated animals showed a stronger protection against infection, indicating that this hormone has a positive effect, stimulating a specific memory response to the pathogen.
Collapse
Affiliation(s)
- Laura Del Rio
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
- Correspondence:
| | - Antonio Murcia-Belmonte
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
| | - Antonio Julián Buendía
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.J.B.); (J.A.N.)
| | - Jose Antonio Navarro
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.J.B.); (J.A.N.)
| | - Nieves Ortega
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
| | - Daniel Alvarez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
| | - Jesús Salinas
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
| | - María Rosa Caro
- Departamento de Sanidad Animal, Facultad de Veterinaria, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, 30100 Murcia, Spain; (A.M.-B.); (N.O.); (D.A.); (J.S.); (M.R.C.)
| |
Collapse
|
16
|
Li Y, Liu T, Chen G, Wang L, Guo A, Li Z, Pan L, Mao L, Luo X. Th17 cell differentiation induced by cytopathogenic biotype BVDV-2 in bovine PBLCs. BMC Genomics 2021; 22:884. [PMID: 34872498 PMCID: PMC8650399 DOI: 10.1186/s12864-021-08194-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background Bovine viral diarrhea virus (BVDV) is a major pathogen that causes bovine viral diarrhea/mucosal disease (BVD-MD), which has become a global infectious disease due to its wide spread and the lack of effective treatment. The process of BVDV infection is complex. Once infected, host immune cells are activated and modulated. As a major immune cell, peripheral blood lymphocyte cells (PBLCs) are the primary target of BVDV. In order to further understand the mechanism of BVDV- host interaction, the expression profiles of host lymphocytes mRNAs associated with BVDV infection were investigated by transcriptomic sequencing analysis. Results The transcriptomic sequencing analysis was performed on bovine PBLCs infected with CP BVDV-2 GS2018 after 12 h of infection. Gene expression profiling demonstrated that 1052 genes were differentially expressed in GS2018 infected PBLCs compared with the control group. Of these genes, 485 genes were up-regulated and 567 were down-regulated. The 19 differential expressed genes (DEGs) were selected for validation using quantitative real-time PCR and the results were consistent with the results of RNA-Seq. Gene ontology enrichment and KEGG pathway analysis showed that 1052 DEGs were significantly enriched in 16 pathways, including cytokine-cytokine receptor interaction, IL17, PI3K-Akt, MAPK and TNF signaling pathway. PPI network analysis showed that IL17A, IFN-γ and TNF-α interacted with various proteins and may play crucial roles in BVDV-2 infection. Of note, we confirmed that GS2018 induced Th17 cell differentiation in PBLCs and persistently increased the expression levels of IL17A. In turn, the replication of GS2018 was inhibited by IL17A. Conclusion In this study, the transcription changes of DEGs related to host immune responses in bovine PBLCs were caused by CP BVDV-2 infection. In particular, the effector molecules IL17A of Th17 cells were significantly up-regulated, which inhibited viral replication. These results will contribute to exploration and further understanding of the host immune response mechanism and interaction between host and BVDV-2. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08194-w.
Collapse
Affiliation(s)
- Yanping Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Tingli Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Guoliang Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Liqun Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Aimin Guo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Zhi Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Li Mao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China.
| | - Xuenong Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China.
| |
Collapse
|
17
|
Network Pharmacology-Based Systematic Analysis of Molecular Mechanisms of Geranium wilfordii Maxim for HSV-2 Infection. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:1009551. [PMID: 34777530 PMCID: PMC8580655 DOI: 10.1155/2021/1009551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/03/2021] [Accepted: 10/13/2021] [Indexed: 11/18/2022]
Abstract
Background Being a traditional Chinese medicine, Geranium wilfordii Maxim (GWM) is used for the treatment of various infectious diseases, and its main active ingredients are the polyphenolic substances such as polyphenols quercetin, corilagin, and geraniin. Previous studies have demonstrated the anti-HSV-1 viral activity of these three main ingredients. Through employing a network pharmacological method, the authors of the present research intend to probe the mechanism of GWM for the therapeutic treatment of HSV-2 infection. Methods The bioactive substances and related targets of GWM were obtained from the TCMSP database. Gene expression discrepancy for HSV-2 infection was obtained from dataset GSE18527. Crossover genes between disease target genes and GWM target genes were gained via Circos package. Distinctively displayed genes (DDGs) during HSV-2 infection were uploaded to the Metascape database with GWM target genes for further analysis. The tissue-specific distribution of the genes was obtained by uploading the genes to the PaGenBase database. Ingredient-gene-pathway (IGP) networks were constructed using Cytoscape software. Molecular docking investigations were carried out utilizing AutoDock Vina software. Results Nine actively involved components were retrieved from the TCMSP database. After taking the intersection among 153 drug target genes and 83 DDGs, 7 crossover genes were screened. Gene enrichment analysis showed that GWM treatment of HSV-2 infection mainly involves cytokine signaling in the immune system, response to virus, epithelial cell differentiation, and type II interferon signaling (IFNG). One hub, three core objectives, and two critical paths were filtered out from the built network. Geraniin showed strong binding activity with HSV-2 gD protein and STING protein in molecular docking. Conclusions This network pharmacological study provides a fundamental molecular mechanistic exploration of GWM for the treatment of HSV-2 infection.
Collapse
|
18
|
VanBenschoten HM, Woodrow KA. Vaginal delivery of vaccines. Adv Drug Deliv Rev 2021; 178:113956. [PMID: 34481031 PMCID: PMC8722700 DOI: 10.1016/j.addr.2021.113956] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/06/2021] [Accepted: 08/28/2021] [Indexed: 11/22/2022]
Abstract
Recent estimates suggest that one in two sexually active individuals will acquire a sexually transmitted infection by age 25, an alarming statistic that amounts to over 1 million new infections per day worldwide. Vaccination against STIs is highly desirable for alleviating this global burden of disease. Vaginal immunization is a promising strategy to combat transmission via the vaginal mucosa. The vagina is typically considered a poor inductive site for common correlates of adaptive immunity. However, emerging evidence suggests that immune tolerance may be overcome by precisely engineered vaccination schemes that orchestrate cell-mediated immunity and establish tissue resident memory immune cells. In this review, we will discuss the unique immunological milieu of the vaginal mucosa and our current understanding of correlates of pathogenesis and protection for several common STIs. We then present a summary of recent vaginal vaccine studies and explore the role that mucosal adjuvants and delivery systems play in enhancing protection according to requisite features of immunity. Finally, we offer perspectives on the challenges and future directions of vaginal vaccine delivery, discussing remaining physiological barriers and innovative vaccine formulations that may overcome them.
Collapse
Affiliation(s)
- Hannah M VanBenschoten
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
19
|
Luo SD, Chiu TJ, Chen WC, Wang CS. Sex Differences in Otolaryngology: Focus on the Emerging Role of Estrogens in Inflammatory and Pro-Resolving Responses. Int J Mol Sci 2021; 22:ijms22168768. [PMID: 34445474 PMCID: PMC8395901 DOI: 10.3390/ijms22168768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 12/02/2022] Open
Abstract
Otolaryngology (also known as ear, nose, and throat (ENT)) diseases can be significantly affected by the level of sex hormones, which indicates that sex differences affect the manifestation, pathophysiology, and outcomes of these diseases. Recently, increasing evidence has suggested that proinflammatory responses in ENT diseases are linked to the level of sex hormones. The sex hormone receptors are present on a wide variety of immune cells; therefore, it is evident that they play crucial roles in regulating the immune system and hence affect the disease progression of ENT diseases. In this review, we focus on how sex hormones, particularly estrogens, regulate ENT diseases, such as chronic rhinosinusitis, vocal fold polyps, thyroid cancer, Sjögren’s syndrome, and head and neck cancers, from the perspectives of inflammatory responses and specialized proresolving mediator-driven resolution. This paper aims to clarify why considering sex differences in the field of basic and medical research on otolaryngology is a key component to successful therapy for both males and females in the future.
Collapse
Affiliation(s)
- Sheng-Dean Luo
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (S.-D.L.); (W.-C.C.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Tai-Jan Chiu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Wei-Chih Chen
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (S.-D.L.); (W.-C.C.)
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-227-361-661 (ext. 5166)
| |
Collapse
|
20
|
Zenobia C, Herpoldt KL, Freire M. Is the oral microbiome a source to enhance mucosal immunity against infectious diseases? NPJ Vaccines 2021; 6:80. [PMID: 34078913 PMCID: PMC8172910 DOI: 10.1038/s41541-021-00341-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/24/2021] [Indexed: 12/14/2022] Open
Abstract
Mucosal tissues act as a barrier throughout the oral, nasopharyngeal, lung, and intestinal systems, offering first-line protection against potential pathogens. Conventionally, vaccines are applied parenterally to induce serotype-dependent humoral response but fail to drive adequate mucosal immune protection for viral infections such as influenza, HIV, and coronaviruses. Oral mucosa, however, provides a vast immune repertoire against specific microbial pathogens and yet is shaped by an ever-present microbiome community that has co-evolved with the host over thousands of years. Adjuvants targeting mucosal T-cells abundant in oral tissues can promote soluble-IgA (sIgA)-specific protection to confer increased vaccine efficacy. Th17 cells, for example, are at the center of cell-mediated immunity and evidence demonstrates that protection against heterologous pathogen serotypes is achieved with components from the oral microbiome. At the point of entry where pathogens are first encountered, typically the oral or nasal cavity, the mucosal surfaces are layered with bacterial cohabitants that continually shape the host immune profile. Constituents of the oral microbiome including their lipids, outer membrane vesicles, and specific proteins, have been found to modulate the Th17 response in the oral mucosa, playing important roles in vaccine and adjuvant designs. Currently, there are no approved adjuvants for the induction of Th17 protection, and it is critical that this research is included in the preparedness for the current and future pandemics. Here, we discuss the potential of oral commensals, and molecules derived thereof, to induce Th17 activity and provide safer and more predictable options in adjuvant engineering to prevent emerging infectious diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Freire
- Departments of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, USA.
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Paiva IA, Badolato-Corrêa J, Familiar-Macedo D, de-Oliveira-Pinto LM. Th17 Cells in Viral Infections-Friend or Foe? Cells 2021; 10:cells10051159. [PMID: 34064728 PMCID: PMC8151546 DOI: 10.3390/cells10051159] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Th17 cells are recognized as indispensable in inducing protective immunity against bacteria and fungi, as they promote the integrity of mucosal epithelial barriers. It is believed that Th17 cells also play a central role in the induction of autoimmune diseases. Recent advances have evaluated Th17 effector functions during viral infections, including their critical role in the production and induction of pro-inflammatory cytokines and in the recruitment and activation of other immune cells. Thus, Th17 is involved in the induction both of pathogenicity and immunoprotective mechanisms seen in the host's immune response against viruses. However, certain Th17 cells can also modulate immune responses, since they can secrete immunosuppressive factors, such as IL-10; these cells are called non-pathogenic Th17 cells. Here, we present a brief review of Th17 cells and highlight their involvement in some virus infections. We cover these notions by highlighting the role of Th17 cells in regulating the protective and pathogenic immune response in the context of viral infections. In addition, we will be describing myocarditis and multiple sclerosis as examples of immune diseases triggered by viral infections, in which we will discuss further the roles of Th17 cells in the induction of tissue damage.
Collapse
|
22
|
Chandrasekar SS, Phanse Y, Hildebrand RE, Hanafy M, Wu CW, Hansen CH, Osorio JE, Suresh M, Talaat AM. Localized and Systemic Immune Responses against SARS-CoV-2 Following Mucosal Immunization. Vaccines (Basel) 2021; 9:132. [PMID: 33562141 PMCID: PMC7914464 DOI: 10.3390/vaccines9020132] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
The rapid transmission of SARS-CoV-2 in the USA and worldwide necessitates the development of multiple vaccines to combat the COVID-19 global pandemic. Previously, we showed that a particulate adjuvant system, quil-A-loaded chitosan (QAC) nanoparticles, can elicit robust immunity combined with plasmid vaccines when used against avian coronavirus. Here, we report on the immune responses elicited by mucosal homologous plasmid and a heterologous immunization strategy using a plasmid vaccine and a Modified Vaccinia Ankara (MVA) expressing SARS-CoV-2 spike (S) and nucleocapsid (N) antigens. Only the heterologous intranasal immunization strategy elicited neutralizing antibodies against SARS-CoV-2 in serum and bronchoalveolar lavage of mice, suggesting a protective vaccine. The same prime/boost strategy led to the induction of type 1 and type 17 T-cell responses and polyfunctional T-cells expressing multiple type 1 cytokines (e.g., IFN-γ, TNFα, IL-2) in the lungs and spleens of vaccinated mice. In contrast, the plasmid homologous vaccine strategy led to the induction of local mono and polyfunctional T-cells secreting IFN-γ. Outcomes of this study support the potential of QAC-nano vaccines to elicit significant mucosal immune responses against respiratory coronaviruses.
Collapse
Affiliation(s)
- Shaswath S. Chandrasekar
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | | | - Rachel E. Hildebrand
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Mostafa Hanafy
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Chia-Wei Wu
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Chungyi H. Hansen
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Colombia Wisconsin One Health Consortium, Universidad Nacional Medellín, Calle 75#79a 5, Colombia
| | - M. Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Adel M. Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Pan Genome Systems, Madison, WI 53719, USA;
| |
Collapse
|
23
|
Khan N, Chen X, Geiger JD. Possible Therapeutic Use of Natural Compounds Against COVID-19. JOURNAL OF CELLULAR SIGNALING 2021; 2:63-79. [PMID: 33768214 PMCID: PMC7990267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The outbreak of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has led to coronavirus disease-19 (COVID-19); a pandemic disease that has resulted in devastating social, economic, morbidity and mortality burdens. SARS-CoV-2 infects cells following receptor-mediated endocytosis and priming by cellular proteases. Following uptake, SARS-CoV-2 replicates in autophagosome-like structures in the cytosol following its escape from endolysosomes. Accordingly, the greater endolysosome pathway including autophagosomes and the mTOR sensor may be targets for therapeutic interventions against SARS-CoV-2 infection and COVID-19 pathogenesis. Naturally existing compounds (phytochemicals) through their actions on endolysosomes and mTOR signaling pathways might provide therapeutic relief against COVID-19. Here, we discuss evidence that some natural compounds through actions on the greater endolysosome system can inhibit SARS-CoV-2 infectivity and thereby might be repurposed for use against COVID-19.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|
24
|
Estradiol Enhances Antiviral CD4 + Tissue-Resident Memory T Cell Responses following Mucosal Herpes Simplex Virus 2 Vaccination through an IL-17-Mediated Pathway. J Virol 2020; 95:JVI.01206-20. [PMID: 33028712 DOI: 10.1128/jvi.01206-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/30/2020] [Indexed: 12/31/2022] Open
Abstract
Estradiol (E2) is a sex hormone which has been shown to be protective against sexually transmitted infections such as herpes simplex virus 2 (HSV-2). However, few studies have examined the underlying mechanisms by which this occurs. Here, we investigated the effect of E2 on the establishment of memory T cells post-intranasal immunization with HSV-2. CD4+ T cell responses first appeared in the upper respiratory tract (URT) within 3 days postimmunization before being detected in the female reproductive tract (FRT) at 7 days. E2 treatment resulted in greater and earlier Th17 responses, which preceded augmented Th1 responses at these sites. The CD4+ T cells persisted in the URT for up to 28 days, and E2 treatment resulted in higher frequencies of memory T cells. Intranasal immunization also led to the establishment of CD4+ tissue-resident memory T cells (TRM cells) in the FRT, and E2 treatment resulted in increased Th1 and Th17 TRM cells. When the migration of circulating T cells into the FRT was blocked by FTY720, immunized E2-treated mice remained completely protected against subsequent genital HSV-2 challenge compared to non-E2 controls, confirming that TRM cells alone are adequate for protection in these mice. Finally, the enhanced vaginal Th1 TRM cells present in E2-treated mice were found to be modulated through an interleukin 17 (IL-17)-mediated pathway, as E2-treated IL-17A-deficient mice had impaired establishment of Th1 TRM cells. This study describes a novel role for E2 in enhancing CD4+ memory T cells and provides insight on potential strategies for generating optimal immunity during vaccination.IMPORTANCE Herpes simplex virus 2 (HSV-2) is a highly prevalent sexually transmitted infection for which there is currently no vaccine available. Interestingly, the female sex hormone estradiol has been shown to be protective against HSV-2. However, the underlying mechanisms by which this occurs remains relatively unknown. Our study demonstrates that under the influence of estradiol treatment, intranasal immunization with an attenuated strain of HSV-2 leads to enhanced establishment of antiviral memory T cell responses in the upper respiratory tract and female reproductive tract. In these sites, estradiol treatment leads to greater Th17 memory cells, which precede enhanced Th1 memory responses. Consequently, the T cell responses mounted by tissue-resident memory cells in the female reproductive tract of estradiol-treated mice are sufficient to protect mice against vaginal HSV-2 challenge. This study offers important insights regarding the regulation of mucosal immunity by hormones and on potential strategies for generating optimal immunity during vaccination.
Collapse
|
25
|
Khan N. Possible protective role of 17β-estradiol against COVID-19. JOURNAL OF ALLERGY AND INFECTIOUS DISEASES 2020; 1:38-48. [PMID: 33196058 PMCID: PMC7665224 DOI: 10.46439/allergy.1.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is the virus that causes coronavirus disease 2019 (COVID-19); a worldwide pandemic as declared by the World Health Organization (WHO). SARS-CoV-2 appears to infect cells by first binding and priming its viral-spike proteins with membrane-associated angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). Through the coordinated actions of ACE2 and TMPRSS2, SARS-CoV-2 spike proteins fuse with plasma membranes and ultimately the virus enters cells. ACE2 is integral to the renin-angiotensin-aldosterone system (RAAS), and SARS-CoV-2 down-regulates protein expression levels of ACE2. Once infected, patients typically develop acute respiratory distress syndrome (ARDS) and a number of other severe complications that result in a high rate of fatality, especially in older (>60 years) adults and in people with pre-existing medical conditions. Data now indicate clearly that among people of all age groups, COVID-19 fatalities are higher in men than women. Here, attention is focused on these sex differences and posit a role of estrogen in these differences as well as possible therapeutic and protective actions of 17β-estradiol against COVID-19.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|
26
|
Jiménez-Saiz R, Anipindi VC, Galipeau H, Ellenbogen Y, Chaudhary R, Koenig JF, Gordon ME, Walker TD, Mandur TS, Abed S, Humbles A, Chu DK, Erjefält J, Ask K, Verdú EF, Jordana M. Microbial Regulation of Enteric Eosinophils and Its Impact on Tissue Remodeling and Th2 Immunity. Front Immunol 2020; 11:155. [PMID: 32117293 PMCID: PMC7033414 DOI: 10.3389/fimmu.2020.00155] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Eosinophils have emerged as multifaceted cells that contribute to tissue homeostasis. However, the impact of the microbiota on their frequency and function at mucosal sites remains unclear. Here, we investigated the role of the microbiota in the regulation of enteric eosinophils. We found that small intestinal (SI) eosinophilia was significantly greater in germ-free (GF) mice compared to specific pathogen free (SPF) controls. This was associated with changes in the production of enteric signals that regulate eosinophil attraction and survival, and was fully reversed by complex colonization. Additionally, SI eosinophils of GF mice exhibited more cytoplasmic protrusions and less granule content than SPF controls. Lastly, we generated a novel strain of eosinophil-deficient GF mice. These mice displayed intestinal fibrosis and were less prone to allergic sensitization as compared to GF controls. Overall, our study demonstrates that commensal microbes regulate intestinal eosinophil frequency and function, which impacts tissue repair and allergic sensitization to food antigens. These data support a critical interplay between the commensal microbiota and intestinal eosinophils in shaping homeostatic, innate, and adaptive immune processes in health and disease.
Collapse
Affiliation(s)
- Rodrigo Jiménez-Saiz
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
- Department of Immunology & Oncology, National Center for Biotechnology (CNB)-CSIC, Madrid, Spain
| | - Varun C. Anipindi
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Heather Galipeau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Yosef Ellenbogen
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Roopali Chaudhary
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Joshua F. Koenig
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Melissa E. Gordon
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Tina D. Walker
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Talveer S. Mandur
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Soumeya Abed
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Alison Humbles
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Gaithersburg, MD, United States
| | - Derek K. Chu
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonas Erjefält
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Kjetil Ask
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| | - Elena F. Verdú
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Manel Jordana
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, ON, Canada
| |
Collapse
|
27
|
Lauzon-Joset JF, Mincham KT, Abad AP, Short BP, Holt PG, Strickland DH, Leffler J. Oestrogen amplifies pre-existing atopy-associated Th2 bias in an experimental asthma model. Clin Exp Allergy 2019; 50:391-400. [PMID: 31785105 DOI: 10.1111/cea.13544] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/03/2019] [Accepted: 11/27/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The prevalence and severity of asthma, particularly the most common (atopic) form of the disease, increase amongst females but not males after puberty, and asthma activity also changes throughout the menstrual cycle and during pregnancy. The contribution of female sex hormones to asthma pathogenesis is incompletely understood. OBJECTIVE To obtain insight into the role of oestrogen (E2) in experimental atopic asthma, and guide future research on sex-related variations in atopic asthma susceptibility/intensity in humans. METHODS We utilized an experimental model comprising rat strains expressing dichotomous Th2-high vs Th2-low immunophenotypes exemplified by eosinophilia, mirroring differences between human atopics/non-atopics. We compared the efficiency of Th2-associated immunoinflammatory mechanisms, which differed markedly between the two strains, and between sexes in the Th2-high strain, and determined the effects of E2 administration on these differences. RESULTS Unique to the Th2-high strain, eosinophil: neutrophil ratios in the airways at baseline and following sensitization/aeroallergen challenge were logfold higher in females relative to males, and this was reflected by higher baseline blood eosinophil numbers in females. Pretreatment of Th2-high males with E2 abrogated this sex difference by selectively boosting Th2-associated genes in the airways and eosinophilia, but was without corresponding effect in the Th2-low strain. In contrast, parallel E2 effects on myeloid and lymphoid cell populations were relatively modest. CONCLUSIONS AND CLINICAL RELEVANCE E2 acts to amplify the eosinophilic component of pre-existing Th2-high immunophenotype, possibly acting at the level of the common eosinophil/neutrophil precursor in bone marrow to preferentially drive eosinophil differentiation. Constitutive granulocyte profiles in which the balance between eosinophils and neutrophils is skewed towards eosinophils have been identified in independent cohort studies as markers of asthma risk, and these findings suggest that more detailed studies on the role of E2 in this context, and in relation to asthma pathogenesis in post-pubertal females in particular, appear warranted.
Collapse
Affiliation(s)
- Jean Francois Lauzon-Joset
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Kyle T Mincham
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Ana P Abad
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Braden P Short
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Deborah H Strickland
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
28
|
Schiffer JT, Gottlieb SL. Biologic interactions between HSV-2 and HIV-1 and possible implications for HSV vaccine development. Vaccine 2019; 37:7363-7371. [PMID: 28958807 PMCID: PMC5867191 DOI: 10.1016/j.vaccine.2017.09.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
Development of a safe and effective vaccine against herpes simplex virus type 2 (HSV-2) has the potential to limit the global burden of HSV-2 infection and disease, including genital ulcer disease and neonatal herpes, and is a global sexual and reproductive health priority. Another important potential benefit of an HSV-2 vaccine would be to decrease HIV infections, as HSV-2 increases the risk of HIV-1 acquisition several-fold. Acute and chronic HSV-2 infection creates ulcerations and draws dendritic cells and activated CD4+ T cells into genital mucosa. These cells are targets for HIV entry and replication. Prophylactic HSV-2 vaccines (to prevent infection) and therapeutic vaccines (to modify or treat existing infections) are currently under development. By preventing or modifying infection, an effective HSV-2 vaccine could limit HSV-associated genital mucosal inflammation and thus HIV risk. However, a vaccine might have competing effects on HIV risk depending on its mechanism of action and cell populations generated in the genital mucosa. In this article, we review biologic interactions between HSV-2 and HIV-1, consider HSV-2 vaccine development in the context of HIV risk, and discuss implications and research needs for future HSV vaccine development.
Collapse
Affiliation(s)
- Joshua T Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, United States; Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States; University of Washington, Department of Medicine, Seattle, WA, United States.
| | - Sami L Gottlieb
- World Health Organization, Department of Reproductive Health and Research, Geneva, Switzerland
| |
Collapse
|
29
|
Wessels JM, Lajoie J, Cooper MIJH, Omollo K, Felker AM, Vitali D, Dupont HA, Nguyen PV, Mueller K, Vahedi F, Kimani J, Oyugi J, Cheruiyot J, Mungai JN, Deshiere A, Tremblay MJ, Mazzulli T, Stearns JC, Ashkar AA, Fowke KR, Surette MG, Kaushic C. Medroxyprogesterone acetate alters the vaginal microbiota and microenvironment in women and increases susceptibility to HIV-1 in humanized mice. Dis Model Mech 2019; 12:dmm.039669. [PMID: 31537512 PMCID: PMC6826019 DOI: 10.1242/dmm.039669] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
The hormonal contraceptive medroxyprogesterone acetate (MPA) is associated with increased risk of human immunodeficiency virus (HIV), via incompletely understood mechanisms. Increased diversity in the vaginal microbiota modulates genital inflammation and is associated with increased HIV-1 acquisition. However, the effect of MPA on diversity of the vaginal microbiota is relatively unknown. In a cohort of female Kenyan sex workers, negative for sexually transmitted infections (STIs), with Nugent scores <7 (N=58 of 370 screened), MPA correlated with significantly increased diversity of the vaginal microbiota as assessed by 16S rRNA gene sequencing. MPA was also significantly associated with decreased levels of estrogen in the plasma, and low vaginal glycogen and α-amylase, factors implicated in vaginal colonization by lactobacilli, bacteria that are believed to protect against STIs. In a humanized mouse model, MPA treatment was associated with low serum estrogen, low glycogen and enhanced HIV-1 susceptibility. The mechanism by which the MPA-mediated changes in the vaginal microbiota may contribute to HIV-1 susceptibility in humans appears to be independent of inflammatory cytokines and/or activated T cells. Altogether, these results suggest MPA-induced hypo-estrogenism may alter key metabolic components that are necessary for vaginal colonization by certain bacterial species including lactobacilli, and allow for greater bacterial diversity in the vaginal microbiota. This article has an associated First Person interview with the first author of the paper. Summary: MPA may increase susceptibility to HIV-1 in sex workers through the suppression of endogenous estrogen, reducing vaginal glycogen and α-amylase levels, which increases bacterial diversity, potentially reducing protective bacterial species such as lactobacilli.
Collapse
Affiliation(s)
- Jocelyn M Wessels
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Julie Lajoie
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Maeve I J Hay Cooper
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Kenneth Omollo
- Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Allison M Felker
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Danielle Vitali
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Haley A Dupont
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Philip V Nguyen
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Kristen Mueller
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Joshua Kimani
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya.,Kenyan AIDS Control Program, P.O. Box 19361 - 00202, Nairobi, Kenya
| | - Julius Oyugi
- Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | | | - John N Mungai
- Kenyan AIDS Control Program, P.O. Box 19361 - 00202, Nairobi, Kenya
| | - Alexandre Deshiere
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec City, Québec G1V 4G2, Canada.,Department of Microbiology and Immunology Medical Biology, Université Laval, Québec City, Québec G1V 0A6, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec City, Québec G1V 4G2, Canada.,Department of Microbiology and Immunology Medical Biology, Université Laval, Québec City, Québec G1V 0A6, Canada
| | - Tony Mazzulli
- Public Health Laboratories, Public Health Ontario, Toronto, Ontario M5G 1V2, Canada.,Mount Sinai Hospital/University Health Network, Department of Microbiology, Toronto, Ontario M5G 1X5, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jennifer C Stearns
- Department of Medicine, Farncombe Family Digestive Health Institute, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Keith R Fowke
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Michael G Surette
- Department of Medicine, Farncombe Family Digestive Health Institute, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,McMaster Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,McMaster Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
30
|
Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia. J Virol 2019; 93:JVI.00814-19. [PMID: 31375576 DOI: 10.1128/jvi.00814-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/24/2019] [Indexed: 12/21/2022] Open
Abstract
BLT (bone marrow-liver-thymus) humanized mice, which reconstitute a functional human immune system, develop prototypic human virus-specific CD8+ T cell responses following infection with human immunodeficiency virus type 1 (HIV-1). We explored the utility of the BLT model for HIV-1 vaccine development by immunizing BLT mice against the conserved viral Gag protein, utilizing a rapid prime-boost protocol of poly(lactic-co-glycolic) acid microparticles and a replication-defective herpes simplex virus (HSV) recombinant vector. After HIV-1 challenge, the mice developed broad, proteome-wide gamma interferon-positive (IFN-γ+) T cell responses against HIV-1 that reached magnitudes equivalent to what is observed in HIV-1-infected individuals. The functionality of these responses was underscored by the consistent emergence of escape mutations in multiple CD8+ T cell epitopes during the course of infection. Although prechallenge vaccine-induced responses were largely undetectable, the Gag immunization increased both the magnitude and the kinetics of anamnestic Gag-specific T cell responses following HIV-1 infection, and the magnitude of these postchallenge Gag-specific responses was inversely correlated with acute HIV-1 viremia. Indeed, Gag immunization was associated with a modest but significant 0.5-log reduction in HIV-1 viral load when analyzed across four experimental groups of BLT mice. Notably, the HSV vector induced elevated plasma concentrations of polarizing cytokines and chemotactic factors, including interleukin-12p70 (IL-12p70) and MIP-1α, which were positively correlated with the magnitude of Gag-specific responses. Overall, these results support the ability of BLT mice to recapitulate human pathogen-specific T cell responses and to respond to immunization; however, additional improvements to the model are required to develop a robust system for testing HIV-1 vaccine efficacy.IMPORTANCE Advances in the development of humanized mice have raised the possibility of a small-animal model for preclinical testing of an HIV-1 vaccine. Here, we describe the capacity of BLT humanized mice to mount broadly directed HIV-1-specific human T cell responses that are functionally active, as indicated by the rapid emergence of viral escape mutations. Although immunization of BLT mice with the conserved viral Gag protein did not result in detectable prechallenge responses, it did increase the magnitude and kinetics of postchallenge Gag-specific T cell responses, which was associated with a modest but significant reduction in acute HIV-1 viremia. Additionally, the BLT model revealed immunization-associated increases in the plasma concentrations of immunomodulatory cytokines and chemokines that correlated with more robust T cell responses. These data support the potential utility of the BLT humanized mouse for HIV-1 vaccine development but suggest that additional improvements to the model are warranted.
Collapse
|
31
|
Ma WT, Yao XT, Peng Q, Chen DK. The protective and pathogenic roles of IL-17 in viral infections: friend or foe? Open Biol 2019; 9:190109. [PMID: 31337278 PMCID: PMC6685926 DOI: 10.1098/rsob.190109] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Viral infections cause substantial human morbidity and mortality, and are a significant health burden worldwide. Following a viral infection, the host may initiate complex antiviral immune responses to antagonize viral invasion and replication. However, proinflammatory antiviral immune responses pose a great threat to the host if not properly held in check. Interleukin (IL)-17 is a pleiotropic cytokine participating in a variety of physiological and pathophysiological conditions, including tissue integrity maintenance, cancer progression, autoimmune disease development and, more intriguingly, infectious diseases. Abundant evidence suggests that while IL-17 plays a crucial role in enhancing effective antiviral immune responses, it may also promote and exacerbate virus-induced illnesses. Accumulated experimental and clinical evidence has broadened our understanding of the seemingly paradoxical role of IL-17 in viral infections and suggests that IL-17-targeted immunotherapy may be a promising therapeutic option. Herein, we summarize current knowledge regarding the protective and pathogenic roles of IL-17 in viral infections, with emphasis on underlying mechanisms. The various and critical roles of IL-17 in viral infections necessitate the development of therapeutic strategies that are uniquely tailored to both the infectious agent and the infection environment.
Collapse
Affiliation(s)
- Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - Xiao-Ting Yao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - Qun Peng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| |
Collapse
|
32
|
Tognarelli EI, Palomino TF, Corrales N, Bueno SM, Kalergis AM, González PA. Herpes Simplex Virus Evasion of Early Host Antiviral Responses. Front Cell Infect Microbiol 2019; 9:127. [PMID: 31114761 PMCID: PMC6503643 DOI: 10.3389/fcimb.2019.00127] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/10/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex viruses type 1 (HSV-1) and type 2 (HSV-2) have co-evolved with humans for thousands of years and are present at a high prevalence in the population worldwide. HSV infections are responsible for several illnesses including skin and mucosal lesions, blindness and even life-threatening encephalitis in both, immunocompetent and immunocompromised individuals of all ages. Therefore, diseases caused by HSVs represent significant public health burdens. Similar to other herpesviruses, HSV-1 and HSV-2 produce lifelong infections in the host by establishing latency in neurons and sporadically reactivating from these cells, eliciting recurrences that are accompanied by viral shedding in both, symptomatic and asymptomatic individuals. The ability of HSVs to persist and recur in otherwise healthy individuals is likely given by the numerous virulence factors that these viruses have evolved to evade host antiviral responses. Here, we review and discuss molecular mechanisms used by HSVs to evade early innate antiviral responses, which are the first lines of defense against these viruses. A comprehensive understanding of how HSVs evade host early antiviral responses could contribute to the development of novel therapies and vaccines to counteract these viruses.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomás F Palomino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Corrales
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
33
|
Pepe G, Locati M, Della Torre S, Mornata F, Cignarella A, Maggi A, Vegeto E. The estrogen-macrophage interplay in the homeostasis of the female reproductive tract. Hum Reprod Update 2019; 24:652-672. [PMID: 30256960 DOI: 10.1093/humupd/dmy026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/10/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Estrogens are known to orchestrate reproductive events and to regulate the immune system during infections and following tissue damage. Recent findings suggest that, in the absence of any danger signal, estrogens trigger the physiological expansion and functional specialization of macrophages, which are immune cells that populate the female reproductive tract (FRT) and are increasingly being recognized to participate in tissue homeostasis beyond their immune activity against infections. Although estrogens are the only female gonadal hormones that directly target macrophages, a comprehensive view of this endocrine-immune communication and its involvement in the FRT is still missing. OBJECTIVE AND RATIONALE Recent accomplishments encourage a revision of the literature on the ability of macrophages to respond to estrogens and induce tissue-specific functions required for reproductive events, with the aim to envision macrophages as key players in FRT homeostasis and mediators of the regenerative and trophic actions of estrogens. SEARCH METHODS We conducted a systematic search using PubMed and Ovid for human, animal (rodents) and cellular studies published until 2018 on estrogen action in macrophages and the activity of these cells in the FRT. OUTCOMES Our search identified the remarkable ability of macrophages to activate biochemical processes in response to estrogens in cell culture experiments. The distribution at specific locations, interaction with selected cells and acquisition of distinct phenotypes of macrophages in the FRT, as well as the cyclic renewal of these properties at each ovarian cycle, demonstrate the involvement of these cells in the homeostasis of reproductive events. Moreover, current evidence suggests an association between estrogen-macrophage signaling and the generation of a tolerant and regenerative environment in the FRT, although a causative link is still missing. WIDER IMPLICATIONS Dysregulation of the functions and estrogen responsiveness of FRT macrophages may be involved in infertility and estrogen- and macrophage-dependent gynecological diseases, such as ovarian cancer and endometriosis. Thus, more research is needed on the physiology and pharmacological control of this endocrine-immune interplay.
Collapse
Affiliation(s)
- Giovanna Pepe
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Segrate, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via fratelli Cervi, Segrate, Italy
| | - Sara Della Torre
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Federica Mornata
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padua, Largo Meneghetti 2, Padua, Italy
| | - Adriana Maggi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti, 9 Milan, Italy
| |
Collapse
|
34
|
Voigt EA, Ovsyannikova IG, Kennedy RB, Grill DE, Goergen KM, Schaid DJ, Poland GA. Sex Differences in Older Adults' Immune Responses to Seasonal Influenza Vaccination. Front Immunol 2019; 10:180. [PMID: 30873150 PMCID: PMC6400991 DOI: 10.3389/fimmu.2019.00180] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Sex differences in immune responses to influenza vaccine may impact efficacy across populations. Methods: In a cohort of 138 older adults (50-74 years old), we measured influenza A/H1N1 antibody titers, B-cell ELISPOT response, PBMC transcriptomics, and PBMC cell compositions at 0, 3, and 28 days post-immunization with the 2010/11 seasonal inactivated influenza vaccine. Results: We identified higher B-cell ELISPOT responses in females than males. Potential mechanisms for sex effects were identified in four gene clusters related to T, NK, and B cells. Mediation analysis indicated that sex-dependent expression in T and NK cell genes can be partially attributed to higher CD4+ T cell and lower NK cell fractions in females. We identified strong sex effects in 135 B cell genes whose expression correlates with ELISPOT measures, and found that cell subset differences did not explain the effect of sex on these genes' expression. Post-vaccination expression of these genes, however, mediated 41% of the sex effect on ELISPOT responses. Conclusions: These results improve our understanding of sexual dimorphism in immunity and influenza vaccine response.
Collapse
Affiliation(s)
- Emily A. Voigt
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | | | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | - Diane E. Grill
- Division of Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Krista M. Goergen
- Division of Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Daniel J. Schaid
- Division of Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
35
|
Wessels JM, Felker AM, Dupont HA, Kaushic C. The relationship between sex hormones, the vaginal microbiome and immunity in HIV-1 susceptibility in women. Dis Model Mech 2018; 11:dmm035147. [PMID: 30154116 PMCID: PMC6177003 DOI: 10.1242/dmm.035147] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The role of sex hormones in regulating immune responses in the female genital tract has been recognized for decades. More recently, it has become increasingly clear that sex hormones regulate susceptibility to sexually transmitted infections through direct and indirect mechanisms involving inflammation and immune responses. The reproductive cycle can influence simian/human immunodeficiency virus (SHIV) infections in primates and HIV-1 infection in ex vivo cervical tissues from women. Exogenous hormones, such as those found in hormonal contraceptives, have come under intense scrutiny because of the increased susceptibility to sexually transmitted infections seen in women using medroxyprogesterone acetate, a synthetic progestin-based contraceptive. Recent meta-analyses concluded that medroxyprogesterone acetate enhanced HIV-1 susceptibility in women by 40%. In contrast, estradiol-containing hormonal contraceptives were not associated with increased susceptibility and some studies reported a protective effect of estrogen on HIV/SIV infection, although the underlying mechanisms remain incompletely understood. Recent studies describe a key role for the vaginal microbiota in determining susceptibility to sexually transmitted infections, including HIV-1. While Lactobacillus spp.-dominated vaginal microbiota is associated with decreased susceptibility, complex microbiota, such as those seen in bacterial vaginosis, correlates with increased susceptibility to HIV-1. Interestingly, sex hormones are inherently linked to microbiota regulation in the vaginal tract. Estrogen has been postulated to play a key role in establishing a Lactobacillus-dominated microenvironment, whereas medroxyprogesterone acetate is linked to hypo-estrogenic effects. The aim of this Review is to contribute to a better understanding of the sex-hormone-microbiome-immunity axis, which can provide key information on the determinants of HIV-1 susceptibility in the female genital tract and, consequently, inform HIV-1 prevention strategies.
Collapse
Affiliation(s)
- Jocelyn M Wessels
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Allison M Felker
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Haley A Dupont
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
36
|
Leffler J, Stumbles PA, Strickland DH. Immunological Processes Driving IgE Sensitisation and Disease Development in Males and Females. Int J Mol Sci 2018; 19:E1554. [PMID: 29882879 PMCID: PMC6032271 DOI: 10.3390/ijms19061554] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 01/15/2023] Open
Abstract
IgE sensitisation has increased significantly over the last decades and is a crucial factor in the development of allergic diseases. IgE antibodies are produced by B cells through the process of antigen presentation by dendritic cells, subsequent differentiation of CD4⁺ Th2 cells, and class switching in B cells. However, many of the factors regulating these processes remain unclear. These processes affect males and females differently, resulting in a significantly higher prevalence of IgE sensitisation in males compared to females from an early age. Before the onset of puberty, this increased prevalence of IgE sensitisation is also associated with a higher prevalence of clinical symptoms in males; however, after puberty, females experience a surge in the incidence of allergic symptoms. This is particularly apparent in allergic asthma, but also in other allergic diseases such as food and contact allergies. This has been partly attributed to the pro- versus anti-allergic effects of female versus male sex hormones; however, it remains unclear how the expression of sex hormones translates IgE sensitisation into clinical symptoms. In this review, we describe the recent epidemiological findings on IgE sensitisation in male and females and discuss recent mechanistic studies casting further light on how the expression of sex hormones may influence the innate and adaptive immune system at mucosal surfaces and how sex hormones may be involved in translating IgE sensitisation into clinical manifestations.
Collapse
Affiliation(s)
- Jonatan Leffler
- Telethon Kids Institute, The University of Western Australia, 100 Roberts Rd, Subiaco, WA 6008, Australia.
| | - Philip A Stumbles
- Telethon Kids Institute, The University of Western Australia, 100 Roberts Rd, Subiaco, WA 6008, Australia.
- School of Paediatrics and Child Health, The University of Western Australia, Subiaco, WA 6008, Australia.
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia.
| | - Deborah H Strickland
- Telethon Kids Institute, The University of Western Australia, 100 Roberts Rd, Subiaco, WA 6008, Australia.
| |
Collapse
|
37
|
Stanfield BA, Rider PJF, Caskey J, Del Piero F, Kousoulas KG. Intramuscular vaccination of guinea pigs with the live-attenuated human herpes simplex vaccine VC2 stimulates a transcriptional profile of vaginal Th17 and regulatory Tr1 responses. Vaccine 2018; 36:2842-2849. [PMID: 29655629 DOI: 10.1016/j.vaccine.2018.03.075] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/31/2022]
Abstract
Herpes simplex virus is a common causative agent of oral and genital diseases. Novel vaccines and therapeutics are needed to combat herpes infections especially after the failure of subunit vaccines in human clinical trials. We have shown that the live-attenuated HSV-1 VC2 vaccine strain is unable to establish latency in vaccinated animals and produces a robust immune response capable of completely protecting mice against lethal vaginal HSV-1 or HSV-2 infections. The guinea pig represents the best small animal model of genital HSV-2 disease. Reported here, twenty-one female Hartley guinea pigs received intramuscular injection with either the VC2 vaccine, or equal volume of conditioned tissue culture media. Animals received 2 booster vaccinations at 21 day intervals following the initial vaccination. After vaccination, animals were challenged with the highly virulent HSV-2 (G) strain. Histologically, VC2 vaccinated animals had little to no apparent inflammation/disease following challenge. Unvaccinated animals developed moderate to severe erosive and ulcerative vaginitis. Quantitative reverse-transcriptase PCR analysis in VC2 vaccinated and challenged animals identified transcriptional signatures of Th17 and regulatory Tr1 cells associated with the inflammatory response primed by VC2 vaccination. Treatment of cultured human vaginal epithelial cells (VK2 cells) with a combination of IL-17A and IL-22 resulted in the significant induction of beta-defensin 3 expression. Further, treatment of VK2 cells with IL-17A, IL-22, IL-36 or beta-defensin 3 resulted in diminished HSV-2 replication. Overall, these results suggest that intramuscular vaccination with the live-attenuated vaccine VC2 primes a mucosal immune response predisposing the adaptive expression of transcripts associated with a Th17 response to challenge and these responses contribute to antiviral immunity.
Collapse
Affiliation(s)
- Brent A Stanfield
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Paul J F Rider
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - John Caskey
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fabio Del Piero
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Konstantin G Kousoulas
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
38
|
Capone I, Marchetti P, Ascierto PA, Malorni W, Gabriele L. Sexual Dimorphism of Immune Responses: A New Perspective in Cancer Immunotherapy. Front Immunol 2018; 9:552. [PMID: 29619026 PMCID: PMC5871673 DOI: 10.3389/fimmu.2018.00552] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/05/2018] [Indexed: 12/17/2022] Open
Abstract
Nowadays, several types of tumors can benefit from the new frontier of immunotherapy, due to the recent increasing knowledge of the role of the immune system in cancer control. Among the new therapeutic strategies, there is the immune checkpoint blockade (ICB), able to restore an efficacious antitumor immunity and significantly prolong the overall survival (OS) of patients with advanced tumors such as melanoma and non-small cell lung cancer (NSCLC). Despite the impressive efficacy of these agents in some patients, treatment failure and resistance are frequently observed. In this regard, the signaling governed by IFN type I (IFN-I) has emerged as pivotal in orchestrating host defense. This pathway displays different activation between sexes, thus potentially contributing to sexual dimorphic differences in the immune responses to immunotherapy. This perspective article aims to critically consider the immune signals, with particular attention to IFN-I, that may differently affect female and male antitumor responses upon immunotherapy.
Collapse
Affiliation(s)
- Imerio Capone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Marchetti
- Department of Oncology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori Fondazione G. Pascale (IRCCS), Naples, Italy
| | - Walter Malorni
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
39
|
Hapgood JP, Kaushic C, Hel Z. Hormonal Contraception and HIV-1 Acquisition: Biological Mechanisms. Endocr Rev 2018; 39:36-78. [PMID: 29309550 PMCID: PMC5807094 DOI: 10.1210/er.2017-00103] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022]
Abstract
Access to effective affordable contraception is critical for individual and public health. A wide range of hormonal contraceptives (HCs), which differ in composition, concentration of the progestin component, frequency of dosage, and method of administration, is currently available globally. However, the options are rather limited in settings with restricted economic resources that frequently overlap with areas of high HIV-1 prevalence. The predominant contraceptive used in sub-Saharan Africa is the progestin-only three-monthly injectable depot medroxyprogesterone acetate. Determination of whether HCs affect HIV-1 acquisition has been hampered by behavioral differences potentially confounding clinical observational data. Meta-analysis of these studies shows a significant association between depot medroxyprogesterone acetate use and increased risk of HIV-1 acquisition, raising important concerns. No association was found for combined oral contraceptives containing levonorgestrel, nor for the two-monthly injectable contraceptive norethisterone enanthate, although data for norethisterone enanthate are limited. Susceptibility to HIV-1 and other sexually transmitted infections may, however, be dependent on the type of progestin present in the formulation. Several underlying biological mechanisms that may mediate the effect of HCs on HIV-1 and other sexually transmitted infection acquisition have been identified in clinical, animal, and ex vivo studies. A substantial gap exists in the translation of basic research into clinical practice and public health policy. To bridge this gap, we review the current knowledge of underlying mechanisms and biological effects of commonly used progestins. The review sheds light on issues critical for an informed choice of progestins for the identification of safe, effective, acceptable, and affordable contraceptive methods.
Collapse
Affiliation(s)
- Janet P Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Charu Kaushic
- Department of Pathology and Molecular Medicine, McMaster University, Ontario, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Zdenek Hel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
40
|
Capone I, Marchetti P, Ascierto PA, Malorni W, Gabriele L. Sexual Dimorphism of Immune Responses: A New Perspective in Cancer Immunotherapy. Front Immunol 2018. [PMID: 29619026 DOI: 10.3389/fimmu.2018.0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Nowadays, several types of tumors can benefit from the new frontier of immunotherapy, due to the recent increasing knowledge of the role of the immune system in cancer control. Among the new therapeutic strategies, there is the immune checkpoint blockade (ICB), able to restore an efficacious antitumor immunity and significantly prolong the overall survival (OS) of patients with advanced tumors such as melanoma and non-small cell lung cancer (NSCLC). Despite the impressive efficacy of these agents in some patients, treatment failure and resistance are frequently observed. In this regard, the signaling governed by IFN type I (IFN-I) has emerged as pivotal in orchestrating host defense. This pathway displays different activation between sexes, thus potentially contributing to sexual dimorphic differences in the immune responses to immunotherapy. This perspective article aims to critically consider the immune signals, with particular attention to IFN-I, that may differently affect female and male antitumor responses upon immunotherapy.
Collapse
Affiliation(s)
- Imerio Capone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Marchetti
- Department of Oncology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori Fondazione G. Pascale (IRCCS), Naples, Italy
| | - Walter Malorni
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
41
|
Lennard K, Dabee S, Barnabas SL, Havyarimana E, Blakney A, Jaumdally SZ, Botha G, Mkhize NN, Bekker LG, Lewis DA, Gray G, Mulder N, Passmore JAS, Jaspan HB. Microbial Composition Predicts Genital Tract Inflammation and Persistent Bacterial Vaginosis in South African Adolescent Females. Infect Immun 2018; 86:e00410-17. [PMID: 29038128 PMCID: PMC5736802 DOI: 10.1128/iai.00410-17] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/10/2017] [Indexed: 11/20/2022] Open
Abstract
Young African females are at an increased risk of HIV acquisition, and genital inflammation or the vaginal microbiome may contribute to this risk. We studied these factors in 168 HIV-negative South African adolescent females aged 16 to 22 years. Unsupervised clustering of 16S rRNA gene sequences revealed three clusters (subtypes), one of which was strongly associated with genital inflammation. In a multivariate model, the microbiome compositional subtype and hormonal contraception were significantly associated with genital inflammation. We identified 40 taxa significantly associated with inflammation, including those reported previously (Prevotella, Sneathia, Aerococcus, Fusobacterium, and Gemella) as well as several novel taxa (including increased frequencies of bacterial vaginosis-associated bacterium 1 [BVAB1], BVAB2, BVAB3, Prevotella amnii, Prevotella pallens, Parvimonas micra, Megasphaera, Gardnerella vaginalis, and Atopobium vaginae and decreased frequencies of Lactobacillus reuteri, Lactobacillus crispatus, Lactobacillus jensenii, and Lactobacillus iners). Women with inflammation-associated microbiomes had significantly higher body mass indices and lower levels of endogenous estradiol and luteinizing hormone. Community functional profiling revealed three distinct vaginal microbiome subtypes, one of which was characterized by extreme genital inflammation and persistent bacterial vaginosis (BV); this subtype could be predicted with high specificity and sensitivity based on the Nugent score (≥9) or BVAB1 abundance. We propose that women with this BVAB1-dominated subtype may have chronic genital inflammation due to persistent BV, which may place them at a particularly high risk for HIV infection.
Collapse
Affiliation(s)
- Katie Lennard
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Smritee Dabee
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Shaun L Barnabas
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Enock Havyarimana
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Anna Blakney
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Shameem Z Jaumdally
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gerrit Botha
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla N Mkhize
- National Institute for Communicable Diseases, Sandringham, Johannesburg, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - David A Lewis
- Western Sydney Sexual Health Centre, Parramatta, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
- Sydney Medical School-Westmead, University of Sydney, Sydney, Australia
- National Institute for Communicable Diseases, Sandringham, Johannesburg, South Africa
| | - Glenda Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Nicola Mulder
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Johannesburg, South Africa
| | - Heather B Jaspan
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
42
|
Novel Role for Interleukin-17 in Enhancing Type 1 Helper T Cell Immunity in the Female Genital Tract following Mucosal Herpes Simplex Virus 2 Vaccination. J Virol 2017; 91:JVI.01234-17. [PMID: 28956763 DOI: 10.1128/jvi.01234-17] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022] Open
Abstract
It is well established that interferon gamma (IFN-γ) production by CD4+ T cells is critical for antiviral immunity against herpes simplex virus 2 (HSV-2) genital infection. However, the role of interleukin-17A (IL-17A) production by CD4+ T cells in HSV-2 antiviral immunity is yet to be elucidated. Here we demonstrate that IL-17A plays an important role in enhancing antiviral T helper type 1 (Th1) responses in the female genital tract (FGT) and is essential for effective protection conferred by HSV-2 vaccination. While IL-17A did not play a critical role during primary genital HSV-2 infection, seen by lack of differences in susceptibility between IL-17A-deficient (IL-17A-/-) and wild-type (WT) C57BL/6 mice, it was critical for mediating antiviral responses after challenge/reexposure. Compared to WT mice, IL-17A-/- mice (i) infected intravaginally and reexposed or (ii) vaccinated intranasally and challenged intravaginally demonstrated poor outcomes. Following intravaginal HSV-2 reexposure or challenge, vaccinated IL-17A-/- mice had significantly higher mortality, greater disease severity, higher viral shedding, and higher levels of proinflammatory cytokines and chemokines in vaginal secretions. Furthermore, IL-17A-/- mice had impaired Th1 cell responses after challenge/reexposure, with significantly lower proportions of vaginal IFN-γ+ CD4+ T cells. The impaired Th1 cell responses in IL-17A-/- mice coincided with smaller populations of IFN-γ+ CD4+ tissue resident memory T (TRM) cells in the genital tract postimmunization. Taken together, these findings describe a novel role for IL-17A in regulating antiviral IFN-γ+ Th1 cell immunity in the vaginal tract. This strategy could be exploited to enhance antiviral immunity following HSV-2 vaccination.IMPORTANCE T helper type 1 (Th1) immunity, specifically interferon gamma (IFN-γ) production by CD4+ T cells, is critical for protection against genital herpesvirus (HSV-2) infection, and enhancing this response can potentially help improve disease outcomes. Our study demonstrated that interleukin-17A (IL-17A) plays an essential role in enhancing antiviral Th1 responses in the female genital tract (FGT). We found that in the absence of IL-17A, preexposed and vaccinated mice showed poor disease outcomes and were unable to overcome HSV-2 reexposure/challenge. IL-17A-deficient mice (IL-17A-/-) had smaller populations of IFN-γ+ CD4+ tissue resident memory T (TRM) cells in the genital tract postimmunization than did wild-type (WT) mice, which coincided with attenuated Th1 responses postchallenge. This has important implications for developing effective vaccines against HSV-2, as we propose that strategies inducing IL-17A in the genital tract may promote more effective Th1 cell immunity and better overall protection.
Collapse
|
43
|
Laffont S, Seillet C, Guéry JC. Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function. Front Immunol 2017; 8:108. [PMID: 28239379 PMCID: PMC5300975 DOI: 10.3389/fimmu.2017.00108] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, and the lineage-committing transcription factors shaping peripheral DC subsets have been identified. Both progenitor cells and mature DC subsets express estrogen receptors (ERs), which are ligand-dependent transcription factors. This suggests that estrogens may contribute to the reported sex differences in immunity by regulating DC biology. Here, we review the recent literature and highlight evidence that estrogen-dependent activation of ERα regulates the development or the functional responses of particular DC subsets. The in vitro model of GM-CSF-induced DC differentiation shows that CD11c+ CD11bint Ly6cneg cells depend on ERα activation by estrogen for their development, and for the acquisition of competence to activate naive CD4+ T lymphocytes and mount a robust pro-inflammatory cytokine response to CD40 stimulation. In this model, estrogen signaling in conjunction with GM-CSF is necessary to promote early interferon regulatory factor (Irf)-4 expression in macrophage-DC progenitors and their subsequent differentiation into IRF-4hi CD11c+ CD11bint Ly6cneg cells, closely related to the cDC2 subset. The Flt3L-induced model of DC differentiation in turn shows that ERα signaling promotes the development of conventional DC (cDC) and plasmacytoid DC (pDC) with higher capability of pro-inflammatory cytokine production in response to TLR stimulation. Likewise, cell-intrinsic ER signaling positively regulates the TLR-driven production of type I interferons (IFNs) in mouse pDCs in vivo. This effect of estrogens likely contributes to the greater proficiency of women's pDCs than men's as regards the production of type I IFNs elicited by TLR7 ligands. In summary, evidence is emerging in support of the notion that estrogen signaling regulates important aspects of cDC and pDC development and/or effector functions, in both mice and humans.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| | - Cyril Seillet
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| |
Collapse
|
44
|
Abstract
The maternal immune system is complex and governed by multiple hormonal and metabolic factors, including those provided to the mother via the fetus. Understanding of the balance between maternal tolerance and protection of the fetus may require thinking from multiple theoretical approaches to the general problem of immune activation and tolerance. This article provides a brief review of the immune system, with aspects relevant to pregnancy. The references include reviews that expand on the elements discussed. The article also uses different models of immune system activation and tolerance to provide a theoretical understanding of the problem of maternal tolerance.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Given Building Room C-246, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| |
Collapse
|
45
|
Abstract
The maternal immune system is complex and governed by multiple hormonal and metabolic factors, including those provided to the mother via the fetus. Understanding of the balance between maternal tolerance and protection of the fetus may require thinking from multiple theoretical approaches to the general problem of immune activation and tolerance. This article provides a brief review of the immune system, with aspects relevant to pregnancy. The references include reviews that expand on the elements discussed. The article also uses different models of immune system activation and tolerance to provide a theoretical understanding of the problem of maternal tolerance.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Given Building Room C-246, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| |
Collapse
|
46
|
Effects of Female Sex Hormones on Susceptibility to HSV-2 in Vaginal Cells Grown in Air-Liquid Interface. Viruses 2016; 8:v8090241. [PMID: 27589787 PMCID: PMC5035955 DOI: 10.3390/v8090241] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/05/2023] Open
Abstract
The lower female reproductive tract (FRT) is comprised of the cervix and vagina, surfaces that are continuously exposed to a variety of commensal and pathogenic organisms. Sexually transmitted viruses, such as herpes simplex virus type 2 (HSV-2), have to traverse the mucosal epithelial lining of the FRT to establish infection. The majority of current culture systems that model the host-pathogen interactions in the mucosal epithelium have limitations in simulating physiological conditions as they employ a liquid-liquid interface (LLI), in which both apical and basolateral surfaces are submerged in growth medium. We designed the current study to simulate in vivo conditions by growing an immortalized vaginal epithelial cell line (Vk2/E6E7) in culture with an air-liquid interface (ALI) and examined the effects of female sex hormones on their growth, differentiation, and susceptibility to HSV-2 under these conditions, in comparison to LLI cultures. ALI conditions induced Vk2/E6E7 cells to grow into multi-layered cultures compared to the monolayers present in LLI conditions. Vk2 cells in ALI showed higher production of cytokeratin in the presence of estradiol (E2), compared to cells grown in progesterone (P4). Cells grown under ALI conditions were exposed to HSV-2-green fluorescent protein (GFP) and the highest infection and replication was observed in the presence of P4. Altogether, this study suggests that ALI cultures more closely simulate the in vivo conditions of the FRT compared to the conventional LLI cultures. Furthermore, under these conditions P4 was found to confer higher susceptibility to HSV-2 infection in vaginal cells. The vaginal ALI culture system offers a better alternative to study host-pathogen interactions.
Collapse
|
47
|
Correction: Estradiol Enhances CD4+ T-Cell Anti-Viral Immunity by Priming Vaginal DCs to Induce Th17 Responses via an IL-1-Dependent Pathway. PLoS Pathog 2016; 12:e1005706. [PMID: 27276086 PMCID: PMC4898707 DOI: 10.1371/journal.ppat.1005706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|