101
|
Walker KA, Le Page LM, Terrando N, Duggan MR, Heneka MT, Bettcher BM. The role of peripheral inflammatory insults in Alzheimer's disease: a review and research roadmap. Mol Neurodegener 2023; 18:37. [PMID: 37277738 PMCID: PMC10240487 DOI: 10.1186/s13024-023-00627-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
Peripheral inflammation, defined as inflammation that occurs outside the central nervous system, is an age-related phenomenon that has been identified as a risk factor for Alzheimer's disease. While the role of chronic peripheral inflammation has been well characterized in the context of dementia and other age-related conditions, less is known about the neurologic contribution of acute inflammatory insults that take place outside the central nervous system. Herein, we define acute inflammatory insults as an immune challenge in the form of pathogen exposure (e.g., viral infection) or tissue damage (e.g., surgery) that causes a large, yet time-limited, inflammatory response. We provide an overview of the clinical and translational research that has examined the connection between acute inflammatory insults and Alzheimer's disease, focusing on three categories of peripheral inflammatory insults that have received considerable attention in recent years: acute infection, critical illness, and surgery. Additionally, we review immune and neurobiological mechanisms which facilitate the neural response to acute inflammation and discuss the potential role of the blood-brain barrier and other components of the neuro-immune axis in Alzheimer's disease. After highlighting the knowledge gaps in this area of research, we propose a roadmap to address methodological challenges, suboptimal study design, and paucity of transdisciplinary research efforts that have thus far limited our understanding of how pathogen- and damage-mediated inflammatory insults may contribute to Alzheimer's disease. Finally, we discuss how therapeutic approaches designed to promote the resolution of inflammation may be used following acute inflammatory insults to preserve brain health and limit progression of neurodegenerative pathology.
Collapse
Affiliation(s)
- Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute On Aging. Baltimore, Baltimore, MD, USA.
| | - Lydia M Le Page
- Departments of Physical Therapy and Rehabilitation Science, and Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Cell Biology and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute On Aging. Baltimore, Baltimore, MD, USA
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
102
|
Nakai M, Yokoyama D, Sato T, Sato R, Kojima C, Shimosawa T. Variation in antibody titers determined by Abbott and Roche Elecsys SARS-CoV-2 assays in vaccinated healthcare workers. Heliyon 2023; 9:e16547. [PMID: 37235203 PMCID: PMC10201891 DOI: 10.1016/j.heliyon.2023.e16547] [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: 05/09/2022] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
SARS-CoV-2-specific antibody measurement is important for evaluating COVID-19 vaccine efficacy. We quantified and compared anti-spike (S) antibodies using different commercial immunoassays. We tested serum samples from 70 SARS-CoV-2-naive health care workers 2 weeks after vaccination with a single dose of BNT162b2, 2 and 4 weeks, and 3 months after the second dose of BNT162b2. The following quantitative assays were used: Roche Elecsys Anti-SARS-CoV-2 S (Roche-S), Abbott SARS-CoV-2 IgG II Quant [Abbott-IgG(S)], and Abbott SARS-CoV-2 IgM (Abbott-IgM). All samples tested positive for Roche-S and Abbott-IgG antibodies after the second dose, with 83.6% Abbott-IgM positive rate. Roche-S and Abbott-IgG(S) correlated significantly in all samples (r = 0.920, p < 0.0001), and the Roche-S and Abbott-IgG(S) assay showed a strong correlation with each other at each time point after vaccination. Roche-S and Abbott-IgG(S) antibody titers were correlated with age; their rate of decline was age-dependent in males but not in females. Abbott-IgG(S) antibody titers decreased from 2 weeks after the second dose. Roche-S antibody titers peaked 2 weeks after the second dose in 76.2% of the participants; the titers recovered 3 months post-vaccination after declining at week 4 in 40.7% of the participants. The concordance between Roche-S and Abbott-IgG(S) antibody titers over time was 47.5%. Most participants presented significantly high Roche-S and Abbott-IgG(S) antibody titers after immunization. Some measurements were inconsistent with titer changes between these assays, possibly because of differences in the immunoglobulin-specificity of the kits.
Collapse
Affiliation(s)
- Miku Nakai
- Department of Clinical Laboratory, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Daisuke Yokoyama
- Department of Clinical Laboratory, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Tomoaki Sato
- Department of Clinical Laboratory, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Ryohei Sato
- Department of Clinical Laboratory, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Chiari Kojima
- Department of Clinical Laboratory, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, International University of Health and Welfare Narita Hospital, Chiba, Japan
| |
Collapse
|
103
|
Wang S, Onyeaghala GC, Pankratz N, Nelson HH, Thyagarajan B, Tang W, Norby FL, Ugoji C, Joshu CE, Gomez CR, Couper DJ, Coresh J, Platz EA, Prizment AE. Associations between MICA and MICB Genetic Variants, Protein Levels, and Colorectal Cancer: Atherosclerosis Risk in Communities (ARIC). Cancer Epidemiol Biomarkers Prev 2023; 32:784-794. [PMID: 36958849 PMCID: PMC10239349 DOI: 10.1158/1055-9965.epi-22-1113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/24/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND The MHC class I chain-related protein A (MICA) and protein B (MICB) participate in tumor immunosurveillance and may be important in colorectal cancer, but have not been examined in colorectal cancer development. METHODS sMICA and sMICB blood levels were measured by SomaScan in Visit 2 (1990-92, baseline) and Visit 3 (1993-95) samples in cancer-free participants in the Atherosclerosis Risk in Communities Study. We selected rs1051792, rs1063635, rs2516448, rs3763288, rs1131896, rs2596542, and rs2395029 that were located in or in the vicinity of MICA or MICB and were associated with cancer or autoimmune diseases in published studies. SNPs were genotyped by the Affymetrix Genome-Wide Human SNP Array. We applied linear and Cox proportional hazards regressions to examine the associations of preselected SNPs with sMICA and sMICB levels and colorectal cancer risk (236 colorectal cancers, 8,609 participants) and of sMICA and sMICB levels with colorectal cancer risk (312 colorectal cancers, 10,834 participants). In genetic analyses, estimates adjusted for ancestry markers were meta-analyzed. RESULTS Rs1051792-A, rs1063635-A, rs2516448-C, rs3763288-A, rs2596542-T, and rs2395029-G were significantly associated with decreased sMICA levels. Rs2395029-G, in the vicinity of MICA and MICB, was also associated with increased sMICB levels. Rs2596542-T was significantly associated with decreased colorectal cancer risk. Lower sMICA levels were associated with lower colorectal cancer risk in males (HR = 0.68; 95% confidence interval, 0.49-0.96) but not in females (Pinteraction = 0.08). CONCLUSIONS Rs2596542-T associated with lower sMICA levels was associated with decreased colorectal cancer risk. Lower sMICA levels were associated with lower colorectal cancer risk in males. IMPACT These findings support an importance of immunosurveillance in colorectal cancer.
Collapse
Affiliation(s)
- Shuo Wang
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Guillaume C. Onyeaghala
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Heather H Nelson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Faye L. Norby
- Department of Cardiology, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Chinenye Ugoji
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Christian R. Gomez
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS
| | - David J. Couper
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Anna E. Prizment
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
104
|
Bugaytsova JA, Moonens K, Piddubnyi A, Schmidt A, Edlund JO, Lisiutin G, Brännström K, Chernov YA, Thorel K, Tkachenko I, Sharova O, Vikhrova I, Butsyk A, Shubin P, Chyzhma R, Johansson DX, Marcotte H, Sjöström R, Shevtsova A, Bylund G, Rakhimova L, Lundquist A, Berhilevych O, Kasianchuk V, Loboda A, Ivanytsia V, Hultenby K, Persson MAA, Gomes J, Matos R, Gartner F, Reis CA, Whitmire JM, Merrell DS, Pan-Hammarström Q, Landström M, Oscarson S, D’Elios MM, Agreus L, Ronkainen J, Aro P, Engstrand L, Graham DY, Kachkovska V, Mukhopadhyay A, Chaudhuri S, Karmakar BC, Paul S, Kravets O, Camorlinga M, Torres J, Berg DE, Moskalenko R, Haas R, Remaut H, Hammarström L, Borén T. Helicobacter pylori attachment-blocking antibodies protect against duodenal ulcer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542096. [PMID: 37292721 PMCID: PMC10245814 DOI: 10.1101/2023.05.24.542096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.
Collapse
Affiliation(s)
- Jeanna A. Bugaytsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
| | - Kristof Moonens
- Structural and Molecular Microbiology, VIB Department of Structural Biology, VIB, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Present address: Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Artem Piddubnyi
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Alexej Schmidt
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
- Present address: Department of Medical Biosciences, Umeå University, SE90185 Umeå, Sweden
| | - Johan Olofsson Edlund
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- The Biochemical Imaging Center Umeå (BICU), Umeå University, SE90187 Umeå, Sweden
| | - Gennadii Lisiutin
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Microbiology, Virology and Biotechnology, Odesa Mechnikov National University, 65082 Odesa, Ukraine
| | - Kristoffer Brännström
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- The Biochemical Imaging Center Umeå (BICU), Umeå University, SE90187 Umeå, Sweden
- Present address: Pfizer Worldwide R&D, BioMedicine Design, 10 555 Science Center Drive, San Diego CA, 92121 USA
| | - Yevgen A. Chernov
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Kaisa Thorel
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Iryna Tkachenko
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Oleksandra Sharova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Iryna Vikhrova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Anna Butsyk
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Pavlo Shubin
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Ruslana Chyzhma
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Daniel X. Johansson
- Department of Clinical Neuroscience, Karolinska Institutet at Center for Molecular Medicine, Karolinska University Hospital, Solna, SE17176 Stockholm, Sweden
| | - Harold Marcotte
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Rolf Sjöström
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Anna Shevtsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Göran Bylund
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Lena Rakhimova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Present address: Department of Odontology, Umeå University, SE90187 Umeå, Sweden
| | - Anders Lundquist
- Department of Statistics, USBE, Umeå University, SE90187 Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, SE90187 Umeå, Sweden
| | - Oleksandra Berhilevych
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Victoria Kasianchuk
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Andrii Loboda
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Volodymyr Ivanytsia
- Department of Microbiology, Virology and Biotechnology, Odesa Mechnikov National University, 65082 Odesa, Ukraine
| | - Kjell Hultenby
- Departments of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
| | - Mats A. A. Persson
- Department of Clinical Neuroscience, Karolinska Institutet at Center for Molecular Medicine, Karolinska University Hospital, Solna, SE17176 Stockholm, Sweden
| | - Joana Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Rita Matos
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Fátima Gartner
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Celso A. Reis
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | | | - D. Scott Merrell
- Department of Microbiology and Immunology, USUHS, Bethesda, MD 20814, USA
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Maréne Landström
- Present address: Department of Medical Biosciences, Umeå University, SE90185 Umeå, Sweden
| | - Stefan Oscarson
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Mario M. D’Elios
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134 Firenze, Italy
| | - Lars Agreus
- Division of Family Medicine and Primary Care, Karolinska Institutet, SE14183 Huddinge, Sweden
| | - Jukka Ronkainen
- University of Oulu, Center for Life Course Health Research and Primary Health Care Center, Tornio Finland
| | - Pertti Aro
- University of Oulu, Center for Life Course Health Research and Primary Health Care Center, Tornio Finland
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE17177 Stockholm, Sweden
- Present address: Science for Life Laboratory, SE17165, Solna, Sweden
| | - David Y. Graham
- Department of Medicine, Molecular Virology and Microbiology, Baylor College of Medicine, Michael E. DeBakey VAMC, 2002 Holcombe Blvd. Houston, TX, 77030 USA
| | - Vladyslava Kachkovska
- Department of Internal Medicine, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Asish Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Sujit Chaudhuri
- Department of Gastroenterology, AMRI Hospital, Salt Lake City. Kolkata, West Bengal 700098, India
| | - Bipul Chandra Karmakar
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Sangita Paul
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Oleksandr Kravets
- Department of Surgery, Traumatology, Orthopedics and Physiology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Margarita Camorlinga
- Unidad de Investigacion en Enfermedades Infecciosas, UMAE Pediatria, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Javier Torres
- Unidad de Investigacion en Enfermedades Infecciosas, UMAE Pediatria, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Douglas E. Berg
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Roman Moskalenko
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Rainer Haas
- German Center for Infection Research (DZIF), Munich Site, 80336 Munich, Germany
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer-Institute, Faculty of Medicine, LMU Munich, Germany
| | - Han Remaut
- Structural and Molecular Microbiology, VIB Department of Structural Biology, VIB, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Lennart Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Thomas Borén
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Lead contact
| |
Collapse
|
105
|
Grifoni A, Alonzi T, Alter G, Noonan DM, Landay AL, Albini A, Goletti D. Impact of aging on immunity in the context of COVID-19, HIV, and tuberculosis. Front Immunol 2023; 14:1146704. [PMID: 37292210 PMCID: PMC10246744 DOI: 10.3389/fimmu.2023.1146704] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023] Open
Abstract
Knowledge of aging biology needs to be expanded due to the continuously growing number of elderly people worldwide. Aging induces changes that affect all systems of the body. The risk of cardiovascular disease and cancer increases with age. In particular, the age-induced adaptation of the immune system causes a greater susceptibility to infections and contributes to the inability to control pathogen growth and immune-mediated tissue damage. Since the impact of aging on immune function, is still to be fully elucidated, this review addresses some of the recent understanding of age-related changes affecting key components of immunity. The emphasis is on immunosenescence and inflammaging that are impacted by common infectious diseases that are characterized by a high mortality, and includes COVID-19, HIV and tuberculosis.
Collapse
Affiliation(s)
- Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
| | - Douglas McClain Noonan
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Alan L. Landay
- Department of Internal Medicine, Rush Medical College, Chicago, IL, United States
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| |
Collapse
|
106
|
Luo E, Zhong Q, Wen Y, Cai J, Xie X, Zhou L. Development and external validation of a prognostic tool for nonsevere COVID-19 inpatients. Epidemiol Infect 2023; 151:e128. [PMID: 37202367 PMCID: PMC10540186 DOI: 10.1017/s0950268823000717] [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: 09/15/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
To develop a machine learning model and nomogram to predict the probability of persistent virus shedding (PVS) in hospitalized patients with coronavirus disease 2019 (COVID-19), the clinical symptoms and signs, laboratory parameters, cytokines, and immune cell data of 429 patients with nonsevere COVID-19 were retrospectively reviewed. Two models were developed using the Akaike information criterion (AIC). The performance of these two models was analyzed and compared by the receiver operating characteristic (ROC) curve, calibration curve, net reclassification index (NRI), and integrated discrimination improvement (IDI). The final model included the following independent predictors of PVS: sex, C-reactive protein (CRP) level, interleukin-6 (IL-6) level, the neutrophil-lymphocyte ratio (NLR), monocyte count (MC), albumin (ALB) level, and serum potassium level. The model performed well in both the internal validation (corrected C-statistic = 0.748, corrected Brier score = 0.201) and external validation datasets (corrected C-statistic = 0.793, corrected Brier score = 0.190). The internal calibration was very good (corrected slope = 0.910). The model developed in this study showed high discriminant performance in predicting PVS in nonsevere COVID-19 patients. Because of the availability and accessibility of the model, the nomogram designed in this study could provide a useful prognostic tool for clinicians and medical decision-makers.
Collapse
Affiliation(s)
- Ensi Luo
- Department of Endocrinology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Qingyang Zhong
- Medical Department, The Ninth People’s Hospital of Dongguan, Dongguan, China
| | - Yongtao Wen
- Department of Respiratory Medicine, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Jie Cai
- Department of Respiratory Medicine, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Xia Xie
- Pain Department, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Lingjuan Zhou
- Nursing Department, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| |
Collapse
|
107
|
Nissen E, Reiner A, Liu S, Wallace RB, Molinaro AM, Salas LA, Christensen BC, Wiencke JK, Koestler DC, Kelsey KT. Assessment of immune cell profiles among post-menopausal women in the Women's Health Initiative using DNA methylation-based methods. Clin Epigenetics 2023; 15:69. [PMID: 37118842 PMCID: PMC10141818 DOI: 10.1186/s13148-023-01488-8] [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: 09/02/2022] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Over the past decade, DNA methylation (DNAm)-based deconvolution methods that leverage cell-specific DNAm markers of immune cell types have been developed to provide accurate estimates of the proportions of leukocytes in peripheral blood. Immune cell phenotyping using DNAm markers, termed immunomethylomics or methylation cytometry, offers a solution for determining the body's immune cell landscape that does not require fresh blood and is scalable to large sample sizes. Despite significant advances in DNAm-based deconvolution, references at the population level are needed for clinical and research interpretation of these additional immune layers. Here we aim to provide some references for immune populations in a group of multi-ethnic post-menopausal American women. RESULTS We applied DNAm-based deconvolution to a large sample of post-menopausal women enrolled in the Women's Health Initiative (baseline, N = 58) or the ancillary Long Life Study (WHI-LLS, N = 1237) to determine the reference ranges of 58 immune parameters, including proportions and absolute counts for 19 leukocyte subsets and 20 derived cell ratios. Participants were 50-94 years old at the time of blood draw, and N = 898 (69.3%) self-identified as White. Using linear regression models, we observed significant associations between age at blood draw and absolute counts and proportions of naïve B, memory CD4+, naïve CD4+, naïve CD8+, memory CD8+ memory, neutrophils, and natural killer cells. We also assessed the same immune profiles in a subset of paired longitudinal samples collected 14-18 years apart across N = 52 participants. Our results demonstrate high inter-individual variability in rates of change of leukocyte subsets over this time. And, when conducting paired t tests to test the difference in counts and proportions between the baseline visit and LLS visit, there were significant changes in naïve B, memory CD4+, naïve CD4+, naïve CD8+, memory CD8+ cells and neutrophils, similar to the results seen when analyzing the association with age in the entire cohort. CONCLUSIONS Here, we show that derived cell counts largely reflect the immune profile associated with proportions and that these novel methods replicate the known immune profiles associated with age. Further, we demonstrate the value this methylation cytometry approach can add as a potential application in epidemiological studies.
Collapse
Affiliation(s)
- Emily Nissen
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexander Reiner
- Division of Public Health Science, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simin Liu
- Departments of Epidemiology, Medicine, and Surgery, Brown University, Providence, RI, USA
| | - Robert B Wallace
- Departments of Epidemiology and Internal Medicine, School of Public Health, University of Iowa, Iowa City, IA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Devin C Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
108
|
Fang Y, Doyle MF, Chen J, Mez J, Satizabal CL, Alosco ML, Qiu WQ, Lunetta KL, Murabito JM. Circulating immune cell phenotypes are associated with age, sex, CMV, and smoking status in the Framingham Heart Study offspring participants. Aging (Albany NY) 2023; 15:3939-3966. [PMID: 37116193 PMCID: PMC10258017 DOI: 10.18632/aging.204686] [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: 01/20/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
Understanding the composition of circulating immune cells with aging and the underlying biologic mechanisms driving aging may provide molecular targets to slow the aging process and reduce age-related disease. Utilizing cryopreserved cells from 996 Framingham Heart Study (FHS) Offspring Cohort participants aged 40 and older (mean 62 years, 48% female), we report on 116 immune cell phenotypes including monocytes, T-, B-, and NK cells and their subtypes, across age groups, sex, cytomegalovirus (CMV) exposure groups, smoking and other cardiovascular risk factors. The major cellular differences with CMV exposure were higher Granzyme B+ cells, effector cells, and effector-memory re-expressing CD45RA (TEMRA) cells for both CD4+ and CD8+. Older age was associated with lower CD3+ T cells, lower naïve cells and naïve/memory ratios for CD4+ and CD8+. We identified many immune cell differences by sex, with males showing lower naïve cells and higher effector and effector memory cells. Current smokers showed lower pro-inflammatory CD8 cells, higher CD8 regulatory type cells and altered B cell subsets. No significant associations were seen with BMI and other cardiovascular risk factors. Our cross-sectional observations of immune cell phenotypes provide a reference to further the understanding of the complexity of immune cells in blood, an easily accessible tissue.
Collapse
Affiliation(s)
- Yuan Fang
- Boston University School of Public Health, Department of Biostatistics, Boston, MA 02118, USA
| | - Margaret F. Doyle
- University of Vermont, Larner College of Medicine, Department of Pathology and Laboratory Medicine, Burlington, VT 05405, USA
| | - Jiachen Chen
- Boston University School of Public Health, Department of Biostatistics, Boston, MA 02118, USA
| | - Jesse Mez
- Boston University Chobanian and Avedisian School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA 02118, USA
- Boston University Chobanian and Avedisian School of Medicine, Department of Neurology, Boston, MA 02118, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University Chobanian and Avedisian School of Medicine, Framingham, MA 01702, USA
| | - Claudia L. Satizabal
- Boston University Chobanian and Avedisian School of Medicine, Department of Neurology, Boston, MA 02118, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University Chobanian and Avedisian School of Medicine, Framingham, MA 01702, USA
- University of Texas Health Science Center at San Antonio, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX 78229, USA
| | - Michael L. Alosco
- Boston University Chobanian and Avedisian School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA 02118, USA
- Boston University Chobanian and Avedisian School of Medicine, Department of Neurology, Boston, MA 02118, USA
| | - Wei Qiao Qiu
- Boston University Chobanian and Avedisian School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA 02118, USA
- Boston University Chobanian and Avedisian School of Medicine, Department of Psychiatry, Boston, MA 02118, USA
- Boston University Chobanian and Avedisian School of Medicine, Department of Pharmacology and Experimental Therapeutics, Boston, MA 02118, USA
| | - Kathryn L. Lunetta
- Boston University School of Public Health, Department of Biostatistics, Boston, MA 02118, USA
| | - Joanne M. Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University Chobanian and Avedisian School of Medicine, Framingham, MA 01702, USA
- Boston University Chobanian and Avedisian School of Medicine, Department of Medicine, Boston, MA 02118, USA
- Boston Medical Center, Department of Adult Primary Care, Boston, MA 02119, USA
| |
Collapse
|
109
|
Wong MCS, Zhang L, Ching JYL, Mak JWY, Huang J, Wang S, Mok CKP, Wong A, Chiu OL, Fung YT, Cheong PK, Tun HM, Ng SC, Chan FKL. Effects of Gut Microbiome Modulation on Reducing Adverse Health Outcomes among Elderly and Diabetes Patients during the COVID-19 Pandemic: A Randomised, Double-Blind, Placebo-Controlled Trial (IMPACT Study). Nutrients 2023; 15:1982. [PMID: 37111201 PMCID: PMC10143994 DOI: 10.3390/nu15081982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Gut microbiota is believed to be a major determinant of health outcomes. We hypothesised that a novel oral microbiome formula (SIM01) can reduce the risk of adverse health outcomes in at-risk subjects during the coronavirus disease 2019 (COVID-19) pandemic. In this single-centre, double-blind, randomised, placebo-controlled trial, we recruited subjects aged ≥65 years or with type two diabetes mellitus. Eligible subjects were randomised in a 1:1 ratio to receive three months of SIM01 or placebo (vitamin C) within one week of the first COVID-19 vaccine dose. Both the researchers and participants were blinded to the groups allocated. The rate of adverse health outcomes was significantly lower in the SIM01 group than the placebo at one month (6 [2.9%] vs. 25 [12.6], p < 0.001) and three months (0 vs. 5 [3.1%], p = 0.025). At three months, more subjects who received SIM01 than the placebo reported better sleep quality (53 [41.4%] vs. 22 [19.3%], p < 0.001), improved skin condition (18 [14.1%] vs. 8 [7.0%], p = 0.043), and better mood (27 [21.2%] vs. 13 [11.4%], p = 0.043). Subjects who received SIM01 showed a significant increase in beneficial Bifidobacteria and butyrate-producing bacteria in faecal samples and strengthened the microbial ecology network. SIM01 reduced adverse health outcomes and restored gut dysbiosis in elderly and diabetes patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Martin C. S. Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica Y. L. Ching
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joyce W. Y. Mak
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shilan Wang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chris K. P. Mok
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Angie Wong
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Oi-Lee Chiu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yee-Ting Fung
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pui-Kuan Cheong
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hein-Min Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
110
|
McGill CJ, Ewald CY, Benayoun BA. Sex-dimorphic expression of extracellular matrix genes in mouse bone marrow neutrophils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530027. [PMID: 36909511 PMCID: PMC10002647 DOI: 10.1101/2023.02.25.530027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The mammalian innate immune system is sex-dimorphic. Neutrophils are the most abundant leukocyte in humans and represent innate immunity's first line of defense. We previously found that primary mouse bone marrow neutrophils show widespread sex-dimorphism throughout life, including at the transcriptional level. Extracellular matrix [ECM]-related terms were observed among the top sex-dimorphic genes. Since the ECM is emerging as an important regulator of innate immune responses, we sought to further investigate the transcriptomic profile of primary mouse bone marrow neutrophils at both the bulk and single-cell level to understand how biological sex may influence ECM component expression in neutrophils throughout life. Here, using curated gene lists of ECM components and unbiased weighted gene co-expression network analysis [WGCNA], we find that multiple ECM-related gene sets show widespread female-bias in expression in primary mouse neutrophils. Since many immune-related diseases (e.g., rheumatoid arthritis) are more prevalent in females, our work may provide insights into the pathogenesis of sex-dimorphic inflammatory diseases.
Collapse
Affiliation(s)
- Cassandra J McGill
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Schwerzenbach, Switzerland
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA 90089, USA
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA
- USC Stem Cell Initiative, Los Angeles, CA 90089, USA
| |
Collapse
|
111
|
Lott N, Gebhard CE, Bengs S, Haider A, Kuster GM, Regitz-Zagrosek V, Gebhard C. Sex hormones in SARS-CoV-2 susceptibility: key players or confounders? Nat Rev Endocrinol 2023; 19:217-231. [PMID: 36494595 PMCID: PMC9734735 DOI: 10.1038/s41574-022-00780-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has a clear sex disparity in clinical outcomes. Hence, the interaction between sex hormones, virus entry receptors and immune responses has attracted major interest as a target for the prevention and treatment of SARS-CoV-2 infections. This Review summarizes the current understanding of the roles of androgens, oestrogens and progesterone in the regulation of virus entry receptors and disease progression of coronavirus disease 2019 (COVID-19) as well as their therapeutic value. Although many experimental and clinical studies have analysed potential mechanisms by which female sex hormones might provide protection against SARS-CoV-2 infectivity, there is currently no clear evidence for a sex-specific expression of virus entry receptors. In addition, reports describing an influence of oestrogen, progesterone and androgens on the course of COVID-19 vary widely. Current data also do not support the administration of oestradiol in COVID-19. The conflicting evidence and lack of consensus results from a paucity of mechanistic studies and clinical trials reporting sex-disaggregated data. Further, the influence of variables beyond biological factors (sex), such as sociocultural factors (gender), on COVID-19 manifestations has not been investigated. Future research will have to fill this knowledge gap as the influence of sex and gender on COVID-19 will be essential to understanding and managing the long-term consequences of this pandemic.
Collapse
Affiliation(s)
- Nicola Lott
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Gabriela M Kuster
- Department of Cardiology and Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Cardiology, Inselspital Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
112
|
Karakaslar EO, Katiyar N, Hasham M, Youn A, Sharma S, Chung C, Marches R, Korstanje R, Banchereau J, Ucar D. Transcriptional activation of Jun and Fos members of the AP-1 complex is a conserved signature of immune aging that contributes to inflammaging. Aging Cell 2023; 22:e13792. [PMID: 36840360 PMCID: PMC10086525 DOI: 10.1111/acel.13792] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 02/26/2023] Open
Abstract
Diverse mouse strains have different health and life spans, mimicking the diversity among humans. To capture conserved aging signatures, we studied long-lived C57BL/6J and short-lived NZO/HILtJ mouse strains by profiling transcriptomes and epigenomes of immune cells from peripheral blood and the spleen from young and old mice. Transcriptional activation of the AP-1 transcription factor complex, particularly Fos, Junb, and Jun genes, was the most significant and conserved aging signature across tissues and strains. ATAC-seq data analyses showed that the chromatin around these genes was more accessible with age and there were significantly more binding sites for these TFs with age across all studied tissues, targeting pro-inflammatory molecules including Il6. Age-related increases in binding sites of JUN and FOS factors were also conserved in human peripheral blood ATAC-seq data. Single-cell RNA-seq data from the mouse aging cell atlas Tabula Muris Senis showed that the expression of these genes increased with age in B, T, NK cells, and macrophages, with macrophages from old mice expressing these molecules more abundantly than other cells. Functional data showed that upon myeloid cell activation via poly(I:C), the levels of JUN protein and its binding activity increased more significantly in spleen cells from old compared to young mice. In addition, upon activation, old cells produced more IL6 compared to young cells. In sum, we showed that the aging-related transcriptional activation of Jun and Fos family members in AP-1 complex is conserved across immune tissues and long- and short-living mouse strains, possibly contributing to increased inflammation with age.
Collapse
Affiliation(s)
- Emin Onur Karakaslar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
- Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Neerja Katiyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Muneer Hasham
- The Jackson Laboratory for Mammalian GeneticsBar HarborMaineUSA
| | | | | | - Cheng‐han Chung
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Radu Marches
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Ron Korstanje
- The Jackson Laboratory for Mammalian GeneticsBar HarborMaineUSA
| | - Jacques Banchereau
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
- ImmunaiNew YorkNew YorkUSA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
- Department of Genetics and Genome SciencesUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
| |
Collapse
|
113
|
Baraibar I, Ros J, Saoudi N, Salvà F, García A, Castells M, Tabernero J, Élez E. Sex and gender perspectives in colorectal cancer. ESMO Open 2023; 8:101204. [PMID: 37018873 PMCID: PMC10163160 DOI: 10.1016/j.esmoop.2023.101204] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 04/05/2023] Open
Abstract
Historically women were frequently excluded from clinical trials and drug usage to protect unborn babies from potential harm. As a consequence, the impact of sex and gender on both tumour biology and clinical outcomes has been largely underestimated. Although interrelated and often used interchangeably, sex and gender are not equivalent concepts. Sex is a biological attribute that defines species according to their chromosomal makeup and reproductive organ, while gender refers to a chosen sexual identity. Sex dimorphisms are rarely taken into account, in either preclinical or clinical research, with inadequate analysis of differences in outcomes according to sex or gender still widespread, reflecting a gap in our knowledge for a large proportion of the target population. Underestimation of sex-based differences in study design and analyses has invariably led to 'one-drug' treatment regimens for both males and females. For patients with colorectal cancer (CRC), sex also has an impact on the disease incidence, clinicopathological features, therapeutic outcomes, and tolerability to anticancer treatments. Although the global incidence of CRC is higher in male subjects, the proportion of patients presenting right-sided tumours and BRAF mutations is higher among females. Concerning sex-related differences in treatment efficacy and toxicity, drug dosage does not take into account sex-specific differences in pharmacokinetics. Toxicity associated with fluoropyrimidines, targeted therapies, and immunotherapies has been reported to be more extensive for females with CRC than for males, although evidence about differences in efficacy is more controversial. This article aims to provide an overview of the research achieved so far into sex and gender differences in cancer and summarize the growing body of literature illustrating the sex and gender perspective in CRC and their impact in relation to tumour biology and treatment efficacy and toxicity. We propose endorsing research on how biological sex and gender influence CRC as an added value for precision oncology.
Collapse
|
114
|
Miller RAJ, Williams AP, Kovats S. Sex chromosome complement and sex steroid signaling underlie sex differences in immunity to respiratory virus infection. Front Pharmacol 2023; 14:1150282. [PMID: 37063266 PMCID: PMC10097973 DOI: 10.3389/fphar.2023.1150282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/08/2023] [Indexed: 04/18/2023] Open
Abstract
Epidemiological studies have revealed sex differences in the incidence and morbidity of respiratory virus infection in the human population, and often these observations are correlated with sex differences in the quality or magnitude of the immune response. Sex differences in immunity and morbidity also are observed in animal models of respiratory virus infection, suggesting differential dominance of specific immune mechanisms. Emerging research shows intrinsic sex differences in immune cell transcriptomes, epigenomes, and proteomes that may regulate human immunity when challenged by viral infection. Here, we highlight recent research into the role(s) of sex steroids and X chromosome complement in immune cells and describe how these findings provide insight into immunity during respiratory virus infection. We focus on the regulation of innate and adaptive immune cells by receptors for androgen and estrogens, as well as genes with a propensity to escape X chromosome inactivation. A deeper mechanistic knowledge of these pathways will help us to understand the often significant sex differences in immunity to endemic or pandemic respiratory pathogens such as influenza viruses, respiratory syncytial viruses and pathogenic coronaviruses.
Collapse
Affiliation(s)
- Reegan A. J. Miller
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Abigael P. Williams
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
115
|
Li X, Wei S, Deng L, Tao H, Liu M, Zhao Z, Du X, Li Y, Hou J. Sex-biased molecular differences in lung adenocarcinoma are ethnic and smoking specific. BMC Pulm Med 2023; 23:99. [PMID: 36964522 PMCID: PMC10039609 DOI: 10.1186/s12890-023-02387-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/14/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND Sex-related differences in cancer epidemiology, tumor biology, immune system activity, and pharmacogenomics have been suggested to be important considerations for precision cancer control. Here we elucidated systematically sex biases in genetic variants, gene expression profiles, and immunological landscapes of lung adenocarcinoma patients (LUADs) with different ancestry and smoking status. METHODS Somatic mutation and mRNA expression data of Asian and Non-Asian LUADs were obtained from public databases. Sex-biased genetic mutations, gene expression, biological pathways, and immune infiltration were identified in the context of smoking status and race. RESULTS Among nonsmokers, male-biased mutations were prevalent in Asian LUADs, while few sex-biased mutations were detected in Non-Asian LUADs. EGFR was the only mutation whose frequency was significantly higher in females than males in both Asian and Non-Asian nonsmokers. More genes exhibited sex-biased expression in Non-Asian LUADs compared to Asian LUADs. Moreover, genes distinctly expressed in females were mainly related to immune-related pathways, whereas those in males were more involved in activation of DNA repair, E2F_targets, and MYC_targets pathways. We also detected sex-specific immune infiltration in the context of genetic variation. In EGFR-mutant LUADs, males had a significantly increased infiltration of CD8 + T cells, whereas resting CD4 + memory T cells were more abundant in females. Additionally, in KRAS-mutant LUADs, CD8 + and CD4 + T cells were more abundant in females than males. In addition, we detected all female patients with high SCGB3A2 expression were exclusively sensitive to immunotherapy, while this phenomenon was not observed in male patients. CONCLUSIONS Our findings provided evidence that sex-related molecular and cellular components are involved in shaping tumor distinct genetic and immune features, which might have important impact on personalized targeted and immune therapy.
Collapse
Affiliation(s)
- Xuetao Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Shuquan Wei
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Liaoyuan Deng
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - HongYan Tao
- Department of Pulmonary Diseases, The Second Affiliated Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Mingkai Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Ziwen Zhao
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Yujun Li
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| | - Jun Hou
- Center for Medical Research On Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| |
Collapse
|
116
|
Broustas CG, Shuryak I, Duval AJ, Amundson SA. Effect of Age and Sex on Gene Expression-Based Radiation Biodosimetry Using Mouse Peripheral Blood. Cytogenet Genome Res 2023; 163:197-209. [PMID: 36928338 PMCID: PMC10585707 DOI: 10.1159/000530172] [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: 12/21/2022] [Accepted: 02/03/2023] [Indexed: 03/18/2023] Open
Abstract
Blood-based gene expression profiles that can reconstruct radiation exposure are being developed as a practical approach to radiation biodosimetry. However, age and sex could potentially limit the accuracy of the approach. In this study, we determined the impact of age on the peripheral blood cell gene expression profile of female mice exposed to radiation and identified differences and similarities with a previously obtained transcriptomic signature of male mice. Young (2 months) and old (24 months) female mice were irradiated with 4 Gy X-rays, total RNA was isolated from blood 24 hours later and subjected to whole-genome microarray analysis. Dose reconstruction analyses using a gene signature trained on gene expression data from irradiated young male mice showed accurate reconstruction of 0 or 4 Gy doses with root mean square error of ±0.75 Gy (R2 = 0.90) in young female mice. Although dose reconstruction for irradiated old female mice was less accurate than young female mice, the deviation from the actual radiation dose was not statistically significant. Pathway analysis of differentially expressed genes revealed that after irradiation, apoptosis-related functions were overrepresented, whereas functions related to quantities of various immune cell subtypes were underrepresented, among differentially expressed genes from young female mice, but not older animals. Furthermore, young mice significantly upregulated genes involved in phagocytosis, a process that eliminates apoptotic cells and preserves tissue homeostasis. Both functions were also overrepresented in young, but not old, male mice following 4 Gy X-irradiation. Lastly, functions associated with neutrophil activation that is essential for killing invading pathogens and regulating the inflammatory response were predicted to be uniquely enriched in young but not old female mice. This work supports the concept that peripheral blood gene expression profiles can be identified in mice that accurately predict physical radiation dose exposure irrespective of age and sex.
Collapse
Affiliation(s)
- Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Axel J. Duval
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Sally A. Amundson
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
117
|
Molecular Markers of Blood Cell Populations Can Help Estimate Aging of the Immune System. Int J Mol Sci 2023; 24:ijms24065708. [PMID: 36982782 PMCID: PMC10055688 DOI: 10.3390/ijms24065708] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Aging of the immune system involves functional changes in individual cell populations, in hematopoietic tissues and at the systemic level. They are mediated by factors produced by circulating cells, niche cells, and at the systemic level. Age-related alterations in the microenvironment of the bone marrow and thymus cause a decrease in the production of naive immune cells and functional immunodeficiencies. Another result of aging and reduced tissue immune surveillance is the accumulation of senescent cells. Some viral infections deplete adaptive immune cells, increasing the risk of autoimmune and immunodeficiency conditions, leading to a general degradation in the specificity and effectiveness of the immune system in old age. During the COVID-19 pandemic, the state-of-the-art application of mass spectrometry, multichannel flow cytometry, and single-cell genetic analysis have provided vast data on the mechanisms of aging of the immune system. These data require systematic analysis and functional verification. In addition, the prediction of age-related complications is a priority task of modern medicine in the context of the increase in the aged population and the risk of premature death during epidemics. In this review, based on the latest data, we discuss the mechanisms of immune aging and highlight some cellular markers as indicators of age-related immune disbalance that increase the risk of senile diseases and infectious complications.
Collapse
|
118
|
Suarez LM, Diaz-Del Cerro E, Felix J, Gonzalez-Sanchez M, Ceprian N, Guerra-Perez N, G Novelle M, Martinez de Toda I, De la Fuente M. Sex differences in neuroimmunoendocrine communication. Involvement on longevity. Mech Ageing Dev 2023; 211:111798. [PMID: 36907251 DOI: 10.1016/j.mad.2023.111798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Endocrine, nervous, and immune systems work coordinately to maintain the global homeostasis of the organism. They show sex differences in their functions that, in turn, contribute to sex differences beyond reproductive function. Females display a better control of the energetic metabolism and improved neuroprotection and have more antioxidant defenses and a better inflammatory status than males, which is associated with a more robust immune response than that of males. These differences are present from the early stages of life, being more relevant in adulthood and influencing the aging trajectory in each sex and may contribute to the different life lifespan between sexes.
Collapse
Affiliation(s)
- Luz M Suarez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain.
| | - Estefania Diaz-Del Cerro
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Judith Felix
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica Gonzalez-Sanchez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Noemi Ceprian
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Natalia Guerra-Perez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Marta G Novelle
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain
| | - Irene Martinez de Toda
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica De la Fuente
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain.
| |
Collapse
|
119
|
Maegele M, Aletti F, Efron PA, Relja B, Orfanos SE. NEW INSIGHTS INTO THE PATHOPHYSIOLOGY OF TRAUMA AND HEMORRHAGE. Shock 2023; 59:6-9. [PMID: 36867756 DOI: 10.1097/shk.0000000000001954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
ABSTRACT Circulatory shock from trauma and hemorrhage remains a clinical challenge with mortality still high within the first hours after impact. It represents a complex disease involving the impairment of a number of physiological systems and organs and the interaction of different pathological mechanisms. Multiple external and patient-specific factors may further modulate and complicate the clinical course. Recently, novel targets and models with complex multiscale interaction of data from different sources have been identified which offer new windows of opportunity. Future works needs to consider patient-specific conditions and outcomes to mount shock research onto the next higher level of precision and personalized medicine.
Collapse
Affiliation(s)
- Marc Maegele
- Department of Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center, Institute for Research in Operative Medicine, University Witten-Herdecke, Cologne, Germany
| | - Federico Aletti
- Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, São José dos Campos, Brazil
| | - Philip A Efron
- Department of Surgery, Division of Acute Care Surgery and Surgical Critical Care, Laboratory of Inflammation Biology and Surgical Science, UF Health Critical Care Organization, Florida
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Stylianos E Orfanos
- 1st Department of Critical Care Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| |
Collapse
|
120
|
Martínez de Toda I, González-Sánchez M, Díaz-Del Cerro E, Valera G, Carracedo J, Guerra-Pérez N. Sex differences in markers of oxidation and inflammation. Implications for ageing. Mech Ageing Dev 2023; 211:111797. [PMID: 36868323 DOI: 10.1016/j.mad.2023.111797] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Sexual dimorphism is a key factor to consider in the ageing process given the impact that it has on life expectancy. The oxidative-inflammatory theory of ageing states that the ageing process is the result of the establishment of oxidative stress which, due to the interplay of the immune system, translates into inflammatory stress, and that both processes are responsible for the damage and loss of function of an organism. We show that there are relevant gender differences in a number of oxidative and inflammatory markers and propose that they may account for the differential lifespan between sexes, given that males display, in general, higher oxidation and basal inflammation. In addition, we explain the significant role of circulating cell-free DNA as a marker of oxidative damage and an inductor of inflammation, connecting both processes and having the potential to become a useful ageing marker. Finally, we discuss how oxidative and inflammatory changes take place differentially with ageing in each sex, which could also have an impact on the sex-differential lifespan. Further research including sex as an essential variable is needed to understand the grounds of sex differences in ageing and to better comprehend ageing itself.
Collapse
Affiliation(s)
- Irene Martínez de Toda
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | - Mónica González-Sánchez
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Estefanía Díaz-Del Cerro
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | - Gemma Valera
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | - Julia Carracedo
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | - Natalia Guerra-Pérez
- Department of Genetics, Physiology, and Microbiology. Unit of Animal Physiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| |
Collapse
|
121
|
Olivieri F, Marchegiani F, Matacchione G, Giuliani A, Ramini D, Fazioli F, Sabbatinelli J, Bonafè M. Sex/gender-related differences in inflammaging. Mech Ageing Dev 2023; 211:111792. [PMID: 36806605 DOI: 10.1016/j.mad.2023.111792] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Geroscience puts mechanisms of aging as a driver of the most common age-related diseases and dysfunctions. Under this perspective, addressing the basic mechanisms of aging will produce a better understanding than addressing each disease pathophysiology individually. Worldwide, despite greater functional impairment, life expectancy is higher in women than in men. Gender differences in the prevalence of multimorbidity lead mandatory to the understanding of the mechanisms underlying gender-related differences in multimorbidity patterns and disability-free life expectancy. Extensive literature suggested that inflammaging is at the crossroad of aging and age-related diseases. In this review, we highlight the main evidence on sex/gender differences in the mechanisms that foster inflammaging, i.e. the age-dependent triggering of innate immunity, modifications of adaptive immunity, and accrual of senescent cells, underpinning some biomarkers of inflammaging that show sex-related differences. In the framework of the "gender medicine perspective", we will also discuss how sex/gender differences in inflammaging can affect sex differences in COVID-19 severe outcomes.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | | | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Deborah Ramini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Francesca Fazioli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Laboratory Medicine Unit, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy.
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
122
|
Duijster JW, Lieber T, Pacelli S, Van Balveren L, Ruijs LS, Raethke M, Kant A, Van Hunsel F. Sex-disaggregated outcomes of adverse events after COVID-19 vaccination: A Dutch cohort study and review of the literature. Front Immunol 2023; 14:1078736. [PMID: 36793715 PMCID: PMC9922710 DOI: 10.3389/fimmu.2023.1078736] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
Background Albeit the need for sex-disaggregated results of adverse events after immunization (AEFIs) is gaining attention since the COVID-19 pandemic, studies with emphasis on sexual dimorphism in response to COVID-19 vaccination are relatively scarce. This prospective cohort study aimed to assess differences in the incidence and course of reported AEFIs after COVID-19 vaccination between males and females in the Netherlands and provides a summary of sex-disaggregated outcomes in published literature. Methods Patient reported outcomes of AEFIs over a six month period following the first vaccination with BioNTech-Pfizer, AstraZeneca, Moderna or the Johnson&Johnson vaccine were collected in a Cohort Event Monitoring study. Logistic regression was used to assess differences in incidence of 'any AEFI', local reactions and the top ten most reported AEFIs between the sexes. Effects of age, vaccine brand, comorbidities, prior COVID-19 infection and the use of antipyretic drugs were analyzed as well. Also, time-to-onset, time-to-recovery and perceived burden of AEFIs was compared between the sexes. Third, a literature review was done to retrieve sex-disaggregated outcomes of COVID-19 vaccination. Results The cohort included 27,540 vaccinees (38.5% males). Females showed around two-fold higher odds of having any AEFI as compared to males with most pronounced differences after the first dose and for nausea and injection site inflammation. Age was inversely associated with AEFI incidence, whereas a prior COVID-19 infection, the use of antipyretic drugs and several comorbidities were positively associated. The perceived burden of AEFIs and time-to-recovery were slightly higher in females. Discussion The results of this large cohort study correspond to existing evidence and contribute to the knowledge gain necessary to disentangle the magnitude of the effect sex in response to vaccination. Whilst females have a significant higher probability of experiencing an AEFI than males, we observed that the course and burden is only to a minor extent different between the sexes.
Collapse
Affiliation(s)
| | - Thomas Lieber
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch, Netherlands
| | - Silvia Pacelli
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch, Netherlands
- School of Pharmacy, Biotechnology, and Sport Sciences, University of Bologna, Bologna, Italy
| | | | - Loes S. Ruijs
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch, Netherlands
| | - Monika Raethke
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch, Netherlands
| | - Agnes Kant
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch, Netherlands
| | | |
Collapse
|
123
|
Karal-ogly DD, Shumeev AN, Keburiya VV, Mintel MV, Rybtsov SA. Age-Related Changes in the Clustering of Blood Populations in Cynomolgus Monkeys Depend on Sex and Immune Status. Life (Basel) 2023; 13:life13020316. [PMID: 36836673 PMCID: PMC9965083 DOI: 10.3390/life13020316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Non-anthropoid primates cynomolgus monkeys (Macaca fascicularis), also known as crab-eating macaques, are increasingly used in biomedical and preclinical studies due to their evolutionary proximity to humans, sharing similar diets, infectious and senile diseases. Age-related changes and sexual dimorphism of the immune system of C. monkeys have not been sufficiently characterized in literature, though age and sex differences affect the course of diseases and sensitivity to medications. Aging in C. monkeys is accompanied by an increase in CD3+CD4+CD8+ (DP-T) cells, plasma B-cells, and a decrease in platelets. Erythromyeloid bias has also been noticed in older animals. There was an increase in eosinophils, haematocrit (HCT) and haemoglobin concentration (HGB). Senile decline in the function of the immune system had sex differences. An increase in the number of monocytes, cytotoxic lymphocytes (CTL) and a decrease in the T-helper population were more pronounced in older females. A significant reduction in the number of B-cells and activated T-cells was detected in males only. A moderate correlation with the regression model of aging was established for DP-T, HCT and HGB. The reduction in the B cells count in males and the increase in CTL level in females are moderately correlated with age. Other blood cell populations did not show significant correlations in the regression models due to their high sample variability. The novel cell population CD3-CD20loCD16/CD56+, presumably NK-cells subset, was revealed. This cell population demonstrated an increase trend with age in both sexes. Population-statistical age norms for different sexes for young and very old macaques were established. The blood population clusters associated with sex and immune status in older animals were also identified.
Collapse
Affiliation(s)
| | - Alexander N. Shumeev
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | | | - Marina V. Mintel
- The Research Institute of Medical Primatology, 354383 Sochi, Russia
| | - Stanislav A. Rybtsov
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK
- Correspondence:
| |
Collapse
|
124
|
García-Pérez R, Ramirez JM, Ripoll-Cladellas A, Chazarra-Gil R, Oliveros W, Soldatkina O, Bosio M, Rognon PJ, Capella-Gutierrez S, Calvo M, Reverter F, Guigó R, Aguet F, Ferreira PG, Ardlie KG, Melé M. The landscape of expression and alternative splicing variation across human traits. CELL GENOMICS 2023; 3:100244. [PMID: 36777183 PMCID: PMC9903719 DOI: 10.1016/j.xgen.2022.100244] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
Understanding the consequences of individual transcriptome variation is fundamental to deciphering human biology and disease. We implement a statistical framework to quantify the contributions of 21 individual traits as drivers of gene expression and alternative splicing variation across 46 human tissues and 781 individuals from the Genotype-Tissue Expression project. We demonstrate that ancestry, sex, age, and BMI make additive and tissue-specific contributions to expression variability, whereas interactions are rare. Variation in splicing is dominated by ancestry and is under genetic control in most tissues, with ribosomal proteins showing a strong enrichment of tissue-shared splicing events. Our analyses reveal a systemic contribution of types 1 and 2 diabetes to tissue transcriptome variation with the strongest signal in the nerve, where histopathology image analysis identifies novel genes related to diabetic neuropathy. Our multi-tissue and multi-trait approach provides an extensive characterization of the main drivers of human transcriptome variation in health and disease.
Collapse
Affiliation(s)
- Raquel García-Pérez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Jose Miguel Ramirez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Aida Ripoll-Cladellas
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Ruben Chazarra-Gil
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Winona Oliveros
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Oleksandra Soldatkina
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Mattia Bosio
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Paul Joris Rognon
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
- Department of Economics and Business, Universitat Pompeu Fabra, Barcelona, Catalonia 08005, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia 08034, Spain
| | - Salvador Capella-Gutierrez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Miquel Calvo
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Ferran Reverter
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Roderic Guigó
- Bioinformatics and Genomics, Center for Genomic Regulation, Barcelona, Catalonia 08003, Spain
| | | | - Pedro G. Ferreira
- Department of Computer Science, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
- Laboratory of Artificial Intelligence and Decision Support, INESC TEC, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto, Institute for Research and Innovation in Health (i3s), R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | | | - Marta Melé
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| |
Collapse
|
125
|
Yiang GT, Wu CC, Lu CL, Hu WC, Tsai YJ, Huang YM, Su WL, Lu KC. Endoplasmic Reticulum Stress in Elderly Patients with COVID-19: Potential of Melatonin Treatment. Viruses 2023; 15:156. [PMID: 36680196 PMCID: PMC9863214 DOI: 10.3390/v15010156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aging processes, including immunosenescence, inflammation, inflammasome formation, genomic instability, telomeric attrition, and altered autophagy, are involved in viral infections and they may contribute to increased pathophysiological responses to the SARS-CoV-2 infection in the elderly; this poses additional risks of accelerated aging, which could be found even after recovery. Aging is associated with oxidative damage. Moreover, SARS-CoV-2 infections may increase the production of reactive oxygen species and such infections will disturb the Ca++ balance via an endoplasmic reticulum (ER) stress-mediated unfolded protein response. Although vaccine development and anti-inflammation therapy lower the severity of COVID-19, the prevalence and mortality rates are still alarming in some countries worldwide. In this review, we describe the involvement of viral proteins in activating ER stress transducers and their downstream signals and in inducing inflammation and inflammasome formation. Furthermore, we propose the potential of melatonin as an ER stress modulator, owing to its antioxidant, anti-inflammatory, and immunoregulatory effects in viral infections. Considering its strong safety profile, we suggest that additive melatonin supplementation in the elderly could be beneficial in treating COVID-19.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei 243, Taiwan
| | - Yiao-Mien Huang
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Wen-Lin Su
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| |
Collapse
|
126
|
Caruso C, Accardi G, Aiello A, Calabrò A, Ligotti ME, Candore G. Centenarians born before 1919 are resistant to COVID-19. Aging Clin Exp Res 2023; 35:217-220. [PMID: 36319938 PMCID: PMC9628492 DOI: 10.1007/s40520-022-02287-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022]
Abstract
Although mortality from COVID-19 progressively increases with age, there are controversial data in the literature on the probability of centenarians dying from COVID-19. Moreover, it has been claimed that men in their 90s and 100s are more resilient than women. To gain insight into this matter, we analysed, according to gender, mortality data during the first year of pandemic of Sicilian nonagenarians and centenarians. We used mortality data from the 2019 as a control. The crude excess mortality between the two years was calculated. Data on deaths of Sicilian 90 + years show that, in line with what is known about the different response to infections between the two genders, oldest females are more resilient to COVID-19 than males. Moreover, centenarians born before 1919, but not "younger centenarians", are resilient to COVID-19. This latter datum should be related to the 1918 Spanish flu epidemic, although the mechanisms involved are not clear.
Collapse
Affiliation(s)
- Calogero Caruso
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Giulia Accardi
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Anna Aiello
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Anna Calabrò
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Mattia Emanuela Ligotti
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| | - Giuseppina Candore
- grid.10776.370000 0004 1762 5517Laboratorio di Immunopatologia e Immunosenescenza, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Palermo, Italy
| |
Collapse
|
127
|
Yang Q, Kennicott K, Zhu R, Kim J, Wakefield H, Studener K, Liang Y. Sex hormone influence on female-biased autoimmune diseases hints at puberty as an important factor in pathogenesis. Front Pediatr 2023; 11:1051624. [PMID: 36793337 PMCID: PMC9923181 DOI: 10.3389/fped.2023.1051624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
The majority of autoimmune diseases affect more women than men, suggesting an important role for sex hormones in regulating immune response. Current research supports this idea, highlighting the importance of sex hormones in both immune and metabolic regulation. Puberty is characterized by drastic changes in sex hormone levels and metabolism. These pubertal changes may be what forms the gulf between men and women in sex bias towards autoimmunity. In this review, a current perspective on pubertal immunometabolic changes and their impact on the pathogenesis of a select group of autoimmune diseases is presented. SLE, RA, JIA, SS, and ATD were focused on in this review for their notable sex bias and prevalence. Due to both the scarcity of pubertal autoimmune data and the differences in mechanism or age-of-onset in juvenile analogues often beginning prior to pubertal changes, data on the connection between the specific adult autoimmune diseases and puberty often relies on sex hormone influence in pathogenesis and established sex differences in immunity that begin during puberty.
Collapse
Affiliation(s)
- Qianfan Yang
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Kameron Kennicott
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Runqi Zhu
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jooyong Kim
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Hunter Wakefield
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Katelyn Studener
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Yun Liang
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Correspondence: Yun Liang
| |
Collapse
|
128
|
Shapiro JR, Roberts CW, Arcovio K, Reade L, Klein SL, Dhakal S. Effects of Biological Sex and Pregnancy on SARS-CoV-2 Pathogenesis and Vaccine Outcomes. Curr Top Microbiol Immunol 2023; 441:75-110. [PMID: 37695426 DOI: 10.1007/978-3-031-35139-6_4] [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: 09/12/2023]
Abstract
SARS-CoV-2 is the causative agent of COVID-19 in humans and has resulted in the death of millions of people worldwide. Similar numbers of infections have been documented in males and females; males, however, are more likely than females to be hospitalized, require intensive care unit, or die from COVID-19. The mechanisms that account for this are multi-factorial and are likely to include differential expression of ACE2 and TMPRSS2 molecules that are required for viral entry into hosts cells and sex differences in the immune response, which are due to modulation of cellular functions by sex hormones and differences in chromosomal gene expression. Furthermore, as comorbidities are also associated with poorer outcomes to SARS-CoV-2 infection and several comorbidities are overrepresented in males, these are also likely to contribute to the observed sex differences. Despite their relative better prognosis following infection with SARS-CoV-2, females do have poorer outcomes during pregnancy. This is likely to be due to pregnancy-induced changes in the immune system that adversely affect viral immunity and disruption of the renin-angiotensin system. Importantly, vaccination reduces the severity of disease in males and females, including pregnant females, and there is no evidence that vaccination has any adverse effects on the outcomes of pregnancy.
Collapse
Affiliation(s)
- Janna R Shapiro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Craig W Roberts
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Kasandra Arcovio
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Lisa Reade
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Sabra L Klein
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
129
|
N’dilimabaka N, Mounguegui DM, Lekana-Douki SE, Yattara MK, Obame-Nkoghe J, Longo-Pendy NM, Koumba IPK, Mve-Ella OLB, Moukouama SK, Dzembo CE, Bolo LY, Biyie-Bi-Ngoghe P, Mangouka GL, Nzenze JR, Lekana-Douki JB. Biochemical and hematological factors associated with COVID-19 severity among Gabonese patients: A retrospective cohort study. Front Cell Infect Microbiol 2022; 12:975712. [PMID: 36619758 PMCID: PMC9816336 DOI: 10.3389/fcimb.2022.975712] [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: 06/22/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
The COVID-19 disease presents a large range of clinical manifestations and includes asymptomatic, mild, and severe cases. The level of severity is related to parameters associated with immunity, genetics, and biochemistry. Africa shows one of the lowest COVID-19 fatality rates but very few data on the biochemical markers of COVID-19 in patients and the factors associated with disease severity are available for the continent. In Gabon, the COVID-19 fatality rate is only 0.63% but almost no data on biomarkers in COVID-19 patients have been published. Both the number of COVID-19 cases and the mortality rate reported in Africa in general, and in Gabon in particular, are lower than in non-African countries. As such, understanding the factors associated with disease severity in Gabonese patients is a crucial step to better understand the disease in the African context and prepare for future COVID-19 waves and other epidemics of emerging diseases. Here, we compared biochemical and hematological markers among 753 Gabonese COVID-19 patients with asymptomatic (184/753), mild/moderate (420/753), and severe/critical (149/753) forms of the disease using an Analysis of Variance (ANOVA) or a Kruskal-Wallis (KW) test. We modeled these parameters together with comorbidities, age, and sex to predict factors associated with disease severity by using a "binomial generalized linear model" utilizing the "package" stats of R software version 4.0.2. Our results showed that almost all the biochemical and hematological parameters (except creatinine, phosphorus, D-dimers, platelets, and monocytes) varied according to disease severity. However, age and the dysfunction of organs like the kidney, liver, and lung together with the decrease of electrolytes (chloride, potassium, and sodium) are the best predictors of disease severity in Gabonese patients.
Collapse
Affiliation(s)
- Nadine N’dilimabaka
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon,Département de Biologie, Faculté des Sciences, Université des Sciences et Techniques de Masuku (USTM), Franceville, Gabon,*Correspondence: Nadine N’dilimabaka,
| | | | - Sonia Etenna Lekana-Douki
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | | | - Judicaël Obame-Nkoghe
- Département de Biologie, Faculté des Sciences, Université des Sciences et Techniques de Masuku (USTM), Franceville, Gabon,Unité Écologie des Systèmes Vectoriels (ESV), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Neil Michel Longo-Pendy
- Unité Écologie des Systèmes Vectoriels (ESV), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Ingrid Precilya Koumba Koumba
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Octavie Lauris Banga Mve-Ella
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Schedy Koumba Moukouama
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Cresh Emelya Dzembo
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - Lauriane Yacka Bolo
- Unité Emergence des Maladies Virales, Département de Virologie, Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | | | | | - Jean-Raymond Nzenze
- Site Coronavirus, Hôpital d’Instruction des Armes d’Akanda, Libreville-Nord, Gabon
| | - Jean-Bernard Lekana-Douki
- Unité Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon,Département de Parasitologie-Mycologie Médecine Tropicale, Faculté de Médecine, Université des Sciences de la Sante, Libreville, Gabon
| |
Collapse
|
130
|
Liang D, Zhang G, Huang M, Wang L, Hong W, Li A, Liang Y, Wang T, Lu J, Ou M, Ren Z, Lu H, Zheng R, Cai X, Pan X, Xia J, Ke C. Progress of the COVID-19: Persistence, Effectiveness, and Immune Escape of the Neutralizing Antibody in Convalescent Serum. Pathogens 2022; 11:pathogens11121531. [PMID: 36558864 PMCID: PMC9782332 DOI: 10.3390/pathogens11121531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a new coronavirus causing Coronavirus Disease 2019 (COVID-19), is a major topic of global human health concern. The Delta and Omicron variants have caused alarming responses worldwide due to their high transmission rates and a number of mutations. During a one-year follow-up (from June 2020 to June 2021), we included 114 patients with SARS-CoV-2 infection to study the long-term dynamics and the correlative factors of neutralizing antibodies (NAbs) in convalescent patients. The blood samples were collected at two detection time points (at 6 and 12 months after discharge). We evaluated the NAbs response of discharged patients by performing a micro-neutralization assay using a SARS-CoV-2 wild type. In addition, a total of 62 serum samples from discharged COVID-19 patients with Alpha, Beta, Delta, and Omicron variants of infection were enrolled to perform cross-neutralization tests using the original SARS-CoV-2 strain and VOCs variants (including Alpha, Beta, Gamma, Delta, and Omicron variants) and to assess the ability of NAbs against the SARS-CoV-2 variants. NAbs seroconversion occurred in 91.46% of patients (n = 82) in the first timepoint and in 89.29% of patients (n = 84) in the second detection point, and three kinds of NAbs kinetics curves were perceived. The NAbs levels in young patients had higher values than those in elder patients. The kinetics of disease duration was accompanied by an opposite trend in NAbs levels. Despite a declining NAbs response, NAbs activity was still detectable in a substantial proportion of recovered patients one year after discharge. Compared to the wild strain, the Omicron strain could lead to a 23.44-, 3.42-, 8.03-, and 2.57-fold reduction in neutralization capacity in "SAlpha", "SBeta", "SDelta", and "SOmicron", respectively, and the NAbs levels against the Omicron strain were significantly lower than those of the Beta and Delta variants. Remarkably, the NAbs activity of convalescent serum with Omicron strain infection was most obviously detectable against six SARS-CoV-2 strains in our study. The role of the vaccination history in NAbs levels further confirmed the previous study that reported vaccine-induced NAbs as the convincing protection mechanism against SARS-CoV-2. In conclusion, our findings highlighted the dynamics of the long-term immune responses after the disappearance of symptoms and revealed that NAbs levels varied among all types of convalescent patients with COVID-19 and that NAbs remained detectable for one year, which is reassuring in terms of protection against reinfection. Moreover, a moderate correlation between the duration of disease and Nabs titers was observed, whereas age was negatively correlated with Nabs titers. On the other hand, compared with other VOCs, the Omicron variant was able to escape the defenses of the immune system more significantly, and the convalescent serum infected with the Omicron variant played a critical part in protection against different SARS-CoV-2 variants. Recovery serum from individuals vaccinated with inactivated vaccine preceding infection with the Omicron strain had a high efficacy against the original strain and the VOCs variants, whereas the convalescent serum of persons vaccinated by inactivated vaccine prior to infection with the Delta variant was only potent against the wild-type strain.
Collapse
Affiliation(s)
- Dan Liang
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- Emergency Key Team, Guangzhou National Laboratory, Guangzhou 510700, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangzhou 511430, China
| | - Guanting Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangzhou 511430, China
| | - Mingxing Huang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Li Wang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenshan Hong
- MPH Education Center, Shantou University Medical College, Shantou 515041, China
| | - An’an Li
- School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yufeng Liang
- School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Tao Wang
- MPH Education Center, Shantou University Medical College, Shantou 515041, China
| | - Jiahui Lu
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Mengdang Ou
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Zhongqiang Ren
- Second People’s Hospital of Zhongshan, Zhongshan 528447, China
| | - Huiyi Lu
- Second People’s Hospital of Zhongshan, Zhongshan 528447, China
| | - Rutian Zheng
- Huizhou Central People’s Hospital, Huizhou 516001, China
| | - Xionghui Cai
- Huizhou Central People’s Hospital, Huizhou 516001, China
| | - Xingfei Pan
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jinyu Xia
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Changwen Ke
- Emergency Key Team, Guangzhou National Laboratory, Guangzhou 510700, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangzhou 511430, China
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- MPH Education Center, Shantou University Medical College, Shantou 515041, China
- School of Public Health, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
131
|
Boehme M, Guzzetta KE, Wasén C, Cox LM. The gut microbiota is an emerging target for improving brain health during ageing. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 4:E2. [PMID: 37179659 PMCID: PMC10174391 DOI: 10.1017/gmb.2022.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The gut microbiota plays crucial roles in maintaining the health and homeostasis of its host throughout lifespan, including through its ability to impact brain function and regulate behaviour during ageing. Studies have shown that there are disparate rates of biologic ageing despite equivalencies in chronologic age, including in the development of neurodegenerative diseases, which suggests that environmental factors may play an important role in determining health outcomes in ageing. Recent evidence demonstrates that the gut microbiota may be a potential novel target to ameliorate symptoms of brain ageing and promote healthy cognition. This review highlights the current knowledge around the relationships between the gut microbiota and host brain ageing, including potential contributions to age-related neurodegenerative diseases. Furthermore, we assess key areas for which gut microbiota-based strategies may present as opportunities for intervention.
Collapse
Affiliation(s)
- Marcus Boehme
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Katherine Elizabeth Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Caroline Wasén
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Laura Michelle Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
132
|
Mazzaschi G, Quaini F, Buti S. Exploring genetic and immune underpinnings of the sexual dimorphism in tumor response to immune checkpoints inhibitors: A narrative review. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100146. [PMID: 36571078 PMCID: PMC9772791 DOI: 10.1016/j.crphar.2022.100146] [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: 10/06/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction In spite of the undisputed relevance of sex as critical biologic variable of the immune landscape, still limited is our understanding of the basic mechanisms implicated in sex-biased immune response thereby conditioning the therapeutic outcome in cancer patients. This hindrance delays the actual attempts to decipher the heterogeneity of cancer and its immune surveillance, further digressing the achievement of predictive biomarkers in the current immunotherapy-driven scenario. Body: The present review concisely reports on genetic, chromosomal, hormonal, and immune features underlying sex-differences in the response to immune checkpoint inhibitors (ICIs). In addition to outline the need of robust data on ICI pharmaco-kinetics/dynamics, our survey might provide new insights on sex determinants of ICI efficacy and suggests uncovered pathways that warrant prospective investigations. Conclusion According to a sharable view, we propose to widely include sex among the co-variates when assessing the clinical response to ICI in cancer patients.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
133
|
Sun SW, Qi C, Xiong XZ. Challenges of COPD Patients during the COVID-19 Pandemic. Pathogens 2022; 11:pathogens11121484. [PMID: 36558818 PMCID: PMC9788471 DOI: 10.3390/pathogens11121484] [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: 10/05/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a severe systemic infection that is a major threat to healthcare systems worldwide. According to studies, chronic obstructive pulmonary disease (COPD) patients with COVID-19 usually have a high risk of developing severe symptoms and fatality, but limited research has addressed the poor condition of COPD patients during the pandemic. This review focuses on the underlying risk factors including innate immune dysfunction, angiotensin converting enzyme 2 (ACE2) expression, smoking status, precocious differentiation of T lymphocytes and immunosenescence in COPD patients which might account for their poor outcomes during the COVID-19 crisis. Furthermore, we highlight the role of aging of the immune system, which may be the culprit of COVID-19. In brief, we list the challenges of COPD patients in this national pandemic, aiming to provide immune-related considerations to support critical processes in COPD patients during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and inspire immune therapy for these patients.
Collapse
Affiliation(s)
- Sheng-Wen Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Chang Qi
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Correspondence: ; Tel.: +027-85726705
| |
Collapse
|
134
|
Medova V, Hulinkova I, Laiferova N, Urdova V, Ciznar P, Dolnikova D, Krasnanova V, Fabri O, Ficek A, Soltysova A. The importance of defining the age-specific TREC/KREC levels for detection of various inborn errors of immunity in pediatric and adult patients. Clin Immunol 2022; 245:109155. [DOI: 10.1016/j.clim.2022.109155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022]
|
135
|
Abstract
Age is the key risk factor for diseases and disabilities of the elderly. Efforts to tackle age-related diseases and increase healthspan have suggested targeting the ageing process itself to 'rejuvenate' physiological functioning. However, achieving this aim requires measures of biological age and rates of ageing at the molecular level. Spurred by recent advances in high-throughput omics technologies, a new generation of tools to measure biological ageing now enables the quantitative characterization of ageing at molecular resolution. Epigenomic, transcriptomic, proteomic and metabolomic data can be harnessed with machine learning to build 'ageing clocks' with demonstrated capacity to identify new biomarkers of biological ageing.
Collapse
Affiliation(s)
- Jarod Rutledge
- Department of Genetics, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA
- Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
136
|
Hadley JB, Kelher MR, D’Alessandro A, Gamboni F, Hansen K, Coleman J, Jones K, Cohen M, Moore EE, Banerjee A, Silliman CC. A pilot study of the metabolic profiles of apheresis platelets modified by donor age and sex and in vitro short-term incubation with sex hormones. Transfusion 2022; 62:2596-2608. [PMID: 36309927 PMCID: PMC9837799 DOI: 10.1111/trf.17165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Platelets are part of innate immunity and comprise the cellular portion of hemostasis. Platelets express sex hormone receptors on their plasma membrane and sex hormones can alter their function in vitro. Little is known about how age and sex may affect platelet biology; thus, we hypothesized that platelets from males and females have different metabolomic profiles, which may be altered by age and in vitro treatment with sex hormones. METHODS Day 1 apheresis platelets were drawn from five 18-53-year-old, premenopausal younger females (YF), five ≥54-year-old, postmenopausal, older females (OF), five 18-44-year-old younger males (YM), and four ≥45-year-old older males (OM). Platelets were normalized to a standard concentration and metabolomics analyses were completed. Unsupervised statistical analyses and hierarchical clustering with principal component analyses were completed. RESULTS Platelets from OM had (1) elevated mono-, di- and tri-carboxylates, (2) increased levels of free fatty acids, acyl-carnitines, and free amino acids, and (3) increased purine breakdown and deamination products. In vitro incubation with sex hormones only affected platelets from OM donors with trends towards increased ATP and other high-energy purines and decreases in L-proline and other amino acids. CONCLUSION Platelets from OM's versus YF, OF, and YM have a different metabolome implying increased energy metabolism, more free fatty acids, acylcarnitines, and amino acids, and increased breakdown of purines and deamination products. However, only platelets from OM were affected by sex hormones in vitro. Platelets from OM are metabolically distinct, which may impart functional differences when transfused.
Collapse
Affiliation(s)
- Jamie B. Hadley
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Marguerite R. Kelher
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA,Vitalant Research Institute, Denver, Colorado, USA
| | - Angelo D’Alessandro
- The Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Fabia Gamboni
- The Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Kirk Hansen
- The Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Julia Coleman
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Kenneth Jones
- Department of Biostatistics, University of Oklahoma School of Medicine, Oklahoma City, Oklahoma, USA
| | - Mitchell Cohen
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Ernest E. Moore
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Anirban Banerjee
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Christopher C. Silliman
- The Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA,Vitalant Research Institute, Denver, Colorado, USA,The Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
137
|
Comeau KD, Shokoples BG, Schiffrin EL. Sex Differences in the Immune System in Relation to Hypertension and Vascular Disease. Can J Cardiol 2022; 38:1828-1843. [PMID: 35597532 DOI: 10.1016/j.cjca.2022.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the leading risk factor for cardiovascular disease and mortality worldwide. Despite intensive research into the mechanisms underlying the development of hypertension, it remains difficult to control blood pressure in a large proportion of patients. Young men have a higher prevalence of hypertension compared with age-matched women, and this holds true until approximately the fifth decade of life. Following the onset of menopause, the incidence of hypertension among women begins to surpass that of men. The immune system has been demonstrated to play a role in the pathophysiology of hypertension, and biological sex and sex hormones can affect the function of innate and adaptive immune cell populations. Recent studies in male and female animal models of hypertension have begun to unravel the relationship among sex, immunity, and hypertension. Hypertensive male animals show a bias toward proinflammatory T-cell subsets, including interleukin (IL) 17-producing TH17 cells, and increased renal infiltration of T cells and inflammatory macrophages. Conversely, premenopausal female animals are largely protected from hypertension, and have a predilection for anti-inflammatory T regulatory cells and production of anti-inflammatory cytokines, such as IL-10. Menopause abrogates female protection from hypertension, which may be due to changes among anti-inflammatory T regulatory cell populations. Since development of novel treatments for hypertension has plateaued, determining the role of sex in the pathophysiology of hypertension may open new therapeutic avenues for both men and women.
Collapse
Affiliation(s)
- Kevin D Comeau
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Brandon G Shokoples
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada; Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
138
|
Dodd KC, Menon M. Sex bias in lymphocytes: Implications for autoimmune diseases. Front Immunol 2022; 13:945762. [PMID: 36505451 PMCID: PMC9730535 DOI: 10.3389/fimmu.2022.945762] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Autoimmune diseases are characterized by a significant sex dimorphism, with women showing increased susceptibility to disease. This is, at least in part, due to sex-dependent differences in the immune system that are influenced by the complex interplay between sex hormones and sex chromosomes, with contribution from sociological factors, diet and gut microbiota. Sex differences are evident in the number and function of lymphocyte populations. Women mount a stronger pro-inflammatory response than males, with increased lymphocyte proliferation, activation and pro-inflammatory cytokine production, whereas men display expanded regulatory cell subsets. Ageing alters the immune landscape of men and women in differing ways, resulting in changes in autoimmune disease susceptibility. Here we review the current literature on sex differences in lymphocyte function, the factors that influence this, and the implications for autoimmune disease. We propose that improved understanding of sex bias in lymphocyte function can provide sex-specific tailoring of treatment strategies for better management of autoimmune diseases.
Collapse
Affiliation(s)
- Katherine C. Dodd
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, United Kingdom
| | - Madhvi Menon
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,*Correspondence: Madhvi Menon,
| |
Collapse
|
139
|
A Glb1-2A-mCherry reporter monitors systemic aging and predicts lifespan in middle-aged mice. Nat Commun 2022; 13:7028. [PMID: 36396643 PMCID: PMC9671911 DOI: 10.1038/s41467-022-34801-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
The progressive decline of physiological function and the increased risk of age-related diseases challenge healthy aging. Multiple anti-aging manipulations, such as senolytics, have proven beneficial for health; however, the biomarkers that label in vivo senescence at systemic levels are lacking, thus hindering anti-aging applications. In this study, we generate a Glb1+/m‒Glb1-2A-mCherry (GAC) reporter allele at the Glb1 gene locus, which encodes lysosomal β-galactosidase-an enzyme elevated in tissues of old mice. A linear correlation between GAC signal and chronological age is established in a cohort of middle-aged (9 to 13 months) Glb1+/m mice. The high GAC signal is closely associated with cardiac hypertrophy and a shortened lifespan. Moreover, the GAC signal is exponentially increased in pathological senescence induced by bleomycin in the lung. Senolytic dasatinib and quercetin (D + Q) reduce GAC signal in bleomycin treated mice. Thus, the Glb1-2A-mCherry reporter mice monitors systemic aging and function decline, predicts lifespan, and may facilitate the understanding of aging mechanisms and help in the development of anti-aging interventions.
Collapse
|
140
|
Shen Z, Patel MV, Rodriguez-Garcia M, Wira CR. Aging beyond menopause selectively decreases CD8+ T cell numbers but enhances cytotoxic activity in the human endometrium. Immun Ageing 2022; 19:55. [PMID: 36371240 PMCID: PMC9652910 DOI: 10.1186/s12979-022-00312-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/23/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Regulation of endometrial (EM) CD8+ T cells, which provide protection through cell-mediated cytotoxicity, is essential for successful reproduction, and protection against sexually transmitted infections and potential tumors. We have previously demonstrated that EM CD8+ T cell cytotoxicity is suppressed directly and indirectly by sex hormones and enhanced after menopause. What remains unclear is whether CD8+ T cell protection and the contribution of tissue-resident (CD103+) and non-resident (CD103-) T cell populations in the EM change as women age following menopause. RESULTS Using hysterectomy EM tissues, we found that EM CD8+ T cell numbers declined significantly in the years following menopause. Despite an overall decline in CD8+ T cells, cytotoxic activity per cell for both CD103- and CD103 + CD8+ T cells increased with age. Investigation of the underlying mechanisms responsible for cytotoxicity indicated that the percentage of total granzyme A and granzyme B positive CD8+ T cells, but not perforin, increased significantly after menopause and remained high and constant as women aged. Additionally, baseline TNFα production by EM CD8+ T cells increased significantly in the years following menopause, and estradiol suppressed TNFα secretion. Moreover, in response to PMA activation, TNFα and IFNγ were significantly up-regulated, and CD103-CD8+ T cells up-regulation of TNFα, IFNγ and IL-6 increased as women aged. CONCLUSIONS Understanding the underlying factors involved in regulating cell-mediated protection of the EM by CD8+ T cells will contribute to the foundation of information essential for developing therapeutic tools to protect women against gynecological cancers and infections as they age.
Collapse
Affiliation(s)
- Zheng Shen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, 03756, USA
| | - Mickey V Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, 03756, USA
| | | | - Charles R Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, 03756, USA.
| |
Collapse
|
141
|
He F, Furones AR, Landegren N, Fuxe J, Sarhan D. Sex dimorphism in the tumor microenvironment - From bench to bedside and back. Semin Cancer Biol 2022; 86:166-179. [PMID: 35278635 DOI: 10.1016/j.semcancer.2022.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/20/2022] [Accepted: 03/06/2022] [Indexed: 01/27/2023]
Abstract
Cancer represents a significant cause of death and suffering in both the developed and developing countries. Key underlying issues in the mortality of cancer are delayed diagnosis and resistance to treatments. However, improvements in biomarkers represent one important step that can be taken for alleviating the suffering caused by malignancy. Precision-based medicine is promising for revolutionizing diagnostic and treatment strategies for cancer patients worldwide. Contemporary methods, including various omics and systems biology approaches, as well as advanced digital imaging and artificial intelligence, allow more accurate assessment of tumor characteristics at the patient level. As a result, treatment strategies can be specifically tailored and adapted for individual and/or groups of patients that carry certain tumor characteristics. This includes immunotherapy, which is based on characterization of the immunosuppressive tumor microenvironment (TME) and, more specifically, the presence and activity of immune cell subsets. Unfortunately, while it is increasingly clear that gender strongly affects immune regulation and response, there is a knowledge gap concerning differences in sex-specific immune responses and how these contribute to the immunosuppressive TME and the response to immunotherapy. In fact, sex dimorphism is poorly understood in cancer progression and is typically ignored in current clinical practice. In this review, we aim to survey the available literature and highlight the existing knowledge gap in order to encourage further studies that would contribute to understanding both gender-biased immunosuppression in the TME and the driver of tumor progression towards invasive and metastatic disease. The review highlights the need to include sex optimized/genderized medicine as a new concept in future medicine cancer diagnostics and treatments.
Collapse
Affiliation(s)
- Fei He
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden; Department of Urology, First affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Andrea Rodgers Furones
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden; Tumor Immunology Department, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Nils Landegren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 751 23, Sweden; Center for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm 171 76, Sweden
| | - Jonas Fuxe
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
142
|
Ma J, Yao Y, Tian Y, Chen K, Liu B. Advances in sex disparities for cancer immunotherapy: unveiling the dilemma of Yin and Yang. Biol Sex Differ 2022; 13:58. [PMID: 36273184 PMCID: PMC9587634 DOI: 10.1186/s13293-022-00469-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
A wide sex disparity has been demonstrated in cancer incidence, tumor aggressiveness, prognosis, and treatment response of different types of cancer. The sex specificity of cancer appears to be a relevant issue in managing the disease, and studies investigating the role of sex and gender are becoming extremely urgent. Immunotherapy plays a leading role in cancer treatment, offering a new perspective on advanced malignancies. Gender has not been considered in standard cancer treatment, suggesting increasing the recognition of sex differences in cancer research and clinical management. This paper provides an overview of sex and gender disparities in cancer immunotherapy efficacy, anti-cancer immune response, predictive biomarkers, and so on. We focus on the molecular differences between male and female patients across a broad range of cancer types to arouse the attention and practice of clinicians and researchers in a sex perspective of new cancer treatment strategies. Sex differences exist in the prevalence, tumor invasiveness, treatment responses, and clinical outcomes of pan-cancer, suggesting that, while not yet incorporated, sex will probably be considered in future practice guidelines. Inherent genetic differences, overlapping epigenetic alterations, and sex hormone influences underpin everything. Androgen receptors influence the sexual differences in TME by regulating epigenetic and transcriptional differentiation programs. It highlights a sex-based therapeutic target for cancer immunotherapy. Proper consideration of sex, age, sex hormones/menopause status, and socio-cultural gender differences in clinical investigation and gene association studies of cancer are needed to fill current gaps and implement precision medicine for patients with cancer.
Collapse
Affiliation(s)
- Junfu Ma
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Yanxin Yao
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Ye Tian
- Department of Senior Ward, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Ben Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
143
|
Gustani-Buss EG, Buss CE, Cavalli LR, Panis C, Tuon FF, Telles JP, Follador FAC, Wendt GW, Lucio LC, Ferreto LED, de Oliveira IM, Carraro E, David LE, Simão ANC, Boldt ABW, Luiza Petzl-Erler M, Silva WA, Figueiredo DLA. Cross-sectional study for COVID-19-related mortality predictors in a Brazilian state-wide landscape: the role of demographic factors, symptoms and comorbidities. BMJ Open 2022; 12:e056801. [PMID: 36253047 PMCID: PMC9577275 DOI: 10.1136/bmjopen-2021-056801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE The Brazilian state of Paraná has suffered from COVID-19 effects, understanding predictors of increased mortality in health system interventions prevent hospitalisation of patients. We selected the best models to evaluate the association of death with demographic characteristics, symptoms and comorbidities based on three levels of clinical severity for COVID-19: non-hospitalised, hospitalised non-ICU ward and ICU ward. DESIGN Cross-sectional survey using binomial mixed models. SETTING COVID-19-positive cases diagnosed by reverse transcription-PCR of municipalities located in Paraná State. PATIENTS Cases of anonymous datasets of electronic medical records from 1 April 2020 to 31 December 2020. PRIMARY AND SECONDARY OUTCOME MEASURES The best prediction factors were chosen based on criteria after a stepwise analysis using multicollinearity measure, lower Akaike information criterion and goodness-of-fit χ2 tests from univariate to multivariate contexts. RESULTS Male sex was associated with increased mortality among non-hospitalised patients (OR 1.76, 95% CI 1.47 to 2.11) and non-ICU patients (OR 1.22, 95% CI 1.05 to 1.43) for symptoms and for comorbidities (OR 1.89, 95% CI 1.59 to 2.25, and OR 1.30, 95% CI 1.11 to 1.52, respectively). Higher mortality occurred in patients older than 35 years in non-hospitalised (for symptoms: OR 4.05, 95% CI 1.55 to 10.54; and for comorbidities: OR 3.00, 95% CI 1.24 to 7.27) and in hospitalised over 40 years (for symptoms: OR 2.72, 95% CI 1.08 to 6.87; and for comorbidities: OR 2.66, 95% CI 1.22 to 5.79). Dyspnoea was associated with increased mortality in non-hospitalised (OR 4.14, 95% CI 3.45 to 4.96), non-ICU (OR 2.41, 95% CI 2.04 to 2.84) and ICU (OR 1.38, 95% CI 1.10 to 1.72) patients. Neurological disorders (OR 2.16, 95% CI 1.35 to 3.46), neoplastic (OR 3.22, 95% CI 1.75 to 5.93) and kidney diseases (OR 2.13, 95% CI 1.36 to 3.35) showed the majority of increased mortality for ICU as well in the three levels of severity jointly with heart disease, diabetes and CPOD. CONCLUSIONS These findings highlight the importance of the predictor's assessment for the implementation of public healthcare policy in response to the COVID-19 pandemic, mainly to understand how non-pharmaceutical measures could mitigate the virus impact over the population.
Collapse
Affiliation(s)
- Emanuele Gustani Gustani-Buss
- Bioinformatics Laboratory, Institute for Cancer Research, IPEC, Guarapuava, Brazil
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
| | - Carlos E Buss
- Bioinformatics Laboratory, Institute for Cancer Research, IPEC, Guarapuava, Brazil
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- MindFlow Genomics, Guarapuava, Brazil
| | - Luciane R Cavalli
- Postgraduate Program in Biotechnology Applied to Child and Adolescent Health at FPP, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, Western Paraná State University-UNIOESTE, Cascavel, Brazil
| | - Felipe F Tuon
- Laboratory of Emerging Infectious Diseases, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Joao P Telles
- Laboratory of Emerging Infectious Diseases, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Franciele A C Follador
- Department of Life Sciences,Postgraduate Program in Applied Health Sciences, Western Paraná State University-UNIOESTE, Francisco Beltrão, Brazil
| | - Guilherme W Wendt
- Department of Life Sciences,Postgraduate Program in Applied Health Sciences, Western Paraná State University-UNIOESTE, Francisco Beltrão, Brazil
| | - Léia C Lucio
- Department of Life Sciences,Postgraduate Program in Applied Health Sciences, Western Paraná State University-UNIOESTE, Francisco Beltrão, Brazil
| | - Lirane E D Ferreto
- Department of Life Sciences,Postgraduate Program in Applied Health Sciences, Western Paraná State University-UNIOESTE, Francisco Beltrão, Brazil
| | - Isabela M de Oliveira
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- Institute for Cancer Research IPEC, Guarapuava, Brazil
| | - Emerson Carraro
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- Virology Laboratory, Midwestern Parana State University-UNICENTRO, Guarapuava, Brazil
| | - Lualis E David
- Virology Laboratory, Midwestern Parana State University-UNICENTRO, Guarapuava, Brazil
| | - Andréa N C Simão
- Laboratory of Research in Applied Immunology, Department of Pathology, Clinical Analysis and Toxicology, State University of Londrina-UEL, Londrina, Brazil
| | - Angelica B W Boldt
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- Postgraduate Program in Genetics, Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná-UFPR, Curitiba, Brazil
| | - Maria Luiza Petzl-Erler
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- Postgraduate Program in Genetics, Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná-UFPR, Curitiba, Brazil
| | - Wilson A Silva
- Institute for Cancer Research IPEC, Guarapuava, Brazil
- Ribeirão Preto Medical School and Center for Cell-Based Therapy (CEPID/FAPESP), University of São Paulo (USP), Ribeirão Preto, Brazil
| | - David L A Figueiredo
- New Arrangements for Research and Innovation - Genomics-Novos Arranjos de Pesquisa e Inovação - Genômica (NAPI-Genômica), Araucária Foundation - FAAP-PR, Curitiba, Parana, Brazil
- Institute for Cancer Research IPEC, Guarapuava, Brazil
- Department of Medicine, Midwestern Paraná State University-UNICENTRO, Guarapuava, Brazil
| |
Collapse
|
144
|
Repurposing SGLT-2 Inhibitors to Target Aging: Available Evidence and Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms232012325. [PMID: 36293181 PMCID: PMC9604287 DOI: 10.3390/ijms232012325] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/04/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Caloric restriction promotes longevity in multiple animal models. Compounds modulating nutrient-sensing pathways have been suggested to reproduce part of the beneficial effect of caloric restriction on aging. However, none of the commonly studied caloric restriction mimetics actually produce a decrease in calories. Sodium-glucose cotransporter 2 inhibitors (SGLT2-i) are a class of drugs which lower glucose by promoting its elimination through urine, thus inducing a net loss of calories. This effect promotes a metabolic shift at the systemic level, fostering ketones and fatty acids utilization as glucose-alternative substrates, and is accompanied by a modulation of major nutrient-sensing pathways held to drive aging, e.g., mTOR and the inflammasome, overall resembling major features of caloric restriction. In addition, preliminary experimental data suggest that SGLT-2i might also have intrinsic activities independent of their systemic effects, such as the inhibition of cellular senescence. Consistently, evidence from both preclinical and clinical studies have also suggested a marked ability of SGLT-2i to ameliorate low-grade inflammation in humans, a relevant driver of aging commonly referred to as inflammaging. Considering also the amount of data from clinical trials, observational studies, and meta-analyses suggesting a tangible effect on age-related outcomes, such as cardiovascular diseases, heart failure, kidney disease, and all-cause mortality also in patients without diabetes, here we propose a framework where at least part of the benefit provided by SGLT-2i is mediated by their ability to blunt the drivers of aging. To support this postulate, we synthesize available data relative to the effect of this class on: 1- animal models of healthspan and lifespan; 2- selected molecular pillars of aging in preclinical models; 3- biomarkers of aging and especially inflammaging in humans; and 4- COVID-19-related outcomes. The burden of evidence might prompt the design of studies testing the potential employment of this class as anti-aging drugs.
Collapse
|
145
|
Sonkodi B. Psoriasis, Is It a Microdamage of Our "Sixth Sense"? A Neurocentric View. Int J Mol Sci 2022; 23:11940. [PMID: 36233237 PMCID: PMC9569707 DOI: 10.3390/ijms231911940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/23/2022] Open
Abstract
Psoriasis is considered a multifactorial and heterogeneous systemic disease with many underlying pathologic mechanisms having been elucidated; however, the pathomechanism is far from entirely known. This opinion article will demonstrate the potential relevance of the somatosensory Piezo2 microinjury-induced quad-phasic non-contact injury model in psoriasis through a multidisciplinary approach. The primary injury is suggested to be on the Piezo2-containing somatosensory afferent terminals in the Merkel cell−neurite complex, with the concomitant impairment of glutamate vesicular release machinery in Merkel cells. Part of the theory is that the Merkel cell−neurite complex contributes to proprioception; hence, to the stretch of the skin. Piezo2 channelopathy could result in the imbalanced control of Piezo1 on keratinocytes in a clustered manner, leading to dysregulated keratinocyte proliferation and differentiation. Furthermore, the author proposes the role of mtHsp70 leakage from damaged mitochondria through somatosensory terminals in the initiation of autoimmune and autoinflammatory processes in psoriasis. The secondary phase is harsher epidermal tissue damage due to the primary impaired proprioception. The third injury phase refers to re-injury and sensitization with the derailment of healing to a state when part of the wound healing is permanently kept alive due to genetical predisposition and environmental risk factors. Finally, the quadric damage phase is associated with the aging process and associated inflammaging. In summary, this opinion piece postulates that the primary microinjury of our “sixth sense”, or the Piezo2 channelopathy of the somatosensory terminals contributing to proprioception, could be the principal gateway to pathology due to the encroachment of our preprogrammed genetic encoding.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sports Sciences, 1123 Budapest, Hungary
| |
Collapse
|
146
|
Wilmanski T, Gibbons SM, Price ND. Healthy aging and the human gut microbiome: why we cannot just turn back the clock. NATURE AGING 2022; 2:869-871. [PMID: 37118282 PMCID: PMC10155257 DOI: 10.1038/s43587-022-00294-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
The aging research field has largely focused on reversing aging-related changes in the body. However, emerging evidence about the gut microbiome indicates that it may not be optimal to just turn back the clock. Here, we advocate for a more tailored and function-focused approach promoting health across the lifespan.
Collapse
Affiliation(s)
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- eScience Institute, University of Washington, Seattle, WA, USA.
| | - Nathan D Price
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Thorne HealthTech, New York, NY, USA.
| |
Collapse
|
147
|
Iannone F, Crocco P, Dato S, Passarino G, Rose G. Circulating miR-181a as a novel potential plasma biomarker for multimorbidity burden in the older population. BMC Geriatr 2022; 22:772. [PMID: 36175844 PMCID: PMC9520903 DOI: 10.1186/s12877-022-03451-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022] Open
Abstract
Background Chronic low-level inflammation is thought to play a role in many age-related diseases and to contribute to multimorbidity and to the disability related to this condition. In this framework, inflamma-miRs, an important subset of miRNA able to regulate inflammation molecules, appear to be key players. This study aimed to evaluate plasma levels of the inflamma-miR-181a in relation to age, parameters of health status (clinical, physical, and cognitive) and indices of multimorbidity in a cohort of 244 subjects aged 65- 97. Methods MiR-181a was isolated from plasma according to standardized procedures and its expression levels measured by qPCR. Correlation tests and multivariate regression analyses were applied on gender-stratified groups. Results MiR-181a levels resulted increased in old men, and significantly correlated with worsened blood parameters of inflammation (such as low levels of albumin and bilirubin and high lymphocyte content), particularly in females. Furthermore, we found miR-181a positively correlated with the overall multimorbidity burden, measured by CIRS Comorbidity Score, in both genders. Conclusions These data support a role of miR-181a in age-related chronic inflammation and in the development of multimorbidity in older adults and indicate that the routes by which this miRNA influence health status are likely to be gender specific. Based on our results, we suggest that miR-181a is a promising biomarker of health status of the older population. Levels of the inflamma-miR-181a correlate with multimorbidity burden in older people. MiR-181a levels correlate with blood inflammation markers in a gender-specific manner. MiR-181a is positively correlated with age in males but not in females. The paths by which miR-181a can influence health status likely differ between genders.
Collapse
Affiliation(s)
- Francesca Iannone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Serena Dato
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, CS, Italy.
| |
Collapse
|
148
|
Humoral immunoresponse elicited against an adenoviral-based SARS-CoV-2 coronavirus vaccine in elderly patients. Aging (Albany NY) 2022; 14:7193-7205. [PMID: 36152043 PMCID: PMC9550251 DOI: 10.18632/aging.204299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022]
Abstract
The early sequencing of the SARS-CoV-2 viral genome allowed for a speedy development of effective vaccines against the virus. Nevertheless, age-related immunosenescence, the inability to mount strong immune responses, still represents a major obstacle. Here, in a group of 149 elderly volunteers (70-96 years old), evolution of the humoral immune response over time to Gam-COVID-Vac (Sputnik V), a vaccine based on heterologous recombinant adenovirus-26 (Ad26) and adenovirus-5 (Ad5) carrying the Spike genome, was analyzed by an anti-RBD ELISA. At 28 days post vaccination (dpv), a seroconversion rate of 91% was achieved, showing the importance of administering at least two doses of Gam-COVID-Vac to elicit a robust immune response, especially in elderly individuals without previous SARS-CoV-2 infection. Interestingly, IgG specific antibodies that reached their highest titers around 28 dpv (median = 740), persisted without significant decrease after 60 dpv (median = 650). After 90 dpv, IgG titers began to drop, and at 180 dpv only 44.7% of the elderly individuals remained with detectable anti-RBD IgG antibodies. No significant differences were observed in specific humoral immune responses between genders at early times point. However, at 60 dpv anti-RBD titers were more persistent in elderly females, and only dropped at 90 dpv (p < 0.0001). As expected, the highest antibodies titers were elicited in the youngest subgroup (70-74 years). Our results show that Gam-COVID-Vac was able to deal with the ageing of the immune system, eliciting a robust immune response in an elderly cohort, which lasted approximately 90 dpv at high levels, and protected against COVID-19.
Collapse
|
149
|
González-Bermúdez B, Abarca-Ortega A, González-Sánchez M, De la Fuente M, Plaza GR. Possibilities of using T-cell biophysical biomarkers of ageing. Expert Rev Mol Med 2022; 24:e35. [PMID: 36111609 PMCID: PMC9884748 DOI: 10.1017/erm.2022.29] [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: 04/29/2022] [Revised: 08/02/2022] [Accepted: 09/10/2022] [Indexed: 11/07/2022]
Abstract
Ageing is interrelated with the development of immunosenescence. This article focuses on one of the cell sets of the adaptive immune system, T cells, and provides a review of the known changes in T cells associated with ageing. Such fundamental changes affect both cell molecular content and internal ordering. However, acquiring a complete description of the changes at these levels would require extensive measurements of parameters and, furthermore, important fine details of the internal ordering that may be difficult to detect. Therefore, an alternative approach for the characterisation of cells consists of the performance of physical measurements of the whole cell, such as deformability measurements or migration measurements: the physical parameters, complementing the commonly used chemical biomarkers, may contribute to a better understanding of the evolution of T-cell states during ageing. Mechanical measurements, among other biophysical measurements, have the advantage of their relative simplicity: one single parameter agglutinates the complex effects of the variety of changes that gradually appear in cells during ageing.
Collapse
Affiliation(s)
- Blanca González-Bermúdez
- Center for Biomedical Technology, Universidad Politécnica de Madrid, E-28223 Pozuelo de Alarcón, Spain
- Department of Materials Science, E.T.S.I. de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, E-28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Aldo Abarca-Ortega
- Center for Biomedical Technology, Universidad Politécnica de Madrid, E-28223 Pozuelo de Alarcón, Spain
- Department of Materials Science, E.T.S.I. de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, E-28040 Madrid, Spain
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago, Chile
| | - Mónica González-Sánchez
- Department of Genetics, Physiology and Microbiology, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Gustavo R. Plaza
- Center for Biomedical Technology, Universidad Politécnica de Madrid, E-28223 Pozuelo de Alarcón, Spain
- Department of Materials Science, E.T.S.I. de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, E-28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| |
Collapse
|
150
|
Baniel A, Petrullo L, Mercer A, Reitsema L, Sams S, Beehner JC, Bergman TJ, Snyder-Mackler N, Lu A. Maternal effects on early-life gut microbiota maturation in a wild nonhuman primate. Curr Biol 2022; 32:4508-4520.e6. [PMID: 36099914 DOI: 10.1016/j.cub.2022.08.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/14/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
Early-life microbial colonization is an important process shaping host physiology,1-3 immunity,4-6 and long-term health outcomes7-10 in humans. However, our understanding of this dynamic process remains poorly investigated in wild animals,11-13 where developmental mechanisms can be better understood within ecological and evolutionarily relevant contexts.11,12 Using one of the largest developmental datasets on a wild primate-the gelada (Theropithecus gelada)-we used 16S rRNA amplicon sequencing to characterize gut microbiota maturation during the first 3 years of life and assessed the role of maternal effects in shaping offspring microbiota assembly. In contrast to recent data on chimpanzees, postnatal microbial colonization in geladas was highly similar to humans:14 microbial alpha diversity increased rapidly following birth, followed by gradual changes in composition until weaning. Dietary changes associated with weaning (from milk- to plant-based diet) were the main drivers of shifts in taxonomic composition and microbial predicted functional pathways. Maternal effects were also an important factor influencing the offspring gut microbiota. During nursing (<12 months), offspring of experienced (multi-time) mothers exhibited faster functional microbial maturation, likely reflecting the general faster developmental pace of infants born to these mothers. Following weaning (>18 months), the composition of the juvenile microbiota tended to be more similar to the maternal microbiota than to the microbiota of other adult females, highlighting that maternal effects may persist even after nursing cessation.15,16 Together, our findings highlight the dynamic nature of early-life gut colonization and the role of maternal effects in shaping this trajectory in a wild primate.
Collapse
Affiliation(s)
- Alice Baniel
- Center for Evolution and Medicine, Arizona State University, E Tyler Mall, Tempe, AZ 85281, USA; School of Life Sciences, Arizona State University, E Tyler Mall, Tempe, AZ 85287, USA.
| | - Lauren Petrullo
- Department of Psychology, University of Michigan, Church St., Ann Arbor, MI 48109, USA
| | - Arianne Mercer
- Department of Psychology, University of Washington, Okanogan Ln., Seattle, WA 98195, USA
| | - Laurie Reitsema
- Department of Anthropology, University of Georgia, Jackson St., Athens, GA 30602, USA
| | - Sierra Sams
- Department of Psychology, University of Washington, Okanogan Ln., Seattle, WA 98195, USA
| | - Jacinta C Beehner
- Department of Psychology, University of Michigan, Church St., Ann Arbor, MI 48109, USA; Department of Anthropology, University of Michigan, S University Ave., Ann Arbor, MI 48109, USA
| | - Thore J Bergman
- Department of Psychology, University of Michigan, Church St., Ann Arbor, MI 48109, USA; Department of Ecology and Evolutionary Biology, University of Michigan, N University Ave., Ann Arbor, MI 48109, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, E Tyler Mall, Tempe, AZ 85281, USA; School of Life Sciences, Arizona State University, E Tyler Mall, Tempe, AZ 85287, USA; Department of Psychology, University of Washington, Okanogan Ln., Seattle, WA 98195, USA; School for Human Evolution and Social Change, Arizona State University, Cady Mall, Tempe, AZ 85287, USA.
| | - Amy Lu
- Department of Anthropology, Stony Brook University, Circle Rd., Stony Brook, NY 11794, USA.
| |
Collapse
|