1
|
Sanyaolu A, Marinkovic A, Prakash S, Balendra V, Hamdy K, Haider N, Abbasi AF, Hosein Z, Younis K, Smith S, Badaru O, Izurieta R. Impact of Prolonged SARS-CoV-2 Viral Shedding on COVID-19 Disease Outcome and Viral Dynamics. Indian J Microbiol 2024; 64:1416-1424. [PMID: 39678987 PMCID: PMC11645332 DOI: 10.1007/s12088-024-01238-1] [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: 03/29/2023] [Accepted: 02/26/2024] [Indexed: 12/17/2024] Open
Abstract
This article aimed to review the current literature on the impact of continuous shedding of the COVID-19 virus in infected patients in relation to disease outcome variables and viral dynamics. Electronic databases PubMed, Google Scholar, and MedlinePlus were searched using relevant keywords, restricting the selection to thirty-two peer-reviewed articles and four gray literatures from the WHO websites. Findings from this study showed that several variables such as sex, age, immune status, treatments, and vaccines were found to affect the outcomes associated with the COVID-19 virus shedding. These findings highlight the need for further research using longitudinal whole-genome sequencing of the virus and its variants to increase the understanding.
Collapse
Affiliation(s)
- Adekunle Sanyaolu
- D’Youville University, Buffalo, NY USA
- Federal Ministry of Health, Abuja, Nigeria
| | | | | | | | - Kareem Hamdy
- Saint James School of Medicine, The Quarter 2640, Anguilla
| | - Nafees Haider
- All Saints University School of Medicine, Roseau, Dominica
| | | | | | | | - Stella Smith
- Nigerian Institute of Medical Research, Lagos, Nigeria
| | | | - Ricardo Izurieta
- Global Communicable Diseases, College of Public Health, University of South Florida, Tampa, FL USA
- Universidad de las Américas, Quito, 170513 Ecuador
| |
Collapse
|
2
|
Willimann M, Grisch‐Chan HM, Rimann N, Rothgangl T, Hruzova M, Schwank G, Thöny B. Therapeutic liver cell transplantation to treat murine PKU. J Inherit Metab Dis 2024; 47:1322-1335. [PMID: 39449255 PMCID: PMC11586590 DOI: 10.1002/jimd.12802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024]
Abstract
For gene therapy of the liver, in vivo applications based on adeno-associated virus are the most advanced vectors despite limitations, including low efficacy and episomal loss, potential integration and safety issues, and high production costs. Alternative vectors and/or delivery routes are of high interest. The regenerative ability of the liver bears the potential for ex vivo therapy using liver cell transplantation for disease correction if provided with a selective advantage to expand and replace the existing cell mass. Here we present such treatment of a mouse model of human phenylketonuria (PKU). Primary hepatocytes from wild-type mice were gene modified in vitro (with a lentiviral vector) that carries a gene editing system (CRISPR) to inhibit Cypor. Cypor inactivation confers paracetamol (or acetaminophen) resistance to hepatocytes and thus a growth advantage to eliminate the pre-existing liver cells upon grafting (via the spleen) and exposure to repeated treatment with paracetamol. Grafting Cypor-inactivated wild-type hepatocytes into inbred young adult enu2 (PKU) mice, followed by selective expansion by paracetamol dosing, resulted in replacing up to 5% of cell mass, normalization of blood phenylalanine, and permanent correction of PKU. Hepatocyte transplantation offers thus an armamentarium of novel therapy options for genetic liver defects.
Collapse
Affiliation(s)
- Melanie Willimann
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Hiu Man Grisch‐Chan
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Nicole Rimann
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Tanja Rothgangl
- University of ZurichInstitute for Pharmacology and ToxicologyZurichSwitzerland
| | - Martina Hruzova
- University of ZurichInstitute for Pharmacology and ToxicologyZurichSwitzerland
| | - Gerald Schwank
- University of ZurichInstitute for Pharmacology and ToxicologyZurichSwitzerland
| | - Beat Thöny
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| |
Collapse
|
3
|
Ju Y, Zhang Z, Liu M, Lin S, Sun Q, Song Z, Liang W, Tong X, Jie Z, Lu H, Cai K, Chen P, Jin X, Zhang W, Xu X, Yang H, Wang J, Hou Y, Xiao L, Jia H, Zhang T, Guo R. Integrated large-scale metagenome assembly and multi-kingdom network analyses identify sex differences in the human nasal microbiome. Genome Biol 2024; 25:257. [PMID: 39380016 PMCID: PMC11463039 DOI: 10.1186/s13059-024-03389-2] [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: 08/27/2022] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Respiratory diseases impose an immense health burden worldwide. Epidemiological studies have revealed extensive disparities in the incidence and severity of respiratory tract infections between men and women. It has been hypothesized that there might also be a nasal microbiome axis contributing to the observed sex disparities. RESULTS Here, we study the nasal microbiome of healthy young adults in the largest cohort to date with 1593 individuals, using shotgun metagenomic sequencing. We compile the most comprehensive reference catalog for the nasal bacterial community containing 4197 metagenome-assembled genomes and integrate the mycobiome, to provide a valuable resource and a more holistic perspective for the understudied human nasal microbiome. We systematically evaluate sex differences and reveal extensive sex-specific features in both taxonomic and functional levels in the nasal microbiome. Through network analyses, we capture markedly higher ecological stability and antagonistic potentials in the female nasal microbiome compared to the male's. The analysis of the keystone bacteria reveals that the sex-dependent evolutionary characteristics might have contributed to these differences. CONCLUSIONS In summary, we construct the most comprehensive catalog of metagenome-assembled-genomes for the nasal bacterial community to provide a valuable resource for the understudied human nasal microbiome. On top of that, comparative analysis in relative abundance and microbial co-occurrence networks identify extensive sex differences in the respiratory tract community, which may help to further our understanding of the observed sex disparities in the respiratory diseases.
Collapse
Affiliation(s)
- Yanmei Ju
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhe Zhang
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
| | - Mingliang Liu
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shutian Lin
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Sun
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
- Department of Statistical Sciences, University of Toronto, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
| | | | - Weiting Liang
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Tong
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
| | - Zhuye Jie
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China
| | - Haorong Lu
- China National Genebank, BGI Research, Shenzhen, 518210, China
| | - Kaiye Cai
- BGI Research, Shenzhen, 518083, China
| | | | - Xin Jin
- BGI Research, Shenzhen, 518083, China
| | | | - Xun Xu
- BGI Research, Shenzhen, 518083, China
| | - Huanming Yang
- BGI Research, Shenzhen, 518083, China
- James D, Watson Institute of Genome Sciences, Hangzhou, 310013, China
| | - Jian Wang
- BGI Research, Shenzhen, 518083, China
| | - Yong Hou
- BGI Research, Shenzhen, 518083, China
| | - Liang Xiao
- BGI Research, Shenzhen, 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, 266555, China
| | - Huijue Jia
- School of Life Sciences, Fudan University, Shanghai, 200433, China.
- Greater Bay Area Institute of Precision Medicine, Guangzhou, 511458, China.
| | - Tao Zhang
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China.
- BGI Research, Wuhan, 430074, China.
| | - Ruijin Guo
- BGI Research, Shenzhen, 518083, China.
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI Research, Shenzhen, 518083, China.
- BGI Research, Wuhan, 430074, China.
| |
Collapse
|
4
|
Sirkis DW, Oddi AP, Jonson C, Bonham LW, Hoang PT, Yokoyama JS. The role of interferon signaling in neurodegeneration and neuropsychiatric disorders. Front Psychiatry 2024; 15:1480438. [PMID: 39421070 PMCID: PMC11484020 DOI: 10.3389/fpsyt.2024.1480438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Recent advances in transcriptomics research have uncovered heightened interferon (IFN) responses in neurodegenerative diseases including Alzheimer's disease, primary tauopathy, Parkinson's disease, TDP-43 proteinopathy, and related mouse models. Augmented IFN signaling is now relatively well established for microglia in these contexts, but emerging work has highlighted a novel role for IFN-responsive T cells in the brain and peripheral blood in some types of neurodegeneration. These findings complement a body of literature implicating dysregulated IFN signaling in neuropsychiatric disorders including major depression and post-traumatic stress disorder. In this review, we will characterize and integrate advances in our understanding of IFN responses in neurodegenerative and neuropsychiatric disease, discuss how sex and ancestry modulate the IFN response, and examine potential mechanistic explanations for the upregulation of antiviral-like IFN signaling pathways in these seemingly non-viral neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Daniel W. Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Alexis P. Oddi
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Caroline Jonson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, United States
- DataTecnica LLC, Washington, DC, United States
| | - Luke W. Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Phuong T. Hoang
- Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Jennifer S. Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
5
|
Johnson L, De Gascun CF, Hassan J. Investigation of SARS-CoV-2 IgG Binding Capability to Variants of the SARS-CoV-2 Virus. Viral Immunol 2024; 37:404-410. [PMID: 39263777 DOI: 10.1089/vim.2024.0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
The SARS-CoV-2 pandemic has confirmed that the ability to rapidly mutate may be extremely beneficial for a virus. Not long after the first wave, new variants emerged with altered infectivity, disease severity, and mortality. These new strains most notably had numerous mutations of the spike (S) protein, a surface protein responsible for binding to and entering the host cell. The Delta and Omicron strains demonstrated increased immune evasion and improved binding affinity to the host cell receptor, angiotensin-converting enzyme 2 (ACE2). This study examines the ability of wild-type SARS-CoV-2 IgG to bind Delta and Omicron antigens, as well as their functional binding capabilities to two different S-ACE2 complexes. Twenty SARS-CoV-2 positive samples from patients who had recovered from infection with ancestral SARS-CoV-2 in the first wave of COVID-19 and 10 pre-pandemic control samples were studied. SARS-CoV-2 exposed patients showed significantly higher levels of IgG to SARS-CoV-2 S1/RBD (p < 0.001), N protein (p < 0.001), and Omicron spike variant (p = 0.01), but not to Delta spike variant (p = 0.966) when compared with controls. Furthermore, patient samples showed significantly greater inhibition of SARS-CoV-2 S1/RBD and E484K spike to ACE2 binding (p < 0.001 and p = 0.015, respectively). Conversely, there was no correlation between the binding inhibition of S1/RBD and E484K spike to ACE2 receptor. This study shows there is considerable cross-reactivity of IgG generated by wild-type SARS-CoV-2 infection to the Delta and Omicron variants.
Collapse
Affiliation(s)
- Lucy Johnson
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
| | - Cillian F De Gascun
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
| | - Jaythoon Hassan
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Qin C, Liu Q, Wang Y, Deng J, Du M, Liu M, Liu J. Disease Burden and Geographic Inequalities in 15 Types of Neonatal Infectious Diseases in 131 Low- and Middle-Income Countries and Territories. HEALTH DATA SCIENCE 2024; 4:0186. [PMID: 39355853 PMCID: PMC11443844 DOI: 10.34133/hds.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 08/13/2024] [Indexed: 10/03/2024]
Abstract
Background: The burden of neonatal infections in low- and middle-income countries and territories (LMICs) is a critical public health challenge, while our understanding of specific burden and secular trends remains limited. Methods: We gathered annual data on 15 types of neonatal infections in LMICs from 1990 to 2019 from the Global Burden of Disease 2019. Numbers, rates, percent changes, and estimated annual percentage changes of incidence and deaths were calculated. We also explored the association between disease burden, socio-demographic index (SDI), and universal health coverage index (UHCI). Results: Enteric infections and upper respiratory infections owned the top highest incidence rates for neonates in 2019. Neonatal sepsis and other neonatal infections, as well as otitis media, demonstrated an increasing trend of incidence across all 3 low- and middle-income regions. The top 3 causes of neonatal mortality in 2019 were neonatal sepsis and other neonatal infections, lower respiratory infections, and enteric infections. Between 1990 and 2019, all of the neonatal infection-related mortality rates suggested an overall decline. Sex differences could be found in the incidence and mortality of some neonatal infections, but most disease burdens decreased more rapidly in males. SDI and UHCI were both negatively associated with most of the disease burden, but there were exceptions. Conclusions: Our study serves as a vital exploration into the realities of neonatal infectious diseases in LMICs. The identified trends and disparities not only provide a foundation for future research but also underscore the critical need for targeted policy initiatives to alleviate on a global scale.
Collapse
Affiliation(s)
- Chenyuan Qin
- School of Public Health, Peking University, Beijing, 100191, China
| | - Qiao Liu
- School of Public Health, Peking University, Beijing, 100191, China
| | - Yaping Wang
- School of Public Health, Peking University, Beijing, 100191, China
| | - Jie Deng
- School of Public Health, Peking University, Beijing, 100191, China
| | - Min Du
- School of Public Health, Peking University, Beijing, 100191, China
| | - Min Liu
- School of Public Health, Peking University, Beijing, 100191, China
| | - Jue Liu
- School of Public Health, Peking University, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, 100871, China
- National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China
- Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health, Peking University, Beijing, 100191, China
| |
Collapse
|
7
|
Anjidani N, Shahpari R, Kafi H, Petrovsky N, Barati S. Effects of age and gender on immunogenicity and reactogenicity of SpikoGen recombinant spike protein vaccine: a post-hoc analysis. Sci Rep 2024; 14:22631. [PMID: 39349494 PMCID: PMC11442574 DOI: 10.1038/s41598-024-67945-3] [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: 07/28/2023] [Accepted: 07/17/2024] [Indexed: 10/02/2024] Open
Abstract
SpikoGen® COVID-19 vaccine is based on the spike protein extracellular domain of the ancestral Wuhan-Hu-1 strain modified by removal of the furin cleavage site and addition of stabilising mutations expressed as a recombinant protein in insect cells. It is formulated with Advax-CpG55.2™ adjuvant to ensure optimal immunogenicity. In this study, data from several SpikoGen® clinical trials was retrospectively analysed to assess for any effect of gender or age on seroconversion, neutralizing antibody levels or the incidence of adverse events. Following the 1st dose, older age was associated with a reduced rate of fatigue (RR 0.97, p < 0.001), headache (RR 0.98, p = 0.034) and myalgia (RR 0.97, p=0.016), following the 2nd dose, the rate of fatigue (RR 0.98, p = 0.017) but following the 3rd dose no effect of age on adverse events was evident. Similarly, following the 1st dose, men reported a 19% lower incidence of fatigue, 36% lower incidence of headache and 28% lower incidence of myalgia when compared to women. Interestingly, there was no relationship between age or gender and serum neutralizing antibody levels, although after each vaccine dose there was a consistent trend to women having a higher seroconversion rate. There was no correlation between neutralizing antibody levels and adverse events. Unlike what is seen with mRNA vaccines, reactogenicity trended lower after each subsequent SpikoGen® dose. Overall, SpikoGen® exhibited positive immunogenicity and low reactogenicity, indicating that a low incidence of adverse events does not equate to poor immunogenicity. SpikoGen® remains a promising protein-based vaccine platform for COVID-19 protection.
Collapse
MESH Headings
- Humans
- Female
- Male
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Middle Aged
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- SARS-CoV-2/immunology
- Age Factors
- Adult
- Immunogenicity, Vaccine
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Sex Factors
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/administration & dosage
- Retrospective Studies
- Aged
- Myalgia
- Headache/chemically induced
- Fatigue/immunology
Collapse
Affiliation(s)
- Nassim Anjidani
- Medical Department, Orchid Pharmed Company, No. 12 Asad Abadi Street, 70th Alley, Tehran, 1969955311, Iran
| | - Ramin Shahpari
- Medical Department, Orchid Pharmed Company, No. 12 Asad Abadi Street, 70th Alley, Tehran, 1969955311, Iran
| | - Hamidreza Kafi
- Medical Department, Orchid Pharmed Company, No. 12 Asad Abadi Street, 70th Alley, Tehran, 1969955311, Iran
| | | | - Saghar Barati
- Medical Department, Orchid Pharmed Company, No. 12 Asad Abadi Street, 70th Alley, Tehran, 1969955311, Iran.
| |
Collapse
|
8
|
Chhabra A, Bashirians G, Petropoulos CJ, Wrin T, Paliwal Y, Henstock PV, Somanathan S, da Fonseca Pereira C, Winburn I, Rasko JE. Global seroprevalence of neutralizing antibodies against adeno-associated virus serotypes used for human gene therapies. Mol Ther Methods Clin Dev 2024; 32:101273. [PMID: 39022744 PMCID: PMC11253686 DOI: 10.1016/j.omtm.2024.101273] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Abstract
Adeno-associated virus (AAV) vectors are promising gene therapy candidates, but pre-existing anti-AAV neutralizing antibodies (NAbs) pose a significant challenge to successful gene delivery. Knowledge of NAb seroprevalence remains limited and inconsistent. We measured activity of NAbs against six clinically relevant AAV serotypes across 10 countries in adults (n = 502) and children (n = 50) using a highly sensitive transduction inhibition assay. NAb prevalence was generally highest for AAV1 and lowest for AAV5. There was considerable variability across countries and geographical regions. NAb prevalence increased with age and was higher in females, participants of Asian ethnicity, and participants in cancer trials. Co-prevalence was most frequently observed between AAV1 and AAV6 and less frequently between AAV5 and other AAVs. Machine learning analyses revealed a unique clustering of AAVs that differed from previous phylogenetic classifications. These results offer insights into the biological relationships between the immunogenicity of AAVs in humans beyond that observed previously using standard clades, which are based on linear capsid sequences. Our findings may inform improved vector design and facilitate the development of AAV vector-mediated clinical gene therapies.
Collapse
Affiliation(s)
| | | | | | - Terri Wrin
- Labcorp-Monogram Biosciences, South San Francisco, CA, USA
| | | | | | | | | | | | - John E.J. Rasko
- University of Sydney, Central Clinical School, Faculty of Medicine & Health, Sydney, NSW, Australia
- Department of Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Almuntashiri S, Dutta S, Zhu Y, Gamare S, Ramírez G, Irineo‐Moreno V, Camarena A, Regino N, Campero P, Hernández‐Cardenas CM, Rodriguez‐ Reyna TS, Zuñiga J, Owen CA, Wang X, Zhang D. Estrogen-dependent gene regulation: Molecular basis of TIMP-1 as a sex-specific biomarker for acute lung injury. Physiol Rep 2024; 12:e70047. [PMID: 39267201 PMCID: PMC11392656 DOI: 10.14814/phy2.70047] [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: 04/25/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Increased circulating tissue inhibitor of metalloproteinases-1 (TIMP-1) levels have been observed in patients with acute lung injury (ALI). However, the sex-specific regulation of TIMP-1 and the underlying molecular mechanisms have not been well elucidated. In this study, we found that plasma TIMP-1 levels were significantly higher in COVID-19 and H1N1 patients compared with those in healthy subjects (n = 25). TIMP-1 concentrations were significantly different between males and females in each disease group. Among female but not male patients, TIMP-1 levels significantly correlated with the PaO2/FiO2 ratio and hospital length of stay. Using the mouse model of ALI induced by the H1N1 virus, we found that TIMP-1 is strikingly induced in PDGFRα-positive cells in the murine lungs. Moreover, female mice showed a higher Timp-1 expression in the lungs on day 3 postinfection. Mechanistically, we observed that estrogen can upregulate TIMP-1 expression in lung fibroblasts, not epithelial cells. In addition, overexpression of estrogen receptor α (ERα) increased the TIMP-1 promoter activity. In summary, TIMP-1 is an estrogen-responsive gene, and its promoter activity is regulated by ERα. Circulating TIMP-1 may serve as a sex-specific marker, reflecting the severity and worst outcomes in female patients with SARS-CoV2- and IAV-related ALI.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
- Department of Clinical Pharmacy, College of PharmacyUniversity of HailHailSaudi Arabia
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Siddhika Gamare
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Valeria Irineo‐Moreno
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Angel Camarena
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Paloma Campero
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | | | - Tatiana S. Rodriguez‐ Reyna
- Department of Immunology and RheumatologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
- Department of Medicine, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
10
|
De Kesel W, Vanden Broecke B, Borremans B, Fourchault L, Willems E, Ceulemans A, Sabuni C, Massawe A, Makundi RH, Leirs H, Peeters M, Verheyen E, Gryseels S, Mariën J, Ariën KK. Antibodies against medically relevant arthropod-borne viruses in the ubiquitous African rodent Mastomys natalensis. PLoS Negl Trop Dis 2024; 18:e0012233. [PMID: 39231158 PMCID: PMC11404846 DOI: 10.1371/journal.pntd.0012233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/16/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Over the past decades, the number of arthropod-borne virus (arbovirus) outbreaks has increased worldwide. Knowledge regarding the sylvatic cycle (i.e., non-human hosts/environment) of arboviruses is limited, particularly in Africa, and the main hosts for virus maintenance are unknown. Previous studies have shown the presence of antibodies against certain arboviruses (i.e., chikungunya-, dengue-, and Zika virus) in African non-human primates and bats. We hypothesize that small mammals, specifically rodents, may function as amplifying hosts in anthropogenic environments. The detection of RNA of most arboviruses is complicated by the viruses' short viremic period within their hosts. An alternative to determine arbovirus hosts is by detecting antibodies, which can persist several months. Therefore, we developed a high-throughput multiplex immunoassay to detect antibodies against 15 medically relevant arboviruses. We used this assay to assess approximately 1,300 blood samples of the multimammate mouse, Mastomys natalensis from Tanzania. In 24% of the samples, we detected antibodies against at least one of the tested arboviruses, with high seroprevalences of antibodies reacting against dengue virus serotype one (7.6%) and two (8.4%), and chikungunya virus (6%). Seroprevalence was higher in females and increased with age, which could be explained by inherent immunity and behavioral differences between sexes, and the increased chance of exposure to an arbovirus with age. We evaluated whether antibodies against multiple arboviruses co-occur more often than randomly and found that this may be true for some members of the Flaviviridae and Togaviridae. In conclusion, the development of an assay against a wide diversity of medically relevant arboviruses enabled the analysis of a large sample collection of one of the most abundant African small mammals. Our findings highlight that Mastomys natalensis is involved in the transmission cycle of multiple arboviruses and provide a solid foundation to better understand the role of this ubiquitous rodent in arbovirus outbreaks.
Collapse
Affiliation(s)
- Wim De Kesel
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Bram Vanden Broecke
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Terrestrial Ecology Unit, Department of Biology, Ghent University, Ghent, Belgium
| | - Benny Borremans
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Wildlife Health Ecology Research Organization, San Diego, California, United States of America
| | - Léa Fourchault
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Elisabeth Willems
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Ann Ceulemans
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Virus Ecology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Christopher Sabuni
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Apia Massawe
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Rhodes H Makundi
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Herwig Leirs
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
| | - Martine Peeters
- TransVIHMI, University of Montpellier, Institut de Recherche pour le Développement (IRD), INSERM, Montpellier, France
| | - Erik Verheyen
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Sophie Gryseels
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Joachim Mariën
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Virus Ecology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Aleyiydi MS, Alshiban NM, Alajmi AM, Alosaimi NF, Alotaibi M, Nassar MS, Alhumaid NK, Almangour TA, Memish ZA, Binjomah AZ, Algarni SM, Al-Jedai A, Almutairi AS, Shibl A, Tawfik EA. Epidemiology of Viral Infectious Diseases Reported in Saudi Arabia. Infect Dis Ther 2024; 13:1893-1905. [PMID: 38967741 PMCID: PMC11266316 DOI: 10.1007/s40121-024-01014-0] [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: 05/16/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024] Open
Abstract
INTRODUCTION Efficient epidemiological monitoring of virus diseases is crucial in evaluating general public health, the prevalence of specific diseases, the pattern of spread, and implementing preventative and control strategies into action. METHODS This study analyzed data obtained from the Field Epidemiology Program (FETP) which is part of the Ministry of Health (MOH) in Saudi Arabia, which contained reported cases of infectious diseases over four years, from January 2018 to December 2021, to investigate and highlight the significant trend and incidence rate for each viral infectious disease. RESULTS Of the reported viral infectious diseases, hepatitis B and C, dengue fever (DF), influenza, chickenpox, and measles were the highest reported viral cases over four years. For the aforementioned diseases, males were often more susceptible to viral infections than females. Except for DF, this viral infection was more common in Saudi citizens. Viral illnesses like hand, foot, and mouth disease were less prevalent, while neurological viral disorders such as acute flaccid paralysis were rarely detected. There was an overall reduction in viral cases recorded during 2020-2021, which may be attributed to the implementation of preventive measures during the Coronavirus Disease 2019 (COVID-19) pandemic or an underreporting of cases during the lockdown of that time. CONCLUSION The prevalence of these common viral infections in the Saudi population suggests that understanding the mechanisms influencing changes in these viruses, methods of transmission, and the burden of these diseases is a priority for health policy. This understanding is necessary to develop effective intervention and preventive strategies.
Collapse
Affiliation(s)
- Munirah S Aleyiydi
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Noura M Alshiban
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Areej M Alajmi
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Nada F Alosaimi
- Wellness and Preventive Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Maryam Alotaibi
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Majed S Nassar
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Nada K Alhumaid
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia
| | - Thamer A Almangour
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Ziad A Memish
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
- Research & Innovation Center, King Saud Medical City, Riyadh, Saudi Arabia
- Hubert Department School of Public Health, Emory University, Atlanta, USA
- Division of Infectious Diseases, Kyung Hee University, Seoul, South Korea
| | - Abdulwahab Z Binjomah
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
- Mycobacteriology Unit, Riyadh Regional Laboratory, Ministry of Health, 12746, Riyadh, Saudi Arabia
| | | | - Ahmed Al-Jedai
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
- Therapeutic Affairs, Ministry of Health, 12631, Riyadh, Saudi Arabia
| | | | - Atef Shibl
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
| | - Essam A Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 11442, Riyadh, Saudi Arabia.
| |
Collapse
|
12
|
Janssen LLG, van Leeuwen-Kerkhoff N, Westers TM, de Gruijl TD, van de Loosdrecht AA. The immunoregulatory role of monocytes and thrombomodulin in myelodysplastic neoplasms. Front Oncol 2024; 14:1414102. [PMID: 39132505 PMCID: PMC11310157 DOI: 10.3389/fonc.2024.1414102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) are clonal disorders of the myeloid lineage leading to peripheral blood cytopenias. Dysregulation of innate immunity is hypothesized to be a potent driver of MDS. A recent study revealed increased thrombomodulin (TM) expression on classical monocytes in MDS, which was associated with prolonged survival. TM is a receptor with immunoregulatory capacities, however, its exact role in MDS development remains to be elucidated. In this review we focus on normal monocyte biology and report on the involvement of monocytes in myeloid disease entities with a special focus on MDS. Furthermore, we delve into the current knowledge on TM and its function in monocytes in health and disease and explore the role of TM-expressing monocytes as driver, supporter or epiphenomenon in the MDS bone marrow environment.
Collapse
Affiliation(s)
- Luca L. G. Janssen
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Nathalie van Leeuwen-Kerkhoff
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Theresia M. Westers
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Institute for Immunity and Infectious Diseases, Amsterdam, Netherlands
| | - Arjan A. van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| |
Collapse
|
13
|
Torrance BL, Cadar AN, Panier HA, Martin DE, Lorenzo EC, Jellison ER, Bartley JM, Haynes L. Cellular senescence is a double-edged sword in regulating aged immune responses to influenza. Aging Cell 2024; 23:e14162. [PMID: 38689516 PMCID: PMC11258475 DOI: 10.1111/acel.14162] [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: 05/19/2023] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024] Open
Abstract
Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective clearance of p16-expressing cells upon administration of ganciclovir (GCV). While p16-expressing cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although clearance of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV-treated mice. Furthermore, GCV-treated mice were unable to mount an optimal memory response and demonstrated increased viral load following heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.
Collapse
Affiliation(s)
- Blake L. Torrance
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Andreia N. Cadar
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Hunter A. Panier
- Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Dominique E. Martin
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Erica C. Lorenzo
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Evan R. Jellison
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Jenna M. Bartley
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Laura Haynes
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| |
Collapse
|
14
|
Donald J, Bilasy SE, Yang C, El-Shamy A. Exploring the Complexities of Long COVID. Viruses 2024; 16:1060. [PMID: 39066223 PMCID: PMC11281588 DOI: 10.3390/v16071060] [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: 06/03/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Since the emergence of the SARS-CoV-2 virus in 2019, nearly 700 million COVID-19 cases and 7 million deaths have been reported globally. Despite most individuals recovering within four weeks, the Center for Disease Control (CDC) estimates that 7.5% to 41% develop post-acute infection syndrome (PAIS), known as 'Long COVID'. This review provides current statistics on Long COVID's prevalence, explores hypotheses concerning epidemiological factors, such as age, gender, comorbidities, initial COVID-19 severity, and vaccine interactions, and delves into potential mechanisms, including immune responses, viral persistence, and gut dysbiosis. Moreover, we conclude that women, advanced age, comorbidities, non-vaccination, and low socioeconomic status all appear to be risk factors. The reasons for these differences are still not fully understood and likely involve a complex relationship between social, genetic, hormonal, and other factors. Furthermore, individuals with Long COVID-19 seem more likely to endure economic hardship due to persistent symptoms. In summary, our findings further illustrate the multifaceted nature of Long COVID and underscore the importance of understanding the epidemiological factors and potential mechanisms needed to develop effective therapeutic strategies and interventions.
Collapse
Affiliation(s)
- Jackson Donald
- College of Graduate Studies, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA; (J.D.); (C.Y.)
| | - Shymaa E. Bilasy
- College of Dental Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA;
| | - Catherine Yang
- College of Graduate Studies, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA; (J.D.); (C.Y.)
| | - Ahmed El-Shamy
- College of Graduate Studies, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA; (J.D.); (C.Y.)
| |
Collapse
|
15
|
Cecchini MP, Pizzini FB, Boschi F, Marcon A, Moro L, Gordon E, Guizard N, Cavedo E, Ricatti MJ, Veronese S, Tamburin S, Tinazzi M, Mansueto G, Sbarbati A. Long-term effects of SARS-CoV-2 infection in patients with and without chemosensory disorders at disease onset: a psychophysical and magnetic resonance imaging exploratory study. Neurol Sci 2024; 45:2409-2418. [PMID: 38441790 PMCID: PMC11082021 DOI: 10.1007/s10072-024-07429-4] [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: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 05/12/2024]
Abstract
A preserved sense of smell and taste allows us to understand many environmental "messages" and results in meaningfully improvements to quality of life. With the COVID-19 pandemic, it became clear how important these senses are for social and nutritional status and catapulted this niche chemosensory research area towards widespread interest. In the current exploratory work, we assessed two groups of post-COVID-19 patients who reported having had (Group 1) or not (Group 2) a smell/taste impairment at the disease onset. The aim was to compare them using validated smell and taste tests as well as with brain magnetic resonance imaging volumetric analysis. Normative data were used for smell scores comparison and a pool of healthy subjects, recruited before the pandemic, served as controls for taste scores. The majority of patients in both groups showed an olfactory impairment, which was more severe in Group 1 (median UPSIT scores: 24.5 Group 1 vs 31.0 Group 2, p = 0.008), particularly among women (p = 0.014). No significant differences emerged comparing taste scores between Group 1 and Group 2, but dysgeusia was only present in Group 1 patients. However, for taste scores, a significant difference was found between Group 1 and controls (p = 0.005). No MRI anatomical abnormalities emerged in any patients while brain volumetric analysis suggested a significant difference among groups for the right caudate nucleus (p = 0.028), although this was not retained following Benjamini-Hochberg correction. This exploratory study could add new information in COVID-19 chemosensory long-lasting impairment and address future investigations on the post-COVID-19 patients' research.
Collapse
Affiliation(s)
- Maria Paola Cecchini
- Department of Neurosciences, Biomedicines and Movement Sciences, Anatomy and Histology Section, Verona University, Strada Le Grazie 8, 37134, Verona, Italy.
| | | | - Federico Boschi
- Department of Engineering for Innovation Medicine, Engineering and Physics Section, Verona University, Verona, Italy
| | - Alessandro Marcon
- Department of Diagnostics and Public Heath, Unit of Epidemiology and Medical Statistics, Verona University, Verona, Italy
| | - Lucia Moro
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore, Negrar, Italy
| | | | | | | | - Maria Jimena Ricatti
- Department of Neurosciences, Biomedicines and Movement Sciences, Anatomy and Histology Section, Verona University, Strada Le Grazie 8, 37134, Verona, Italy
| | - Sheila Veronese
- Department of Neurosciences, Biomedicines and Movement Sciences, Anatomy and Histology Section, Verona University, Strada Le Grazie 8, 37134, Verona, Italy
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicines and Movement Sciences, Neurology Unit, Verona University, Verona, Italy
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicines and Movement Sciences, Neurology Unit, Verona University, Verona, Italy
| | - Giancarlo Mansueto
- Department of Engineering for Innovation Medicine, Radiology Unit, Verona University, Verona, Italy
| | - Andrea Sbarbati
- Department of Neurosciences, Biomedicines and Movement Sciences, Anatomy and Histology Section, Verona University, Strada Le Grazie 8, 37134, Verona, Italy
| |
Collapse
|
16
|
Nambulli S, Escriou N, Rennick LJ, Demers MJ, Tilston‑Lunel NL, McElroy AK, Barbeau DJ, Crossland NA, Hoehl RM, Schrauf S, White AG, Borish HJ, Tomko JA, Frye LJ, Scanga CA, Flynn JL, Martin A, Gerke C, Hartman AL, Duprex WP. A measles-vectored vaccine candidate expressing prefusion-stabilized SARS-CoV-2 spike protein brought to phase I/II clinical trials: protection of African green monkeys from COVID-19 disease. J Virol 2024; 98:e0176223. [PMID: 38563762 PMCID: PMC11092351 DOI: 10.1128/jvi.01762-23] [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: 11/10/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and is responsible for the largest human pandemic in 100 years. Thirty-four vaccines are currently approved for use worldwide, and approximately 67% of the world population has received a complete primary series of one, yet countries are dealing with new waves of infections, variant viruses continue to emerge, and breakthrough infections are frequent secondary to waning immunity. Here, we evaluate a measles virus (MV)-vectored vaccine expressing a stabilized prefusion SARS-CoV-2 spike (S) protein (MV-ATU3-S2PΔF2A; V591) with demonstrated immunogenicity in mouse models (see companion article [J. Brunet, Z. Choucha, M. Gransagne, H. Tabbal, M.-W. Ku et al., J Virol 98:e01693-23, 2024, https://doi.org/10.1128/jvi.01693-23]) in an established African green monkey model of disease. Animals were vaccinated with V591 or the control vaccine (an equivalent MV-vectored vaccine with an irrelevant antigen) intramuscularly using a prime/boost schedule, followed by challenge with an early pandemic isolate of SARS-CoV-2 at 56 days post-vaccination. Pre-challenge, only V591-vaccinated animals developed S-specific antibodies that had virus-neutralizing activity as well as S-specific T cells. Following the challenge, V591-vaccinated animals had lower infectious virus and viral (v) RNA loads in mucosal secretions and stopped shedding virus in these secretions earlier. vRNA loads were lower in these animals in respiratory and gastrointestinal tract tissues at necropsy. This correlated with a lower disease burden in the lungs as quantified by PET/CT at early and late time points post-challenge and by pathological analysis at necropsy.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the largest human pandemic in 100 years. Even though vaccines are currently available, countries are dealing with new waves of infections, variant viruses continue to emerge, breakthrough infections are frequent, and vaccine hesitancy persists. This study uses a safe and effective measles vaccine as a platform for vaccination against SARS-CoV-2. The candidate vaccine was used to vaccinate African green monkeys (AGMs). All vaccinated AGMs developed robust antigen-specific immune responses. After challenge, these AGMs produced less virus in mucosal secretions, for a shorter period, and had a reduced disease burden in the lungs compared to control animals. At necropsy, lower levels of viral RNA were detected in tissue samples from vaccinated animals, and the lungs of these animals lacked the histologic hallmarks of SARS-CoV-2 disease observed exclusively in the control AGMs.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Chlorocebus aethiops
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Measles virus/immunology
- Measles virus/genetics
- COVID-19 Vaccines/immunology
- Humans
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Genetic Vectors
- Vero Cells
- Pandemics/prevention & control
- Female
- Betacoronavirus/immunology
- Betacoronavirus/genetics
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Pneumonia, Viral/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Coronavirus Infections/veterinary
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Disease Models, Animal
Collapse
Affiliation(s)
- Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicolas Escriou
- Département de Santé Globale, Institut Pasteur, Université de Paris Cite, Paris, France
| | - Linda J. Rennick
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natasha L. Tilston‑Lunel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anita K. McElroy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dominique J. Barbeau
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sabrina Schrauf
- Themis Bioscience GmbH, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jaime A. Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lonnie J. Frye
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Charles A. Scanga
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - JoAnne L. Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Annette Martin
- CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université de Paris, Paris, France
| | - Christiane Gerke
- Vaccine Programs, Institut Pasteur, Université de Paris Cite, Innovation Office, Paris, France
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - W. Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Santana-Sánchez P, Vaquero-García R, Legorreta-Haquet MV, Chávez-Sánchez L, Chávez-Rueda AK. Hormones and B-cell development in health and autoimmunity. Front Immunol 2024; 15:1385501. [PMID: 38680484 PMCID: PMC11045971 DOI: 10.3389/fimmu.2024.1385501] [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: 02/12/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
The development of B cells into antibody-secreting plasma cells is central to the adaptive immune system as they induce protective and specific antibody responses against invading pathogens. Various studies have shown that, during this process, hormones can play important roles in the lymphopoiesis, activation, proliferation, and differentiation of B cells, and depending on the signal given by the receptor of each hormone, they can have a positive or negative effect. In autoimmune diseases, hormonal deregulation has been reported to be related to the survival, activation and/or differentiation of autoreactive clones of B cells, thus promoting the development of autoimmunity. Clinical manifestations of autoimmune diseases have been associated with estrogens, prolactin (PRL), and growth hormone (GH) levels. However, androgens, such as testosterone and progesterone (P4), could have a protective effect. The objective of this review is to highlight the links between different hormones and the immune response mediated by B cells in the etiopathogenesis of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). The data collected provide insights into the role of hormones in the cellular, molecular and/or epigenetic mechanisms that modulate the B-cell response in health and disease.
Collapse
Affiliation(s)
| | | | | | | | - Adriana Karina Chávez-Rueda
- Unidad de Investigación Médica en Inmunología, Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico
| |
Collapse
|
18
|
Meyer-Schwickerath C, Weber C, Hornuss D, Rieg S, Hitzenbichler F, Hagel S, Ankert J, Hennigs A, Glossmann J, Jung N. Complexity of patients with or without infectious disease consultation in tertiary-care hospitals in Germany. Infection 2024; 52:577-582. [PMID: 38277092 PMCID: PMC10955003 DOI: 10.1007/s15010-023-02166-w] [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: 11/14/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024]
Abstract
PURPOSE Patients seen by infectious disease (ID) specialists are more complex compared to patients treated by other subspecialities according to Tonelli et al. (2018). However, larger studies on the complexity of patients related to the involvement of ID consultation services are missing. METHODS Data of patients being treated in 2015 and 2019 in four different German university hospitals was retrospectively collected. Data were collected from the hospitals' software system and included whether the patients received an ID consultation as well as patient clinical complexity level (PCCL), case mix index (CMI) and length of stay (LOS) as a measurement for the patients' complexity. Furthermore, a comparison of patients with distinct infectious diseases treated with or without an ID consultation was initiated. RESULTS In total, 215.915 patients were included in the study, 3% (n = 6311) of those were seen by an ID consultant. Patients receiving ID consultations had a significantly (p < 0.05) higher PCCL (median 4 vs. 0), CMI (median 3,8 vs. 1,1) and deviation of the expected mean LOS (median 7 days vs. 0 days) than patients in the control group. No differences among hospitals or between years were observed. Comparing patients with distinct infectious diseases treated with or without an ID consultation, the differences were confirmed throughout the groups. CONCLUSION Patients receiving ID consultations are highly complex, frequently need further treatment after discharge and have a high economic impact. Thus, ID specialists should be clinically trained in a broad spectrum of diseases and treating these complex patients should be sufficiently remunerated.
Collapse
Affiliation(s)
- C Meyer-Schwickerath
- Department I of Internal Medicine, Division of Infectious Diseases, University of Cologne, Cologne, Germany
| | - C Weber
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - D Hornuss
- Faculty of Medicine, Department of Medicine II, Division of Infectious Diseases, Medical Center - University of Freiburg, Freiburg, Germany
| | - S Rieg
- Faculty of Medicine, Department of Medicine II, Division of Infectious Diseases, Medical Center - University of Freiburg, Freiburg, Germany
| | - F Hitzenbichler
- Department of Infection Prevention and Infectious Diseases, University Hospital of Regensburg, Regensburg, Germany
| | - S Hagel
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - J Ankert
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - A Hennigs
- I. Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - J Glossmann
- Center of Integrated Oncology Aachen Bonn Cologne Düsseldorf, University of Cologne, Cologne, Germany
| | - N Jung
- Department I of Internal Medicine, Division of Infectious Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
19
|
Naidoo KK, Highton AJ, Baiyegunhi OO, Bhengu SP, Dong KL, Bunders MJ, Altfeld M, Ndung’u T. Early Initiation of Antiretroviral Therapy Preserves the Metabolic Function of CD4+ T Cells in Subtype C Human Immunodeficiency Virus 1 Infection. J Infect Dis 2024; 229:753-762. [PMID: 37804102 PMCID: PMC10938216 DOI: 10.1093/infdis/jiad432] [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: 05/11/2023] [Revised: 09/19/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Immune dysfunction often persists in people living with human immunodeficiency virus (HIV) who are on antiretroviral therapy (ART), clinically manifesting as HIV-1-associated comorbid conditions. Early ART initiation may reduce incidence of HIV-1-associated immune dysfunction and comorbid conditions. Immunometabolism is a critical determinant of functional immunity. We investigated the effect of HIV-1 infection and timing of ART initiation on CD4+ T cell metabolism and function. METHODS Longitudinal blood samples from people living with HIV who initiated ART during hyperacute HIV-1 infection (HHI; before peak viremia) or chronic HIV-1 infection (CHI) were assessed for the metabolic and immune functions of CD4+ T cells. Metabolite uptake and mitochondrial mass were measured using fluorescent analogues and MitoTracker Green accumulation, respectively, and were correlated with CD4+ T cell effector functions. RESULTS Initiation of ART during HHI prevented dysregulation of glucose uptake by CD4+ T cells, but glucose uptake was reduced before and after ART initiation in CHI. Glucose uptake positively correlated with interleukin-2 and tumor necrosis factor-α production by CD4+ T cells. CHI was associated with elevated mitochondrial mass in effector memory CD4+ T cells that persisted after ART and correlated with PD-1 expression. CONCLUSIONS ART initiation in HHI largely prevented metabolic impairment of CD4+ T cells. ART initiation in CHI was associated with persistently dysregulated immunometabolism of CD4+ T cells, which was associated with impaired cellular functions and exhaustion.
Collapse
Affiliation(s)
- Kewreshini K Naidoo
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Andrew J Highton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | | | - Sindiswa P Bhengu
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Krista L Dong
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute, Durban, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
20
|
Kelkar NS, Goldberg BS, Dufloo J, Bruel T, Schwartz O, Hessell AJ, Ackerman ME. Sex- and species-associated differences in complement-mediated immunity in humans and rhesus macaques. mBio 2024; 15:e0028224. [PMID: 38385704 PMCID: PMC10936177 DOI: 10.1128/mbio.00282-24] [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: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024] Open
Abstract
The complement system can be viewed as a "moderator" of innate immunity, "instructor" of humoral immunity, and "regulator" of adaptive immunity. While sex is known to affect humoral and cellular immune systems, its impact on complement in humans and rhesus macaques, a commonly used non-human primate model system, has not been well studied. To address this knowledge gap, we analyzed serum samples from 90 humans and 72 rhesus macaques for the abundance and activity of the complement system components. While sequences of cascade proteins were highly conserved, dramatically different levels were observed between species. Whereas the low levels detected in rhesus samples raised questions about the suitability of the test for use with macaque samples, differences in levels of complement proteins were observed in male and female humans. Levels of total and antibody-dependent deposition of C1q and C3b on a glycosylated antigen differed between humans and rhesus, suggesting differential recognition of glycans and balance between classical and alternative activation pathways. Functional differences in complement-mediated lysis of antibody-sensitized cells were observed in multiple assays and showed that human females frequently exhibited higher lytic activity than human males or rhesus macaques, which typically did not exhibit such sex-associated differences. Other differences between species and sexes were observed in more narrow contexts-for only certain antibodies, antigens, or assays. Collectively, these results expand knowledge of sex-associated differences in the complement system in humans, identifying differences absent from rhesus macaques.IMPORTANCEThe complement system is a critical part of host defense to many bacterial, fungal, and viral infections. In parallel, rich epidemiological, clinical, and biomedical research evidence demonstrates that sex is an important biological variable in immunity, and many sex-specific differences in immune system are intimately tied with disease outcomes. This study focuses on the intersection of these two factors to define the impact of sex on complement pathway components and activities. This work expands our knowledge of sex-associated differences in the complement system in humans and also identifies the differences that appear to be absent in rhesus macaques, a popular non-human primate model. Whereas differences between species suggest potential limitations in the ability of macaque model to recapitulate human biology, knowledge of sex-based differences in humans has the potential to inform clinical research and practice.
Collapse
Affiliation(s)
- Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
21
|
Björkhem-Bergman L, Schultz T, Strang P. Age and co-morbidities as independent risk factors of infections leading to hospital admission in the last year of life among the elderly: A retrospective registry-based study. Ups J Med Sci 2024; 129:10504. [PMID: 38571881 PMCID: PMC10989213 DOI: 10.48101/ujms.v129.10504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024] Open
Abstract
Background The immune system declines with age, but the impact of chronological age may be affected by sex, co-morbidities, and sociodemographic factors. Objective The article aims to study infections associated with hospital admission in the elderly in their last year of life and the impact of age, sex, co-morbidities, and sociodemographic factors. Method A retrospective study based on registry data covering all care visits in Stockholm Region, Sweden, for 7 years was conducted. All deceased subjects with at least one hospital admission with infection as the main diagnosis in the last year of life were compared with subjects with no such admission. Subjects were categorized into three different age-groups 65-79, 80-89, and 90 years and above. Co-morbidity was measured by the Charlson Comorbidity Index (CCI) and sociodemographic factors were assessed using the 'Mosaic-system'. Subjects living in nursing homes were analyzed separately. Uni- and multivariable logistic regressions were conducted. Results Of the 55,238 subjects in the study population, 14,192 (26%) had at least one hospital admission due to infection in the last year of life. The risk of having a severe infection increased with age, adjusted odds ratio (OR): 1.30 (1.25-1.36), and 1.60 (1.52-1.69) for the age-groups 80-89 and ≥ 90 compared to the age-group 65-79. The most important factor for infection was a high co-morbidity score; adjusted OR: 1.75 (1.68-1.82). Male sex and living in a less affluent area were weaker risk factors for infections. Conclusion Chronological age and co-morbidities are independent risk factors of infections associated with hospital admission in the last year in life while male sex and sociodemographic factors have less impact.
Collapse
Affiliation(s)
- Linda Björkhem-Bergman
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
- Palliative Medicine, Stockholms Sjukhem, Stockholm, Sweden
| | | | - Peter Strang
- Palliative Medicine, Stockholms Sjukhem, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Guillien A, Niespodziana K, Mauclin M, Boudier A, Varraso R, Leynaert B, Dumas O, Le Moual N, Schlederer T, Bajic M, Borochova K, Errhalt P, Vernet R, Nadif R, Bousquet J, Bouzigon E, Valenta R, Siroux V. Determinants of immunoglobulin G responses to respiratory syncytial virus and rhinovirus in children and adults. Front Immunol 2024; 15:1355214. [PMID: 38500884 PMCID: PMC10945029 DOI: 10.3389/fimmu.2024.1355214] [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: 12/13/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
Introduction Exposure to respiratory viruses is a significant cause of morbidity and affects virus-specific antibody levels. Little is known about determinants associated with immune response to these viruses. We aimed to investigate the determinants of respiratory syncytial virus (RSV)- and rhinovirus (RV)- specific IgG responses in both children and adults. Methods The study is based on the EGEA cohort, composed of 530 samples of children in EGEA1 (1991-95) and 1241 samples of adults in EGEA2 (2003-07). Cumulative RV-specific IgG levels (species A, B and C) and IgG levels to RSV-G protein were measured by using micro-array technoloy. Multiple linear mixed models (random effect to account for familial dependence) were performed to assess associations between age, sex, body mass index (BMI), tobacco smoke exposure and season of blood sampling with RSV-and RV-specific IgG levels. Results In children (11.1 ± 2.8 years old, 57% boys), higher RV-specific IgG levels were associated with older age (only for RV-B), female sex and lower BMI, while only older age was associated with higher RSV-specific IgG levels. In adults (43.5 ± 16.7 years old, 48% men), younger age, female sex, lower BMI, active smoking and all seasons except summer were associated with higher RV-specific IgG levels. Older age, active smoking and all seasons except summer were associated with higher RSV-specific IgG levels. Conclusion Personal and seasonal determinants of RSV- and RV-specific IgG levels seem to vary according to the respiratory virus type and between children and adults, suggesting different patterns of responses along the life course.
Collapse
Affiliation(s)
- Alicia Guillien
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team in Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Katarzyna Niespodziana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Marion Mauclin
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team in Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Anne Boudier
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team in Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
- CHU Grenoble Alpes, Grenoble, France
| | - Raphäelle Varraso
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Bénédicte Leynaert
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Orianne Dumas
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Nicole Le Moual
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Thomas Schlederer
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Maja Bajic
- Karl Landsteiner University, Krems, Austria
| | - Kristina Borochova
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Errhalt
- Karl Landsteiner University, Krems, Austria
- Department of Pneumology, University Hospital, Krems, Austria
| | - Raphaël Vernet
- Université Paris Cité, Inserm, UMRS 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris, France
| | - Rachel Nadif
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Jean Bousquet
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Equipe d’Epidémiologie Respiratoire Intégrative, Centre de recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Emmanuelle Bouzigon
- Université Paris Cité, Inserm, UMRS 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris, France
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University, Krems, Austria
| | - Valérie Siroux
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team in Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
23
|
Eposse Ekoube C, Heles Nsang E, Épée P, Mandeng Ma Linwa E, Djike Puepi Y, Mbono Betoko R, Noukeu Njinkui D, Enyama D, Mbonjo Bitsie D, Disso Massako J, Abba S, Ngo Linwa EE, Ida Penda C. Predictors of prolonged length of hospital stay and in-hospital mortality in patients aged 1-24 months with acute bronchiolitis in Douala, Cameroon. BMC Pediatr 2024; 24:150. [PMID: 38424505 PMCID: PMC10903055 DOI: 10.1186/s12887-024-04653-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION In Cameroon, acute bronchiolitis has been reported as the third commonest lower respiratory infection and is usually associated with low mortality. Nonetheless, respiratory distress associated with non-adherence to management guidelines can prolong hospital stay. This study aimed to explore predictors of prolonged hospital stay (≥ 5 days) and mortality in patients aged < 2years hospitalised for acute bronchiolitis. METHODOLOGY We conducted a retrospective cohort study at three paediatric units in the city of Douala, Cameroon. Factors associated with prolonged hospital stay and mortality were determined using multivariable linear regression model. Threshold for significance was set at p ≤ 0.05. RESULTS A total of 215 patients with bronchiolitis were included with mean age of 6.94 ± 5.71 months and M/F sex ratio of 1.39/1. Prolonged hospital stay was reported in 46.98% and mortality in 10.70% of patients hospitalised for bronchiolitis. Factors independently associated with prolonged hospital duration were oxygen administration [b = 0.36, OR = 2.35 (95% CI:1.16-4.74), p = 0.017], abnormal respiratory rate [b = 0.38, OR = 2.13 (1.00-4.55), p = 0.050] and patients presenting with cough [b = 0.33, OR = 2.35 (95% CI: 1.22-4.51), p = 0.011], and diarrhoea [b = 0.71, OR = 6.44 (95% CI: 1.6-25.86), p = 0.009] on admission. On the other hand, factors independently associated with mortality were age of the patient [b= -0.07, OR = 0.84 (95% CI: 0.74-0.97), p = 0.014] and oxygen administration [b = 1.08, OR = 9.64 (95% CI:1.16-79.85), p = 0.036] CONCLUSION: Acute bronchiolitis represented 1.24% of admissions and was common in the rainy season, in males and 3-11-month-old patients. Management guidelines were poorly respected. Prolonged length of stay was reported in half of the patients hospitalized and mortality was high, especially in younger patients and in patients receiving oxygen.
Collapse
Affiliation(s)
- Charlotte Eposse Ekoube
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon.
- Paediatric unit, Douala Laquintinie Hospital, Douala, Cameroon.
| | - Emmanuel Heles Nsang
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | - Patricia Épée
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | | | | | - Ritha Mbono Betoko
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | - Diomède Noukeu Njinkui
- Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Dominique Enyama
- Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Dora Mbonjo Bitsie
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | | | - Soumaiyatou Abba
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| | | | - Calixte Ida Penda
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
| |
Collapse
|
24
|
Sampson OL, Jay C, Adland E, Csala A, Lim N, Ebbrecht SM, Gilligan LC, Taylor AE, George SS, Longet S, Jones LC, Barnes E, Frater J, Klenerman P, Dunachie S, Carrol M, Hawley J, Arlt W, Groll A, Goulder P. Gonadal androgens are associated with decreased type I interferon production by plasmacytoid dendritic cells and increased IgG titres to BNT162b2 following co-vaccination with live attenuated influenza vaccine in adolescents. Front Immunol 2024; 15:1329805. [PMID: 38481993 PMCID: PMC10933029 DOI: 10.3389/fimmu.2024.1329805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/13/2024] [Indexed: 04/09/2024] Open
Abstract
mRNA vaccine technologies introduced following the SARS-CoV-2 pandemic have highlighted the need to better understand the interaction of adjuvants and the early innate immune response. Type I interferon (IFN-I) is an integral part of this early innate response that primes several components of the adaptive immune response. Women are widely reported to respond better than men to tri- and quadrivalent influenza vaccines. Plasmacytoid dendritic cells (pDCs) are the primary cell type responsible for IFN-I production, and female pDCs produce more IFN-I than male pDCs since the upstream pattern recognition receptor Toll-like receptor 7 (TLR7) is encoded by X chromosome and is biallelically expressed by up to 30% of female immune cells. Additionally, the TLR7 promoter contains several putative androgen response elements, and androgens have been reported to suppress pDC IFN-I in vitro. Unexpectedly, therefore, we recently observed that male adolescents mount stronger antibody responses to the Pfizer BNT162b2 mRNA vaccine than female adolescents after controlling for natural SARS-CoV-2 infection. We here examined pDC behaviour in this same cohort to determine the impact of IFN-I on anti-spike and anti-receptor-binding domain IgG titres to BNT162b2. Through flow cytometry and least absolute shrinkage and selection operator (LASSO) modelling, we determined that serum-free testosterone was associated with reduced pDC IFN-I, but contrary to the well-described immunosuppressive role for androgens, the most bioactive androgen dihydrotestosterone was associated with increased IgG titres to BNT162b2. Also unexpectedly, we observed that co-vaccination with live attenuated influenza vaccine boosted the magnitude of IgG responses to BNT162b2. Together, these data support a model where systemic IFN-I increases vaccine-mediated immune responses, yet for vaccines with intracellular stages, modulation of the local IFN-I response may alter antigen longevity and consequently improve vaccine-driven immunity.
Collapse
Affiliation(s)
- Oliver L. Sampson
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Cecilia Jay
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Anna Csala
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Nicholas Lim
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Stella M. Ebbrecht
- Department of Statistics, Technical University of Dortmund, Dortmund, Germany
| | - Lorna C. Gilligan
- Steroid Metabolome Analysis Core, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Angela E. Taylor
- Steroid Metabolome Analysis Core, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Sherley Sherafin George
- Biochemistry Department, Clinical Science Building, Wythenshawe Hospital, Manchester, United Kingdom
| | - Stephanie Longet
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Lucy C. Jones
- Department of Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Ellie Barnes
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Susie Dunachie
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Miles Carrol
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - James Hawley
- Biochemistry Department, Clinical Science Building, Wythenshawe Hospital, Manchester, United Kingdom
| | - Wiebke Arlt
- Steroid Metabolome Analysis Core, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Medical Research Council London Institute of Medical Sciences (MRC LMS), Imperial College London, London, United Kingdom
| | - Andreas Groll
- Department of Statistics, Technical University of Dortmund, Dortmund, Germany
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Di Pietro C, Haberman AM, Lindenbach BD, Smith PC, Bruscia EM, Allore HG, Vander Wyk B, Tyagi A, Zeiss CJ. Prior Influenza Infection Mitigates SARS-CoV-2 Disease in Syrian Hamsters. Viruses 2024; 16:246. [PMID: 38400021 PMCID: PMC10891789 DOI: 10.3390/v16020246] [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: 01/11/2024] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Seasonal infection rates of individual viruses are influenced by synergistic or inhibitory interactions between coincident viruses. Endemic patterns of SARS-CoV-2 and influenza infection overlap seasonally in the Northern hemisphere and may be similarly influenced. We explored the immunopathologic basis of SARS-CoV-2 and influenza A (H1N1pdm09) interactions in Syrian hamsters. H1N1 given 48 h prior to SARS-CoV-2 profoundly mitigated weight loss and lung pathology compared to SARS-CoV-2 infection alone. This was accompanied by the normalization of granulocyte dynamics and accelerated antigen-presenting populations in bronchoalveolar lavage and blood. Using nasal transcriptomics, we identified a rapid upregulation of innate and antiviral pathways induced by H1N1 by the time of SARS-CoV-2 inoculation in 48 h dual-infected animals. The animals that were infected with both viruses also showed a notable and temporary downregulation of mitochondrial and viral replication pathways. Quantitative RT-PCR confirmed a decrease in the SARS-CoV-2 viral load and lower cytokine levels in the lungs of animals infected with both viruses throughout the course of the disease. Our data confirm that H1N1 infection induces rapid and transient gene expression that is associated with the mitigation of SARS-CoV-2 pulmonary disease. These protective responses are likely to begin in the upper respiratory tract shortly after infection. On a population level, interaction between these two viruses may influence their relative seasonal infection rates.
Collapse
Affiliation(s)
- Caterina Di Pietro
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06519, USA; (C.D.P.); (E.M.B.)
| | - Ann M. Haberman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Brett D. Lindenbach
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06519, USA;
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Peter C. Smith
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Emanuela M. Bruscia
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06519, USA; (C.D.P.); (E.M.B.)
| | - Heather G. Allore
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (H.G.A.); (B.V.W.)
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06519, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (H.G.A.); (B.V.W.)
| | - Antariksh Tyagi
- Department of Genetics, Yale Center for Genome Analysis, New Haven, CT 06519, USA;
| | - Caroline J. Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| |
Collapse
|
26
|
Harbecke R, Oxman MN, Selke S, Ashbaugh ME, Lan KF, Koelle DM, Wald A. Prior Herpes Simplex Virus Infection and the Risk of Herpes Zoster. J Infect Dis 2024; 229:64-72. [PMID: 37410908 PMCID: PMC10786259 DOI: 10.1093/infdis/jiad259] [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: 01/04/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND The incidence of herpes zoster (HZ) has increased in the United States concurrent with decrease in herpes simplex virus (HSV) prevalence. We hypothesized that lack of HSV-elicited cross-reactive immunity to varicella-zoster virus (VZV) results in an increased risk of HZ. Using specimens from the placebo arm of the Shingles Prevention Study, we investigated whether persons who develop HZ are less likely to have prior HSV infection than persons who do not develop HZ, and whether HZ is less severe in persons with HSV than in HSV seronegative persons. METHODS We conducted a nested case-control (1:2) study comparing the seroprevalence of HSV-1 and HSV-2 in cases (persons with polymerase chain reaction-confirmed HZ) to age-, sex-, and health-matched controls (persons without HZ). RESULTS Sera from 639 study participants (213 cases and 426 controls) yielded definitive HSV antibody results and were analyzed. Overall, HSV seropositivity rate was 75%. HSV seronegativity was significantly higher in HZ cases than controls (30.5% vs 22.3%; P = .024), with a 55% higher risk of HZ in HSV seronegative than HSV seropositive participants. HSV seropositivity was associated with more severe HZ (P = .021). CONCLUSIONS Our study demonstrated that prior infection with HSV partly protects against HZ.
Collapse
Affiliation(s)
- Ruth Harbecke
- Department of Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Medicine, University of California San Diego, San Diego, California, USA
| | - Michael N Oxman
- Department of Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Medicine, University of California San Diego, San Diego, California, USA
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Stacy Selke
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mark E Ashbaugh
- Department of Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Kristine F Lan
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - David M Koelle
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Benaroya Research Institute, Seattle, Washington, USA
| | - Anna Wald
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
27
|
Rudolph TE, Roths M, Freestone AD, White-Springer SH, Rhoads RP, Baumgard LH, Selsby JT. Heat stress alters hematological parameters in barrows and gilts. J Anim Sci 2024; 102:skae123. [PMID: 38706303 PMCID: PMC11141298 DOI: 10.1093/jas/skae123] [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: 10/03/2023] [Accepted: 05/02/2024] [Indexed: 05/07/2024] Open
Abstract
The purpose of this investigation was to establish the role biological sex plays in circulating factors following heat stress (HS). Barrows and gilts (36.8 ± 3.7 kg body weight) were kept in either thermoneutral (TN; 20.8 ± 1.6 °C; 62.0% ± 4.7% relative humidity; n = 8/sex) conditions or exposed to HS (39.4 ± 0.6 °C; 33.7% ± 6.3% relative humidity) for either 1 (HS1; n = 8/sex) or 7 (HS7; n = 8/sex) d. Circulating glucose decreased as a main effect of the environment (P = 0.03). Circulating non-esterified fatty acid (NEFA) had an environment × sex interaction (P < 0.01) as HS1 barrows had increased NEFA compared to HS1 gilts (P = 0.01) and NEFA from HS7 gilts increased compared to HS1 gilts (P = 0.02) and HS7 barrows (P = 0.04). Cortisol, insulin, glucagon, T3, and T4 were reduced as a main effect of environment (P ≤ 0.01). Creatinine was increased in HS1 and HS7 animals compared to TN (P ≤ 0.01), indicative of decreased glomerular filtration rate. White blood cell populations exhibited differential patterns based on sex and time. Neutrophils and lymphocytes had an environment × sex interaction (P ≤ 0.05) as circulating neutrophils were increased in HS1 barrows compared to TN and HS7 barrows, and HS1 gilts (P ≤ 0.01) and HS7 barrows had less neutrophils compared to TN barrows (P = 0.01), whereas they remained similar in gilts. In contrast, barrow lymphocyte numbers were similar between groups, but in HS7 gilts they were decreased compared to TN and HS1 gilts (P ≤ 0.04). In total, these data demonstrate that HS alters a host of circulating factors and that biological sex mediates, at least in part, the physiological response to HS.
Collapse
Affiliation(s)
- Tori E Rudolph
- Department of Animal Science, Iowa State University, Ames, IA, 50010, USA
| | - Melissa Roths
- Department of Animal Science, Iowa State University, Ames, IA, 50010, USA
| | - Alyssa D Freestone
- Department of Animal Science, Iowa State University, Ames, IA, 50010, USA
| | - Sarah H White-Springer
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX, 77843, USA
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, 77843, USA
| | - Robert P Rhoads
- School of Animal Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA, 50010, USA
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, IA, 50010, USA
| |
Collapse
|
28
|
Yang L, Lu Z, Bian J, Li F, Zou H. Association between chronic viral infection-related hospitalization and risk of cardiovascular disease: A population-based cohort study. J Med Virol 2024; 96:e29350. [PMID: 38180233 DOI: 10.1002/jmv.29350] [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: 09/17/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024]
Abstract
Chronic viral infection induces immunosenescence and systemic low-grade inflammation, leading to worsened long-term outcomes. We sought to explore the short- and long-term effects of chronic viral infection on cardiovascular disease (CVD). Based on UK Biobank data, exposed group was identified as individuals who had chronic virus infection-related hospitalization (IRH). Unexposed group was randomly selected, matched by 5-year age interval, sex, and Charlson comorbidity index at a ratio up to 1:10. Restricted cubic splines were used to model time-varying effects of IRH in nonproportional Cox models. A cut-off value of 5 years was recorded and used in piecewise Cox proportional hazards models as we estimated short- and long-term effects of IRH on CVD. A total of 2826 exposed participants and 28 212 matched unexposed participants were included. Chronic viral IRH was associated with increased risk of CVD (0-5 years: hazard ratio, 1.57 [95% confidence interval: 1.32, 1.87] and 5+ years: 1.31 [1.06, 1.61]). Elevated risk of stroke was only observed within the initial 5-year follow-up (0-5 years: 1.91 [1.30, 2.81]). The short- and long-term associations were observed in herpes or hepatitis virus IRH with risk of CVD (all p < 0.05). Subgroup analysis revealed long-term association between chronic viral IRH and CVD among female (5+ years: 1.68 [1.27, 2.22]) but not among male. The association between chronic viral infection and elevated CVD risk appeared to be stronger among individuals who did not take cholesterol-lowering medication, antithrombotic medication, or certain antihypertensive medications (all p for interaction < 0.05). The risk of CVD event remained persistently higher within and over 5 years following chronic viral IRH, especially in individuals infected with herpes and hepatitis virus.
Collapse
Affiliation(s)
- Luoyao Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Zhen Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Junye Bian
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Feng Li
- Department of Cardiac and Vascular Surgery, The 1st Affiliated Hospital of AnHui Medical University, Hefei, China
| | - Huachun Zou
- School of Public Health, Fudan University, Shanghai, China
- School of Public Health, Southwest Medical University, Luzhou, China
- Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
29
|
Abdul-Nabi ZN, Mohamed-Jawad NK, Fareed NY, Neamah NF, Shari FH. Adverse Effects Post COVID-19 Vaccination and its Association with Age, Gender and Comorbid Disease in Basrah City Southern of Iraq. Curr Drug Saf 2024; 19:248-254. [PMID: 37231751 DOI: 10.2174/1574886318666230525142152] [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: 01/22/2023] [Revised: 03/10/2023] [Accepted: 04/06/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Vaccination against COVID-19 virus is the most valuable tool available for protection during the pandemic of coronavirus. The clinical manifestation post-vaccination is a barrier to vaccination for many people in Iraq and worldwide. OBJECTIVES The objective of this study is identifying various clinical manifestations occurring after receiving vaccines among individuals in Basrah Governorate. Moreover, we examine its association with respondents' demographics and the type of vaccine they received. METHODS A cross-section study was conducted in Basrah, southern Iraq. Research data were collected through an online questionnaire. The data were analyzed using both descriptive and analytic statistical tools using the SPSS program. RESULTS Most of the participants (86.68%) received the vaccine. The side effects were reported in 71.61% of vaccinated individuals. Fever and muscle pain were the two most experienced clinical manifestations, while lymph node enlargement and disturbances in taste and/or smell sensations were reported infrequently. Adverse effects were mostly reported with the Pfizer BioNTech vaccine receiver. Females and those in the younger age group also reported a significantly higher incidence of side effects. CONCLUSION Most adverse effects related to the COVID-19 vaccine were minor and could be tolerated without the need for hospital admission.
Collapse
Affiliation(s)
| | | | - Noor Yousif Fareed
- Department of Pharmaceutics, College of Pharmacy, University of Basrah, Basrah, Iraq
| | | | - Falah Hassan Shari
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Basra, Iraq
| |
Collapse
|
30
|
Lee HL, Squire E, Fotio Y, Mabou Tagne A, Lee J, Yoon JJ, Hong Y, Kim LH, Jung KM, Piomelli D. Frequent low-impact exposure to THC during adolescence causes persistent sexually dimorphic alterations in the response to viral infection in mice. Pharmacol Res 2024; 199:107049. [PMID: 38159785 DOI: 10.1016/j.phrs.2023.107049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Adolescent exposure to Δ9-tetrahydrocannabinol (THC) has enduring effects on energy metabolism and immune function. Prior work showed that daily administration of a low-impact dose of THC (5 mg/kg, intraperitoneal) during adolescence alters transcription in adult microglia and disrupts their response to bacterial endotoxin or social stress. To explore the lasting impact of adolescent THC exposure on the brain's reaction to viral infection, we administered THC (5 mg/kg, intraperitoneal) in male and female mice once daily on postnatal day (PND) 30-43. When the mice reached adulthood (PND 70), we challenged them with the viral mimic, polyinosinic acid:polycytidylic acid [Poly(I:C)], and assessed sickness behavior (motor activity, body temperature) and whole brain gene transcription. Poly(I:C) caused an elevation in body temperature which was lessened by prior THC exposure in female but not male mice. Adolescent THC exposure did not affect the locomotor response to Poly(I:C) in either sex. Transcriptomic analyses showed that Poly(I:C) produced a substantial upregulation of immune-related genes in the brain, which was decreased by THC in females. Additionally, the viral mimic caused a male-selective downregulation in transcription of genes involved in neurodevelopment and synaptic transmission, which was abrogated by adolescent THC treatment. The results indicate that Poly(I:C) produces complex transcriptional alterations in the mouse brain, which are sexually dimorphic and differentially affected by early-life THC exposure. In particular, adolescent THC dampens the brain's antiviral response to Poly(I:C) in female mice and prevents the transcriptional downregulation of neuron-related genes caused by the viral mimic in male mice.
Collapse
Affiliation(s)
- Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Jungyeon Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - John Jeongwoo Yoon
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Yedam Hong
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Laura Hyunseo Kim
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, USA; Department of Biological Chemistry, University of California, Irvine, USA; Department of Pharmaceutical Sciences, University of California, Irvine, USA.
| |
Collapse
|
31
|
Anticoli S, Dorrucci M, Iessi E, Chiarotti F, Di Prinzio RR, Vinci MR, Zaffina S, Puro V, Colavita F, Mizzoni K, Meschi S, Vonesch N, Albano C, Ortona E, Ruggieri A, Tomao P. Association between sex hormones and anti-S/RBD antibody responses to COVID-19 vaccines in healthcare workers. Hum Vaccin Immunother 2023; 19:2273697. [PMID: 37961893 PMCID: PMC10760357 DOI: 10.1080/21645515.2023.2273697] [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: 06/05/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Healthcare workers (HCWs) are the target population for vaccination against coronavirus disease (COVID-19) as they are at a high risk of exposure and transmission of pathogens to patients. Neutralizing antibodies developed after COVID-19 vaccination decline within few months of vaccination. Several factors, including age and sex, can affect the intensity, efficacy, and duration of immune response to vaccines. However, sex-specific analyses of humoral responses to COVID-19 vaccines are lacking. This study aimed to evaluate sex-based differences in anti-S/RBD (Receptor Binding Domain) responses at three different time points after the second dose of mRNA COVID-19 vaccine in HCWs in relation to age, and to investigate the role of sex hormones as potential markers of response. Anti-S/RBD levels after two doses of the mRNA vaccine were collected from 521 HCWs naïve to COVID-19, working at two Italian Clinical Centers. Multiple regression analysis was applied to evaluate the association between anti-S levels and sex, age, and plasma levels of sex hormones. Significantly higher anti-S/RBD response to the COVID-19 vaccination was found in female HCWs, and a significant and more abrupt decline in response with time was observed in women than that in men. A novel, positive association of testosterone plasma levels and higher anti-S levels in male HCWs was found, suggesting its potential role as sex specific marker in males. In conclusion, understanding the sex-based differences in humoral immune responses to vaccines may potentially improve vaccination strategies and optimize surveillance programs for HCWs.
Collapse
Affiliation(s)
- Simona Anticoli
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Maria Dorrucci
- Department of Infectious Diseases, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Elisabetta Iessi
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Flavia Chiarotti
- Reference Center for the Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, [Italian National Institute of Health], Rome, Italy
| | | | - Maria Rosaria Vinci
- Occupational Medicine Unit, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Salvatore Zaffina
- Occupational Medicine Unit, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Vincenzo Puro
- UOC Emerging Infections and CRAIDS, National Institute for Infectious Diseases L. Spallanzani IRCSS, Rome, Italy
| | - Francesca Colavita
- UOC Lab of Virology, National Institute for Infectious Diseases L. Spallanzani IRCSS, Rome, Italy
| | - Klizia Mizzoni
- UOC Lab of Virology, National Institute for Infectious Diseases L. Spallanzani IRCSS, Rome, Italy
| | - Silvia Meschi
- UOC Lab of Virology, National Institute for Infectious Diseases L. Spallanzani IRCSS, Rome, Italy
| | - Nicoletta Vonesch
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers’ Compensation Authority (INAIL), Monte Porzio Catone, Rome, Italy
| | - Christian Albano
- B cell Lab, Immunology Research Area, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Elena Ortona
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Anna Ruggieri
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Paola Tomao
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers’ Compensation Authority (INAIL), Monte Porzio Catone, Rome, Italy
| |
Collapse
|
32
|
Calvet GA, Kara EO, Bôtto-Menezes CHA, da Costa Castilho M, de Oliveira Franca RF, Habib N, Neto AM, Pereira GFM, Giozza SP, Bermúdez XPD, Fernandes TJ, Modjarrad K, Brasil P, Broutet NJN, de Filippis AMB. Detection and persistence of Zika virus in body fluids and associated factors: a prospective cohort study. Sci Rep 2023; 13:21557. [PMID: 38057382 PMCID: PMC10700488 DOI: 10.1038/s41598-023-48493-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
This study aimed to analyze the detection and duration of the Zika virus (ZIKV) in plasma, urine, saliva, sweat, rectal swabs, vaginal secretions, breast milk, and semen and to explore risk factors associated with prolonged viral persistence. A prospective cohort study of symptomatic patients and their household contacts was conducted in Brazil from July 2017 to June 2019. A total of 260 individuals (184 women and 76 men) with confirmed ZIKV infection were enrolled and followed up for 12 months. ZIKV RNA was present in all body fluid specimens and detectable for extended periods in urine, sweat, rectal swabs, and semen. The longest detection duration was found in semen, with high viral loads in the specimens. ZIKV RNA clearance was associated with several factors, including age, sex, education level, body mass index, non-purulent conjunctivitis, joint pain, and whether the participant had a history of yellow fever vaccination. The influence of each of these factors on the low or fast viral clearance varied according to the specific body fluid under investigation. Recurrent ZIKV detection events after total viral clearance were observed in the cohort. Our findings provide valuable insights into the persistence and potential recurrence of ZIKV infection, highlighting the need for continued monitoring and follow-up of individuals infected with ZIKV and for effective prevention measures to reduce the risk of transmission.
Collapse
Affiliation(s)
- Guilherme Amaral Calvet
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Edna Oliveira Kara
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Camila Helena Aguiar Bôtto-Menezes
- Department of Malaria, Tropical Medicine Foundation Doctor Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
- School of Health Sciences, Amazonas State University (UEA), Manaus, Amazonas, Brazil
| | - Marcia da Costa Castilho
- Department of Malaria, Tropical Medicine Foundation Doctor Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
| | | | - Ndema Habib
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Armando Menezes Neto
- Department of Virology and Experimental Therapy, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Gerson Fernando Mendes Pereira
- Department of HIV/AIDS, Tuberculosis, Viral Hepatitis and Sexually Transmitted Infections (DATHI), Ministry of Health, Brasília, Brazil
| | - Silvana Pereira Giozza
- Department of HIV/AIDS, Tuberculosis, Viral Hepatitis and Sexually Transmitted Infections (DATHI), Ministry of Health, Brasília, Brazil
| | | | - Tatiana Jorge Fernandes
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Patrícia Brasil
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Maria Bispo de Filippis
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Alarcon PC, Damen MSMA, Ulanowicz CJ, Sawada K, Oates JR, Toth A, Wayland JL, Chung H, Stankiewicz TE, Moreno-Fernandez ME, Szabo S, Zacharias WJ, Divanovic S. Obesity amplifies influenza virus-driven disease severity in male and female mice. Mucosal Immunol 2023; 16:843-858. [PMID: 37730122 PMCID: PMC10842771 DOI: 10.1016/j.mucimm.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Influenza virus-induced respiratory pneumonia remains a major public health concern. Obesity, metabolic diseases, and female sex are viewed as independent risk factors for worsened influenza virus-induced lung disease severity. However, lack of experimental models of severe obesity in female mice limits discovery-based studies. Here, via utility of thermoneutral housing (30 °C) and high-fat diet (HFD) feeding, we induced severe obesity and metabolic disease in female C57BL/6 mice and compared their responses to severely obese male C57BL/6 counterparts during influenza virus infection. We show that lean male and female mice have similar lung edema, inflammation, and immune cell infiltration during influenza virus infection. At standard housing conditions, HFD-fed male, but not female, mice exhibit severe obesity, metabolic disease, and exacerbated influenza disease severity. However, combining thermoneutral housing and HFD feeding in female mice induces severe obesity and metabolic disease, which is sufficient to amplify influenza virus-driven disease severity to a level comparable to severely obese male counterparts. Lastly, increased total body weights of male and female mice at time of infection correlated with worsened influenza virus-driven disease severity metrics. Together, our findings confirm the impact of obesity and metabolic disease as key risk factors to influenza disease severity and present a novel mouse experimental model suitable for future mechanistic interrogation of sex, obesity, and metabolic disease traits in influenza virus-driven disease severity.
Collapse
Affiliation(s)
- Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Cassidy J Ulanowicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrea Toth
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jennifer L Wayland
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Gastroenterology, Hepatology and Nutrition Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sara Szabo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - William J Zacharias
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
34
|
Graham VA, Easterbrook L, Kennedy E, Rayner E, Findlay-Wilson S, Flett L, Wise EL, Treagus S, Fotheringham S, Kempster S, Almond N, Dowall S. Pathogenesis of Rift Valley Fever Virus in a BALB/c Mouse Model Is Affected by Virus Culture Conditions and Sex of the Animals. Viruses 2023; 15:2369. [PMID: 38140610 PMCID: PMC10747589 DOI: 10.3390/v15122369] [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: 11/06/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen causing disease in livestock and humans. Whilst initially restricted to the African continent, recent spread to the Arabian Peninsula has highlighted the likelihood of entry into new regions. Due to the absence of a regulatory-approved human vaccine, work is ongoing to develop and assess countermeasures. As such, small animal models play a pivotal role in providing information on disease pathogenesis and elucidating which intervention strategies confer protection. To develop and establish the BALB/c mouse model, we challenged mice with RVFV grown from two separate cell lines: one derived from mosquitoes (C6/36) and the other mammalian derived (Vero E6). Following infection, we assessed the clinical course of disease progression at days 1 and 3 post-challenge and evaluated viral tropism and immune analytes. The results demonstrated that RVFV infection was affected by the cell line used to propagate the challenge virus, with those grown in insect cells resulting in a more rapid disease progression. The lowest dose that caused uniform severe disease remained the same across both virus preparations. In addition, to demonstrate reproducibility, the lowest dose was used for a subsequent infection study using male and female animals. The results further demonstrated that male mice succumbed to infection more rapidly than their female counterparts. Our results establish an RVFV mouse model and key parameters that affect the course of disease progression in BALB/c mice.
Collapse
Affiliation(s)
- Victoria A. Graham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Linda Easterbrook
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Emma Kennedy
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Emma Rayner
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Stephen Findlay-Wilson
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Lucy Flett
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Emma Louise Wise
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Samantha Treagus
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Susan Fotheringham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| | - Sarah Kempster
- Medicines and Healthcare Products Regulatory Agency (MHRA), Blanche Ln, South Mimms, Potters Bar EN6 3QG, UK; (S.K.); (N.A.)
| | - Neil Almond
- Medicines and Healthcare Products Regulatory Agency (MHRA), Blanche Ln, South Mimms, Potters Bar EN6 3QG, UK; (S.K.); (N.A.)
| | - Stuart Dowall
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, UK; (V.A.G.); (L.E.); (E.K.); (E.R.); (S.F.-W.); (L.F.); (E.L.W.); (S.T.); (S.F.)
| |
Collapse
|
35
|
Brandt L, Angelino P, Martinez R, Cristinelli S, Ciuffi A. Sex and Age Impact CD4+ T Cell Susceptibility to HIV In Vitro through Cell Activation Dynamics. Cells 2023; 12:2689. [PMID: 38067117 PMCID: PMC10706042 DOI: 10.3390/cells12232689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Cellular composition and the responsiveness of the immune system evolve upon aging and are influenced by biological sex. CD4+ T cells from women living with HIV exhibit a decreased viral replication ex vivo compared to men's. We, thus, hypothesized that these findings could be recapitulated in vitro and infected primary CD4+ T cells with HIV-based vectors pseudotyped with VSV-G or HIV envelopes. We used cells isolated from twenty donors to interrogate the effect of sex and age on permissiveness over a six-day activation kinetics. Our data identified an increased permissiveness to HIV between 24 and 72 h post-stimulation. Sex- and age-based analyses at these time points showed an increased susceptibility to HIV of the cells isolated from males and from donors over 50 years of age, respectively. A parallel assessment of surface markers' expression revealed higher frequencies of activation marker CD69 and of immune checkpoint inhibitors (PD-1 and CTLA-4) in the cells from highly permissive donors. Furthermore, positive correlations were identified between the expression kinetics of CD69, PD-1 and CTLA-4 and HIV expression kinetics. The cell population heterogeneity was assessed using a single-cell RNA-Seq analysis and no cell subtype enrichment was identified according to sex. Finally, transcriptomic analyses further highlighted the role of activation in those differences with enriched activation and cell cycle gene sets in male and older female cells. Altogether, this study brought further evidence about the individual features affecting HIV replication at the cellular level and should be considered in latency reactivation studies for an HIV cure.
Collapse
Affiliation(s)
- Ludivine Brandt
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, 1010 Lausanne, Switzerland; (L.B.)
| | - Paolo Angelino
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, 1010 Lausanne, Switzerland; (L.B.)
- Translational Data Science (TDS)-Facility, AGORA Cancer Research Center, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Raquel Martinez
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, 1010 Lausanne, Switzerland; (L.B.)
| | - Sara Cristinelli
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, 1010 Lausanne, Switzerland; (L.B.)
| | - Angela Ciuffi
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, 1010 Lausanne, Switzerland; (L.B.)
| |
Collapse
|
36
|
Jozwiak M, Doyen D, Denormandie P, Goury A, Marey J, Pène F, Cariou A, Mira JP, Dellamonica J, Nguyen LS. Impact of sex differences on cardiac injury in critically ill patients with COVID-19. Respir Res 2023; 24:292. [PMID: 37986157 PMCID: PMC10662091 DOI: 10.1186/s12931-023-02581-5] [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: 09/09/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND COVID-19 infections are associated with accrued inflammatory responses which may result in cardiac injury. Immune response to infection appears different between men and women, suggesting that COVID-19 patients' outcomes may differ according to biological sex. However, the impact of biological sex on the occurrence of cardiac injury during intensive care unit (ICU) stay in COVID-19 patients remain unclear. METHODS In this multicenter and prospective study, we included consecutive patients admitted to ICU for severe COVID-19 pneumonia, during the first two pandemic waves. Biological, electrocardiogram (ECG) and echocardiographic variables were collected on ICU admission. Cardiac injury was defined by increased troponin above 99th percentile of upper norm value and newly diagnosed ECG and/or echocardiographic abnormalities. The primary endpoint was the proportion of patients with cardiac injury during ICU stay according to biological sex. The impact of biological sex on other subsequent clinical outcomes was also evaluated. RESULTS We included 198 patients with a median age of 66 (56-73) years, 147 (74%) patients were men and 51 (26%) were women. Overall, 119 (60%) patients had cardiac injury during ICU stay and the proportion of patients with cardiac injury during ICU stay was not different between men and women (60% vs. 61%, p = 1.00). Patients with cardiac injury during ICU stay showed more cardiovascular risk factors and chronic cardiac disease and had a higher ICU mortality rate. On ICU admission, they had a more marked lymphopenia (0.70 (0.40-0.80) vs. 0.80 (0.50-1.10) × 109/L, p < 0.01) and inflammation (C-Reactive Protein (155 (88-246) vs. 111 (62-192) mg/L, p = 0.03); D-Dimers (1293 (709-2523) vs. 900 (560-1813) µg/L, p = 0.03)). Plasmatic levels of inflammatory biomarkers on ICU admission correlated with SAPS-2 and SOFA scores but not with the different echocardiographic variables. Multivariate analysis confirmed cardiovascular risk factors (OR = 2.31; 95%CI (1.06-5.02), p = 0.03) and chronic cardiac disease (OR = 8.58; 95%CI (1.01-73.17), p = 0.04) were independently associated with the occurrence of cardiac injury during ICU stay, whereas biological sex (OR = 0.88; 95%CI (0.42-1.84), p = 0.73) was not. Biological sex had no impact on the occurrence during ICU stay of other clinical outcomes. CONCLUSIONS Most critically ill patients with COVID-19 were men and experienced cardiac injury during ICU stay. Nevertheless, biological sex had no impact on the occurrence of cardiac injury during ICU stay or on other clinical outcomes. Clinical trial registration NCT04335162.
Collapse
Affiliation(s)
- Mathieu Jozwiak
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France.
- Université Paris Cité, Paris, France.
- UR2CA, Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, Nice, France.
| | - Denis Doyen
- UR2CA, Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, Nice, France
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Nice, Hôpital L'Archet 1, 151 Rue Saint Antoine de Ginestière, 06200, Nice, France
| | - Pierre Denormandie
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
| | - Antoine Goury
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Reims, Rue du Général Koenig, 51092, Reims, France
| | - Jonathan Marey
- Unité de Soins Intensifs Pneumologiques, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
| | - Frédéric Pène
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
- Université Paris Cité, Paris, France
| | - Alain Cariou
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
- Université Paris Cité, Paris, France
| | - Jean-Paul Mira
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
- Université Paris Cité, Paris, France
| | - Jean Dellamonica
- UR2CA, Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, Nice, France
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Nice, Hôpital L'Archet 1, 151 Rue Saint Antoine de Ginestière, 06200, Nice, France
| | - Lee S Nguyen
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
- Recherche et Innovation, Groupe hospitalier privé Ambroise Paré, Hartmann, 48Ter Bd Victor Hugo, 92200, Neuilly-Sur-Seine, France
| |
Collapse
|
37
|
Grünhagel B, Borggrewe M, Hagen SH, Ziegler SM, Henseling F, Glau L, Thiele RJ, Pujantell M, Sivayoganathan V, Padoan B, Claussen JM, Düsedau A, Hennesen J, Bunders MJ, Bonn S, Tolosa E, Krebs CF, Dorn C, Altfeld M. Reduction of IFN-I responses by plasmacytoid dendritic cells in a longitudinal trans men cohort. iScience 2023; 26:108209. [PMID: 37953956 PMCID: PMC10637924 DOI: 10.1016/j.isci.2023.108209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Type I interferons (IFN-I) are important mediators of antiviral immunity and autoimmune diseases. Female plasmacytoid dendritic cells (pDCs) exert an elevated capacity to produce IFN-I upon toll-like receptor 7 (TLR7) activation compared to male pDCs, and both sex hormones and X-encoded genes have been implicated in these sex-specific differences. Using longitudinal samples from a trans men cohort receiving gender-affirming hormone therapy (GAHT), the impact of testosterone injections on TLR7-mediated IFN-I production by pDCs was assessed. Single-cell RNA analyses of pDCs showed downregulation of IFN-I-related gene expression signatures but also revealed transcriptional inter-donor heterogeneity. Longitudinal quantification showed continuous reduction of IFN-I protein production by pDCs and reduced expression of IFN-I-stimulated genes in peripheral blood mononuclear cells (PBMCs). These studies in trans men demonstrate that testosterone administration reduces IFN-I production by pDCs over time and provide insights into the immune-modulatory role of testosterone in sex-specific IFN-I-mediated immune responses.
Collapse
Affiliation(s)
- Benjamin Grünhagel
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Malte Borggrewe
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
| | - Sven Hendrik Hagen
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Susanne M. Ziegler
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Florian Henseling
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Laura Glau
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Rebecca-Jo Thiele
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Maria Pujantell
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Varshi Sivayoganathan
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- Hamburg Center for Translational Immunology, 20251 Hamburg, Germany
| | - Benedetta Padoan
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Janna M. Claussen
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Arne Düsedau
- Technology Platform Flow Cytometry/FACS, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Jana Hennesen
- Technology Platform Flow Cytometry/FACS, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Madeleine J. Bunders
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
| | - Eva Tolosa
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christian F. Krebs
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- Hamburg Center for Translational Immunology, 20251 Hamburg, Germany
| | | | - Marcus Altfeld
- Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| |
Collapse
|
38
|
Vlachovsky SG, Di Ciano LA, Oddo EM, Azurmendi PJ, Silberstein C, Ibarra FR. Role of Female Sex Hormones and Immune Response in Salt-Sensitive Hypertension Development: Evidence from Experimental Models. Curr Hypertens Rep 2023; 25:405-419. [PMID: 37676461 DOI: 10.1007/s11906-023-01257-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 09/08/2023]
Abstract
PURPOSEOF REVIEW Female sex hormones have systemic effects unrelated to their reproductive function. We describe experiences of different research groups and our own, on aspects related to the importance of female sex hormones on blood pressure (BP) regulation and salt-sensitivity-mediated BP response and salt sensitivity without alterations in BP, as well as renal sodium handling and interactions with the immune system. RECENT FINDINGS Changes in sodium intake in normotensive premenopausal women cause more BP variations than in men. After menopause, women often develop arterial hypertension (HT) with a profile of sodium sensitivity. Besides, experimental results have shown that in adult rat models resembling the postmenopausal hormonal state induced by ovariectomy, controlling BP is not enough to avoid renal and other tissue infiltration with immune cells, which does not occur when sodium intake is low or normal. Therefore, excess sodium promotes an inflammatory state with the involvement of immune cells. The evidence of activation of adaptive immunity, besides changes in T cell subpopulations, includes changes in sodium transporters and receptors. More studies are needed to evaluate the particular sodium sensitivity of women and its meaning. Changes in lifestyle and sodium intake reduction are the main therapeutic steps. However, to face the actual burden of salt-sensitive HT in postmenopausal women and its associated inflammatory/immune changes, it seems reasonable to work on immune cell activity by considering the peripheral blood mononuclear cell phenotypes of molecules and transport proteins related to sodium handle, both to screen for and treat cell activation.
Collapse
Affiliation(s)
- Sandra G Vlachovsky
- Universidad de Buenos Aires, Instituto de Investigaciones Medicas A. Lanari, Laboratorio de Nefrología Experimental y Bioquímica Molecular, Combatientes de Malvinas 3150, Buenos Aires, 1427, Argentina
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
| | - Luis A Di Ciano
- Universidad de Buenos Aires, Instituto de Investigaciones Medicas A. Lanari, Laboratorio de Nefrología Experimental y Bioquímica Molecular, Combatientes de Malvinas 3150, Buenos Aires, 1427, Argentina
| | - Elisabet M Oddo
- Universidad de Buenos Aires, Instituto de Investigaciones Medicas A. Lanari, Laboratorio de Nefrología Experimental y Bioquímica Molecular, Combatientes de Malvinas 3150, Buenos Aires, 1427, Argentina
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
| | - Pablo J Azurmendi
- Universidad de Buenos Aires, Instituto de Investigaciones Medicas A. Lanari, Laboratorio de Nefrología Experimental y Bioquímica Molecular, Combatientes de Malvinas 3150, Buenos Aires, 1427, Argentina
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
| | - Claudia Silberstein
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Instituto de Fisiología y Biofísica B. Houssay (IFIBIO-Houssay), Laboratorio de Fisiología Renal, Paraguay 2155, piso 4, Buenos Aires, 1121, Argentina.
| | - Fernando R Ibarra
- Universidad de Buenos Aires, Instituto de Investigaciones Medicas A. Lanari, Laboratorio de Nefrología Experimental y Bioquímica Molecular, Combatientes de Malvinas 3150, Buenos Aires, 1427, Argentina.
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina.
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Instituto de Fisiología y Biofísica B. Houssay (IFIBIO-Houssay), Laboratorio de Fisiología Renal, Paraguay 2155, piso 4, Buenos Aires, 1121, Argentina.
| |
Collapse
|
39
|
Patlin B, Schwerdtfeger L, Tobet S. Neuropeptide stimulation of physiological and immunological responses in precision-cut lung slices. Physiol Rep 2023; 11:e15873. [PMID: 37994278 PMCID: PMC10665790 DOI: 10.14814/phy2.15873] [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: 06/14/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/24/2023] Open
Abstract
Organotypic lung slices, sometimes known as precision-cut lung slices (PCLS), provide an environment in which numerous cell types and interactions can be maintained outside the body (ex vivo). PCLS were maintained ex vivo for up to a week and demonstrated health via the presence of neurons, maintenance of tissue morphology, synthesis of mucopolysaccharides, and minimal cell death. Multiple phenotypes of neuronal fibers were present in lung slices with varied size, caliber, and neurotransmitter immunoreactivity. Of the neuropeptides present in fibers, calcitonin gene-related peptide (CGRP) was the most prevalent. Exposing PCLS to recombinant CGRP resulted in the proliferation and dispersion of CD19+ B cells in slices taken selectively from females. The number of granules containing immunoreactive (ir) surfactant protein C (SPC), which are representative of alveolar type 2 cells, increased in slices from females within 24 h of exposure to CGRP. Additionally, ir-SPC granule size increased in slices from males and females across 48 h of exposure to CGRP. Exposure of PCLS to exogenous CGRP did not alter the number of solitary pulmonary neuroendocrine cells (PNEC) but did result in neuroendocrine bodies that had significantly more cells. Neuronal fiber numbers were unchanged based on ir-peripherin; however, ir-CGRP became non-detectable in fibers while unchanged in PNECs. The effects of exogenous CGRP provide insight into innate immune and neuroendocrine responses in the lungs that may be partially regulated by neural fibers. The sex-dependent nature of these changes may point to the basis for sex-selective outcomes among respiratory diseases.
Collapse
Affiliation(s)
- B. Patlin
- Department of Biochemistry and Molecular BiologyColorado State UniversityFort CollinsColoradoUSA
| | - L. Schwerdtfeger
- Department of NeurologyHarvard Medical School and Ann Romney Center for Neurologic Diseases, Brigham and Women's HospitalBostonMassachusettsUSA
| | - S. Tobet
- School of Biomedical Engineering and Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
40
|
Kelkar NS, Goldberg BS, Dufloo J, Bruel T, Schwartz O, Hessell AJ, Ackerman ME. Sex and species associated differences in Complement-mediated immunity in Humans and Rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563614. [PMID: 37961263 PMCID: PMC10634758 DOI: 10.1101/2023.10.23.563614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The complement system can be viewed as a 'moderator' of innate immunity, 'instructor' of humoral immunity, and 'regulator' of adaptive immunity. While sex and aging are known to affect humoral and cellular immune systems, their impact on the complement pathway in humans and rhesus macaques, a commonly used non-human primate model system, have not been well-studied. To address this knowledge gap, we analyzed serum samples from 90 humans and 75 rhesus macaques for the abundance and activity of the complement system components. While sequences of cascade proteins were highly conserved, dramatically different levels were observed between species. Whereas the low levels detected in rhesus samples raised questions about the suitability of the test, differences in levels of complement proteins were observed in male and female humans. Levels of total and antibody-dependent deposition of C1q and C3b on a glycosylated antigen differed between human and rhesus, suggesting differential recognition of glycans. Functional differences in complement-mediated lysis of antibody-sensitized cells were observed in multiple assays and showed that human females frequently exhibited higher lytic activity than human males or rhesus macaques, which typically did not exhibit such sexual dimorphism. Other differences between species and sexes were observed in more narrow contexts-for only certain antibodies, antigens, or assays. Collectively, these results expand our knowledge of sexual dimorphism in the complement system in humans, identifying differences that appear to be absent from rhesus macaques.
Collapse
Affiliation(s)
- Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Benjamin S. Goldberg
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Present Address: Metaphore Biotechnologies Inc., Cambridge, MA, USA
| | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Present Address: Institute for Integrative Systems Biology (I2SysBio), Universitat da Valencia-CSIC, 46980 Valencia, Spain
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Vaccine Research Institute, 9400 Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Vaccine Research Institute, 9400 Créteil, France
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
41
|
Chen C, Talifu Z, Wu Y, Su B, Dai W. Changing Patterns of Mortality in Viral Hepatitis - China, 1987-2021. China CDC Wkly 2023; 5:933-937. [PMID: 38026100 PMCID: PMC10646165 DOI: 10.46234/ccdcw2023.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
What is already known about this topic? Viral hepatitis continues to present a major global public health challenge, with China shouldering the heaviest burden of this disease worldwide. What is added by this report? This study examined evolving trends and assessed the impacts of age, period, and cohort on viral hepatitis mortality from 1987 to 2021 in both urban and rural settings across China. What are the implications for public health practice? This research provides critical insights, enabling policymakers to develop precise and effective intervention strategies that are specifically tailored to address the needs of high-risk older adults.
Collapse
Affiliation(s)
- Chen Chen
- Department of Population Health and Aging Science, School of Population Medicine and Public Health, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yu Wu
- Department of Population Health and Aging Science, School of Population Medicine and Public Health, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Binbin Su
- Department of Health Economics, School of Population Medicine and Public Health, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Wanwei Dai
- Office of Academic Research of Peking University Third Hospital, Beijing, China
| |
Collapse
|
42
|
Sengupta A, Al-Otaibi N, Hinkula J. Sex-Specific Immune Responses to Seasonal Influenza Vaccination in Diabetic Individuals: Implications for Vaccine Efficacy. J Immunol Res 2023; 2023:3111351. [PMID: 37881338 PMCID: PMC10597737 DOI: 10.1155/2023/3111351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 10/27/2023] Open
Abstract
Seasonal influenza vaccination has different implications on the immune response depending on the comorbidities. Diabetes is one such critical disease that increases the patient's susceptibility to influenza and suppresses vaccine efficacy and immunity. The sex of the individuals also plays a definitive role in the immune responses to both the vaccine and the infection. This study aims to understand the efficacy of the seasonal vaccine against influenza in diabetic groups and undergoing immune mechanisms in different sexes (females and males). In this study, we are reporting about a switching of the immune response of the infected and vaccinated diabetic females towards stronger Th1/Th17 responses with suppressed humoral immunity. They show increased cDC1, enhanced proinflammatory activities within T cells, CD8T activation, Th17 proliferation, and the majority of IgG2 antibody subtypes with reduced neutralization potential. Males with diabetes exhibit enhanced humoral Th2-immunity than the nondiabetic group. They exhibit higher cDC2, and DEC205 levels within them with an increase in plasma B lymphocytes, higher IgG1 subtypes in plasma cells, and influenza-hemagglutinin-specific IgG titer with stronger virus neutralization potential. Males with diabetes recovered better than the females as observed from the changes in their body weight. This study highlights the critical immune mechanisms and sex-specific swapping of their preferred immune response pathways against influenza after vaccination during diabetes. We propose a need for a sex-specific customized vaccine regimen to be implemented against influenza for individuals having diabetes to exploit the manifested strength and weakness in their protective immunity.
Collapse
Affiliation(s)
- Anirban Sengupta
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden
| | - Noha Al-Otaibi
- King Abdulaziz City for Science and Technology (KACST), Riyad 11442, Saudi Arabia
| | - Jorma Hinkula
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden
| |
Collapse
|
43
|
Vancolen S, Sébire G, Robaire B. Influence of androgens on the innate immune system. Andrology 2023; 11:1237-1244. [PMID: 36840517 DOI: 10.1111/andr.13416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND Sexual dimorphism is observed in the occurrence, course, and severity of human disease. The difference in immune response between males and females can in part be attributed to sexual genotype. However, immunological differences can also be explained by endocrine-immune interactions. Specifically, androgens possess the ability of directly modulating the development and function of immune cells. Although androgens generally contribute to immunosuppressive effects, this is not necessarily always the case. AIM The aim of the review is to uncover the role of androgens in shaping the innate immune response. MATERIAL & METHODS Authors included papers in this review which discussed the impact of androgens on specific innate immune cells. RESULTS Androgens modulate the innate immune response through various mechanisms. However, there is conflicting evidence in the literature regarding the interplay betwen androgens and the innate immune system. DISCUSSION Conflicting evidence presented in this review could in part be explained by the limitations present in interpreting results. CONCLUSION This review is of great importance for our understanding of occurence and mechanism of human inflammatory disease.
Collapse
Affiliation(s)
- Seline Vancolen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics and Department of Neurology, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Guillaume Sébire
- Department of Pediatrics and Department of Neurology, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Bernard Robaire
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
44
|
Abdulkader MA, Merza MA. Immediate and Long-Term Adverse Events of COVID-19 Vaccines: A One-Year Follow-Up Study From the Kurdistan Region of Iraq. Cureus 2023; 15:e47670. [PMID: 38021955 PMCID: PMC10671599 DOI: 10.7759/cureus.47670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background The administration of COVID-19 vaccines has been critical in controlling the spread of the virus. However, understanding the potential adverse events (AEs) associated with these vaccines is crucial for public health. While most previous studies observed only short-term AEs, this study aimed to investigate the immediate and long-term AEs following the first and second doses of Pfizer, AstraZeneca, and Sinopharm vaccines, providing valuable long-term insights. Methodology A prospective, one-year, follow-up study was conducted by tracking 922 vaccinated individuals to assess short-term and long-term AEs. Demographics, clinical characteristics, vaccine types, and dose effects were taken into consideration. AEs were classified based on severity and duration. Statistical analyses were performed to compare differences among the vaccine groups, with p-values <0.05 considered significant. Bowker's and chi-square tests were performed using JMP Pro 14.3.0. Results Of the 922 participants, 55.53% (n = 512) were vaccinated with Pfizer, and 23.32% (n = 215) and 21.15% (n = 195) were vaccinated with Sinopharm and AstraZeneca, respectively. Overall, 72.34% of participants (n = 667) were suffering from AEs after the first dose, with a lower prevalence of AEs after the second dose (52.71%, n = 486). Pfizer exhibited the highest percentage and severity of AEs, followed by AstraZeneca and Sinopharm. Most AEs reported in this study were mild and resolved within 72 hours, with females experiencing more frequent AEs. The common short-term AEs observed were fever, injection-site pain, myalgia, fatigue, and headache. Notably, there were no chronic AEs, and only one case of myocarditis was associated with AstraZeneca. Conclusions Despite the variation in the prevalence of AEs among the three vaccines, the vaccination process proved to be safe with no serious short-term AEs. However, the long-term AEs associated with AstraZeneca and the decrease in the prevalence of AEs after the second dose of the COVID-19 vaccines warrant further investigations and priority for future research.
Collapse
Affiliation(s)
- Manhal A Abdulkader
- Department of Clinical Pharmacy, College of Pharmacy, University of Duhok, Duhok, IRQ
| | - Muayad A Merza
- Department of Internal Medicine, University of Duhok, Duhok, IRQ
| |
Collapse
|
45
|
Fardisi M, Thelen K, Groenendal A, Rajput M, Sebastian K, Contreras GA, Moeser AJ. Early weaning and biological sex shape long-term immune and metabolic responses in pigs. Sci Rep 2023; 13:15907. [PMID: 37741873 PMCID: PMC10517948 DOI: 10.1038/s41598-023-42553-9] [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: 06/23/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023] Open
Abstract
During the early pre and postnatal life, host and environmental factors can impart a major influence on immune development, thus shaping lifelong disease resistance. Two major factors known to influence immune function and mortality in animals and people are early life stress and biological sex. How these two factors interact to shape long-term immune development and later life disease risk is poorly understood. Here we investigated how early weaning, a common early life stressor in pigs, and biological sex impacts long-term systemic inflammatory responses and hypothalamic-pituitary-adrenal axis (HPA axis) activation later in life. Ten-week-old female (F), intact-male (IM) and castrated-male (CM) pigs that were randomly assigned to early weaning (EW) and later weaning (LW) (at 15 or 28 days of age, respectively) were intramuscularly injected with either saline vehicle or lipopolysaccharide (LPS) to induce a systemic inflammatory response. Complete blood counts (CBC), proinflammatory cytokines, cortisol, testosterone, estradiol, and rectal temp were measured at 0 h, 2 h, and 4 h post-LPS challenge. At 4 h post-LPS, peritoneal fluid (PF) and white blood cells (WBC) were collected for differential analysis. LPS challenge significantly increased rectal temp and plasma cortisol level in all treatment groups. Together, the CBC results and immune cell counts in peritoneal cavity indicated that EW-F exhibited greater systemic immune response characterized by increased neutrophils to lymphocytes ratio (NLR) and enhanced neutrophil trafficking to the peritoneal cavity. Early weaning had an opposite effect on IM and CM pigs, which exhibited a suppressed LPS-induced neutrophil migration. Early weaning induced significantly greater cortisol responses only in IM pigs indicating a heightened HPA axis responses in EW-IM. how early weaning and biological sex affect immune and stress responses in pigs. Together, these results demonstrate that early weaning and biological sex and castration shape later life immune responses in pigs and provides insight into potential mechanisms driving sex differences in later life inflammatory disease risk and mortality.
Collapse
Affiliation(s)
- Mahsa Fardisi
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Kyan Thelen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Allegra Groenendal
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Mrigendra Rajput
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Kimberly Sebastian
- Department of Pathology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Adam J Moeser
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
46
|
Crawford JD, Wang H, Trejo-Zambrano D, Cimbro R, Talbot CC, Thomas MA, Curran AM, Girgis AA, Schroeder JT, Fava A, Goldman DW, Petri M, Rosen A, Antiochos B, Darrah E. The XIST lncRNA is a sex-specific reservoir of TLR7 ligands in SLE. JCI Insight 2023; 8:e169344. [PMID: 37733447 PMCID: PMC10634230 DOI: 10.1172/jci.insight.169344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with a dramatic sex bias, affecting 9 times more women than men. Activation of Toll-like receptor 7 (TLR7) by self-RNA is a central pathogenic process leading to aberrant production of type I interferon (IFN) in SLE, but the specific RNA molecules that serve as TLR7 ligands have not been defined. By leveraging gene expression data and the known sequence specificity of TLR7, we identified the female-specific X-inactive specific transcript (XIST) long noncoding RNA as a uniquely rich source of TLR7 ligands in SLE. XIST RNA stimulated IFN-α production by plasmacytoid DCs in a TLR7-dependent manner, and deletion of XIST diminished the ability of whole cellular RNA to activate TLR7. XIST levels were elevated in blood leukocytes from women with SLE compared with controls, correlated positively with disease activity and the IFN signature, and were enriched in extracellular vesicles released from dying cells in vitro. Importantly, XIST was not IFN inducible, suggesting that XIST is a driver, rather than a consequence, of IFN in SLE. Overall, our work elucidated a role for XIST RNA as a female sex-specific danger signal underlying the sex bias in SLE.
Collapse
Affiliation(s)
| | - Hong Wang
- Division of Rheumatology, Department of Medicine
| | | | | | - C. Conover Talbot
- The Single Cell and Transcriptomics Core, Institute for Basic Biomedical Sciences; and
| | | | | | | | - John T. Schroeder
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea Fava
- Division of Rheumatology, Department of Medicine
| | | | | | - Antony Rosen
- Division of Rheumatology, Department of Medicine
| | | | - Erika Darrah
- Division of Rheumatology, Department of Medicine
| |
Collapse
|
47
|
Taylor M, Rayner JO. Immune Response to Chikungunya Virus: Sex as a Biological Variable and Implications for Natural Delivery via the Mosquito. Viruses 2023; 15:1869. [PMID: 37766276 PMCID: PMC10538149 DOI: 10.3390/v15091869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne virus with significant public health implications around the world. Climate change, as well as rapid urbanization, threatens to expand the population range of Aedes vector mosquitoes globally, increasing CHIKV cases worldwide in return. Epidemiological data suggests a sex-dependent response to CHIKV infection. In this review, we draw attention to the importance of studying sex as a biological variable by introducing epidemiological studies from previous CHIKV outbreaks. While the female sex appears to be a risk factor for chronic CHIKV disease, the male sex has recently been suggested as a risk factor for CHIKV-associated death; however, the underlying mechanisms for this phenotype are unknown. Additionally, we emphasize the importance of including mosquito salivary components when studying the immune response to CHIKV. As with other vector-transmitted pathogens, CHIKV has evolved to use these salivary components to replicate more extensively in mammalian hosts; however, the response to natural transmission of CHIKV has not been fully elucidated.
Collapse
Affiliation(s)
| | - Jonathan O. Rayner
- Department of Microbiology & Immunology, Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA;
| |
Collapse
|
48
|
Petersen PT, Bodilsen J, Jepsen MPG, Larsen L, Storgaard M, Hansen BR, Helweg-Larsen J, Wiese L, Lüttichau HR, Andersen CØ, Nielsen H, Brandt CT. Clinical features and prognostic factors in adults with viral meningitis. Brain 2023; 146:3816-3825. [PMID: 36929167 PMCID: PMC10473559 DOI: 10.1093/brain/awad089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/08/2023] [Accepted: 02/25/2023] [Indexed: 03/18/2023] Open
Abstract
Clinical features applicable to the entire spectrum of viral meningitis are limited, and prognostic factors for adverse outcomes are undetermined. This nationwide population-based prospective cohort study included all adults with presumed and microbiologically confirmed viral meningitis in Denmark from 2015 until 2020. Prognostic factors for an unfavourable outcome (Glasgow Outcome Scale score of 1-4) 30 days after discharge were examined by modified Poisson regression. In total, 1066 episodes of viral meningitis were included, yielding a mean annual incidence of 4.7 episodes per 100 000 persons. Pathogens were enteroviruses in 419/1066 (39%), herpes simplex virus type 2 in 171/1066 (16%), varicella-zoster virus in 162/1066 (15%), miscellaneous viruses in 31/1066 (3%) and remained unidentified in 283/1066 (27%). The median age was 33 years (IQR 27-44), and 576/1066 (54%) were females. In herpes simplex virus type 2 meningitis, 131/171 (77%) were females. Immunosuppression [32/162 (20%)] and shingles [90/149 (60%)] were frequent in varicella-zoster virus meningitis. The triad of headache, neck stiffness and hyperacusis or photophobia was present in 264/960 (28%). The median time until lumbar puncture was 3.0 h (IQR 1.3-7.1), and the median CSF leucocyte count was 160 cells/µl (IQR 60-358). The outcome was unfavourable in 216/1055 (20%) 30 days after discharge. Using unidentified pathogen as the reference, the adjusted relative risk of an unfavourable outcome was 1.34 (95% CI 0.95-1.88) for enteroviruses, 1.55 (95% CI 1.00-2.41) for herpes simplex virus type 2, 1.51 (95% CI 0.98-2.33) for varicella-zoster virus and 1.37 (95% CI 0.61-3.05) for miscellaneous viruses. The adjusted relative risk of an unfavourable outcome was 1.34 (95% CI 1.03-1.75) for females. Timing of acyclovir or valacyclovir was not associated with the outcome in meningitis caused by herpes simplex virus type 2 or varicella-zoster virus. In summary, the outcome of viral meningitis was similar among patients with different aetiologies, including those with presumed viral meningitis but without an identified pathogen. Females had an increased risk of an unfavourable outcome. Early antiviral treatment was not associated with an improved outcome in meningitis caused by herpes simplex virus type 2 or varicella-zoster virus.
Collapse
Affiliation(s)
- Pelle Trier Petersen
- Department of Pulmonary and Infectious Diseases, Nordsjællands Hospital, 3400 Hillerød, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jacob Bodilsen
- Department of Infectious Diseases, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | | | - Lykke Larsen
- Department of Infectious Diseases, Odense University Hospital, 5000 Odense, Denmark
| | - Merete Storgaard
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | | | | | - Lothar Wiese
- Department of Medicine, Sjællands University Hospital, 4000 Roskilde, Denmark
| | | | | | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | | | | |
Collapse
|
49
|
Roth L, Michl P, Rosendahl J. [Sex-specific differences in gastroenterological diseases]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023; 64:736-743. [PMID: 36884055 DOI: 10.1007/s00108-023-01491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/09/2023]
Abstract
Diseases of the gastrointestinal tract present with substantial sex differences that have a potential impact on patient outcome. This fact is not sufficiently addressed either in basic research or in clinical studies. For example, most animal studies utilize male animals. Despite differences in incidence, sex may affect complication rates, prognosis, or therapeutic response. The incidence of gastrointestinal cancers is frequently higher in males, but this observation cannot solely rely on a distinct risk behaviour. Here, differences in immune response and p53 signalling may be factors responsible for this finding. Nevertheless, taking sex differences into account and improving our understanding of relevant mechanisms is crucial and will most likely have a substantial impact on disease outcome. This overview aims to highlight sex differences in the context of various gastroenterological diseases, primarily to enhance awareness. Attention to sex-specific differences is essential to improve individualized treatment.
Collapse
Affiliation(s)
- Laura Roth
- Universitätsklinik und Poliklinik für Innere Medizin I, Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland.
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| | - Patrick Michl
- Universitätsklinik und Poliklinik für Innere Medizin I, Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland
- Klinik für Innere Medizin IV, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Jonas Rosendahl
- Universitätsklinik und Poliklinik für Innere Medizin I, Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland.
| |
Collapse
|
50
|
Horn C, Sprute R, Kretschmer AC, Do C, Cornely OA, Jung N, Lehmann C, Fischer J. [Sex in infectious diseases-How sex differences influence the immune response to infections]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023; 64:752-757. [PMID: 37016079 PMCID: PMC10072806 DOI: 10.1007/s00108-023-01498-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 04/05/2023]
Abstract
The humoral and cellular immune responses to antigen stimulation, vaccinations and infections differ between women and men. Genetic, epigenetic and hormonal factors contribute to the sex-specific immunity. The expression of genes on the X‑chromosome and the effect of sex hormones substantially influence the immune defence against infections. Females show stronger cellular and humoral immune responses to infections than males, but the enhanced immune response often leads to aberrant inflammatory reactions and autoimmune diseases. Men are principally more prone to bacterial, viral and fungal infections and more often show severe disease courses. In contrast, a more reactive female immune system results in significantly more adverse reactions to vaccinations. In order to be able to better identify the multiple sex-specific that have an influence on the immune system, sex-specific differences should be investigated in a differentiated way. The better understanding of the sex-specific differences in the immune response will have a long-term influence on the prevention, diagnostics and treatment of infectious diseases, and will ultimately contribute to improving healthcare of both women and men.
Collapse
Affiliation(s)
- Carola Horn
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland
| | - Rosanne Sprute
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Lehrstuhl für Translationale Forschung, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Universität zu Köln, Köln, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Köln, Excellenz Zentrum Medizinische Mykologie (ECMM), Universität zu Köln, Köln, Deutschland
| | - Alina Chloé Kretschmer
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Zentrum für Molekulare Medizin (ZMMK), Universität zu Köln, Köln, Deutschland
| | - Carolin Do
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Zentrum für Molekulare Medizin (ZMMK), Universität zu Köln, Köln, Deutschland
| | - Oliver A Cornely
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Lehrstuhl für Translationale Forschung, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Universität zu Köln, Köln, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Zentrum für klinische Studien (ZKS Köln), Universität zu Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Zentrum für Molekulare Medizin (ZMMK), Universität zu Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Köln, Excellenz Zentrum Medizinische Mykologie (ECMM), Universität zu Köln, Köln, Deutschland
| | - Norma Jung
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
| | - Clara Lehmann
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland
- Medizinische Fakultät und Uniklinik Köln, Zentrum für Molekulare Medizin (ZMMK), Universität zu Köln, Köln, Deutschland
| | - Julia Fischer
- Medizinische Fakultät und Uniklinik Köln, Klinik 1 für Innere Medizin, Universität zu Köln, Köln, Deutschland.
- Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Bonn-Köln, Köln, Deutschland.
- Medizinische Fakultät und Uniklinik Köln, Zentrum für Molekulare Medizin (ZMMK), Universität zu Köln, Köln, Deutschland.
- Klinik 1 für Innere Medizin, Uniklinik Köln, Kerpener Str. 62, 50931, Köln, Deutschland.
| |
Collapse
|