1
|
Chen J, Hou R, Xu X, Xie N, Tang J, Li Y, Nie X, Meyer NJ, Su L, Christiani DC, Chen F, Zhang R. Integrative omics and multi-cohort identify IRF1 and biological targets related to sepsis-associated acute respiratory distress syndrome. J Biomed Res 2025; 39:1-12. [PMID: 40420582 DOI: 10.7555/jbr.39.20250066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Interferon-related genes are involved in antiviral responses, inflammation, and immunity, which are closely related to sepsis-associated acute respiratory distress syndrome (ARDS). We analyzed 1972 participants with genotype data and 681 with gene expression data from the Molecular Epidemiology of ARDS (MEARDS), the Molecular Epidemiology of Sepsis in the ICU (MESSI), and the Molecular Diagnosis and Risk Stratification of Sepsis (MARS) cohorts in a three-step study focusing on sepsis-associated ARDS and sepsis-only controls. First, we identified and validated interferon-related genes associated with sepsis-associated ARDS risk using genetically regulated gene expression (GReX). Second, we examined the association of the confirmed gene (interferon regulatory factor 1, IRF1) with ARDS risk and survival and conducted a mediation analysis. Through discovery and validation, we found that the GReX of IRF1 was associated with ARDS risk (OR MEARDS = 0.84, P = 0.008; OR MESSI = 0.83, P = 0.034). Furthermore, individual-level measured IRF1 expression was associated with reduced ARDS risk (OR = 0.58, P = 8.67×10 -4), and improved overall survival in ARDS patients (HR 28-day = 0.49, P = 0.009) and sepsis patients (HR 28-day = 0.76, P = 0.008). Mediation analysis revealed that IRF1 may activate immune functions by regulating the major histocompatibility complex, including HLA-F, which mediated over 70% of the protective effects of IRF1 on ARDS. The findings were validated by in vitro biological experiments involving time-series infection dynamics, overexpression, knockout, and chromatin immunoprecipitation sequencing. Early prophylactic interventions to activate IRF1 in sepsis patients, thereby regulating HLA-F, might reduce the risk of ARDS development and mortality, especially in severely illness patients.
Collapse
Affiliation(s)
- Jiajin Chen
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University School of Medicine, Xiamen, Fujian 361006, China
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ruili Hou
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaowen Xu
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ning Xie
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiaqi Tang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yi Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaoqing Nie
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University School of Medicine, Xiamen, Fujian 361006, China
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Feng Chen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ruyang Zhang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213164, China
| |
Collapse
|
2
|
Manoharan MS, Lee GC, Harper N, Meunier JA, Restrepo MI, Jimenez F, Karekatt S, Branum AP, Gaitan AA, Andampour K, Smith AM, Mader M, Noronha M, Tripathy D, Zhang N, Moreira AG, Pandranki L, Sanchez-Reilly S, Trinh HD, Barnett C, Angel L, Segal LN, Nicholson S, Clark RA, He W, Okulicz JF, Ahuja SK. The 15-Year Survival Advantage: Immune Resilience as a Salutogenic Force in Healthy Aging. Aging Cell 2025:e70063. [PMID: 40264357 DOI: 10.1111/acel.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Human aging presents an evolutionary paradox: while aging rates remain constant, healthspan and lifespan vary widely. We address this conundrum via salutogenesis-the active production of health-through immune resilience (IR), the capacity to resist disease despite aging and inflammation. Analyzing ~17,500 individuals across lifespan stages and inflammatory challenges, we identified a core salutogenic mechanism: IR centered on TCF7, a conserved transcription factor maintaining T-cell stemness and regenerative potential. IR integrates innate and adaptive immunity to counter three aging and mortality drivers: chronic inflammation (inflammaging), immune aging, and cellular senescence. By mitigating these aging mechanisms, IR confers survival advantages: At age 40, individuals with poor IR face a 9.7-fold higher mortality rate-a risk equivalent to that of 55.5-year-olds with optimal IR-resulting in a 15.5-year gap in survival. Optimal IR preserves youthful immune profiles at any age, enhances vaccine responses, and reduces burdens of cardiovascular disease, Alzheimer's, and serious infections. Two key salutogenic evolutionary themes emerge: first, female-predominant IR, including TCF7, likely reflects evolutionary pressures favoring reproductive success and caregiving; second, midlife (40-70 years) is a critical window where optimal IR reduces mortality by 69%. After age 70, mortality rates converge between resilient and non-resilient groups, reflecting biological limits on longevity extension. TNFα-blockers restore salutogenesis pathways, indicating IR delays aging-related processes rather than altering aging rates. By reframing aging as a salutogenic-pathogenic balance, we establish TCF7-centered IR as central to healthy longevity. Targeted midlife interventions to enhance IR offer actionable strategies to maximize healthspan before biological constraints limit benefits.
Collapse
Affiliation(s)
- Muthu Saravanan Manoharan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Grace C Lee
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Nathan Harper
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Justin A Meunier
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Marcos I Restrepo
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Fabio Jimenez
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Sreenath Karekatt
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anne P Branum
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Alvaro A Gaitan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Kian Andampour
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alisha M Smith
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Michael Mader
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Michelle Noronha
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Devjit Tripathy
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Nu Zhang
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alvaro G Moreira
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lavanya Pandranki
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sandra Sanchez-Reilly
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Hanh D Trinh
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Clea Barnett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Luis Angel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Susannah Nicholson
- Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert A Clark
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Weijing He
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | | | - Sunil K Ahuja
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
3
|
Miller JR, Feng C, Ranum J, Striker R. Viruses tipping the scales: The role of the CD4/CD8 ratio in determining viral outcome. Virology 2025; 603:110333. [PMID: 39637475 DOI: 10.1016/j.virol.2024.110333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Here we review recent data on how animal viruses alter the T cell dynamics, and how stressed T cell populations alter viral disease outcomes. The CD4/CD8 ratio of T cells is regulated by the thymus well into adulthood, and determined by both host genetics and environmental exposures. Human and animal data now show many chronic viral infections interact with this ratio and Immune Health Grades, but this raises new questions and justifies new experimental systems. Immune Health Grades and the ratio generally can quantify subtle immunodeficiency for viral infections. Whether or not an animal recovers from a viral infection, and how carcinogenisis proceeds for certain oncogenic viral infections can now be traced back to how "stressed that animal's immune system is as judged by a low CD4/CD8 ratio or elevated Immune Health Grade. We provide this review to encourage basic science virologists using animal models and veterinary virologists to further explore the interactions between the CD4/CD8 T cell ratio and viruses to improve both human and veterinary health.
Collapse
Affiliation(s)
| | | | | | - Rob Striker
- University of Wisconsin-Madison, United States.
| |
Collapse
|
4
|
Pellegrino R, Paganelli R, Di Iorio A, Candeloro M, Volpato S, Bandinelli S, Moretti A, Iolascon G, Tanaka T, Ferrucci L. Role for neurological and immunological resilience in the pathway of the aging muscle powerpenia: InCHIANTI study longitudinal results. GeroScience 2025:10.1007/s11357-025-01536-6. [PMID: 39885114 DOI: 10.1007/s11357-025-01536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
The neuromuscular junction shows several degenerative changes with aging, resulting in a reduction of transmission efficacy. These changes, paired with low-grade chronic inflammation, were considered triggers of the aging muscle processes. The main objective of this study is to assess the role of leukocyte count-derived ratios, nerve conduction velocity (NCV), and compound muscle action potential (CMAP) in determining time-dependent reduction in lower limb muscle explosive strength, a condition that has been defined as powerpenia. The InCHIANTI study enrolled a representative sample from the registry lists of two towns in Tuscany, Italy. Baseline data were collected in 1998, with follow-up visits every 3 years. For the purpose of this analysis, we used 1229 subjects and 3814 follow-up assessments. Subjects with lower values of monocyte-to-lymphocyte ratio (ML-ratio) had higher nerve conduction velocity and higher proximal and distal action potential values; moreover, considering the interaction between age for ML-ratio effect, a statistically significant direct association is found with all the electromyography-parameters. Lower limb muscle power shows a gender dimorphism, male subjects having higher values at baseline, but experiencing steeper decline rate during the follow-up, compared to females. Muscle power was inversely associated with ML-ratio, proximal CMAP, distal CMAP, and NCV. Moreover, we found a direct and statistically significant second-order interaction (age for ML-ratio), meaning that at the same age, increasing ML-ratio increases lower limb muscle power. Lastly, also body composition variation across aging is directly associated with lower limb muscle power. Reduced immunological and neurological homeostasis affects the powerpenia phenotype in a large representative sample of Italian men and women.
Collapse
Affiliation(s)
- Raffaello Pellegrino
- Department of Scientific Research, Campus Ludes, Off-Campus Semmelweis University, 6912, Lugano-Pazzallo, Switzerland
| | - Roberto Paganelli
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Angelo Di Iorio
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Viale Abruzzo 322, 66100, Chieti-Pescara, Italy.
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Matteo Candeloro
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Viale Abruzzo 322, 66100, Chieti-Pescara, Italy
| | - Stefano Volpato
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Bandinelli
- Azienda USL Toscana Centro, InCHIANTI, Villa Margherita, Primo Piano Viale Michelangelo, 41, 50125, Florence, Italy
| | - Antimo Moretti
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| |
Collapse
|
5
|
Hung J, Vonasek B, Rosenberg D, Vo T, Striker R. Using T-Cell Subsets to Better Characterize Immunoresiliency and Immunodeficiency in Patients with Recurrent Infections. Infect Dis Rep 2024; 16:1230-1239. [PMID: 39728019 DOI: 10.3390/idr16060097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Common Variable Immunodeficiency Disease (CVID) and other immunodeficiencies can present in subtle and variable ways. Whether or not a genetic lesion can be identified, there are not well understood biomarkers that quantitatively describe how severe a deficiency is. Here we discuss two possible ranking systems, CD4/CD8 T cell ratios and Immune Health Grades, and how such data maybe applicable to some immunodeficiencies. METHODS This is not a systematic review, but we identify papers relating to immunodeficiencies with enough data to comment on the CD4/CD8 and Immune Health Grade. We also summarized relevant data publicly available from USIDNET, a website that compiles data on immunodeficiencies, and provide two new cases that illustrate ways that this information can alter clinical assessment. RESULTS We review the HIV literature on CD4/CD8 T cell data and how this correlates with both immunologic function and comorbidity better than CD4 count alone. The ratio aslso relates to a new system called Immune Health Grades (IHG) derived from young adult to elderly subjects from many NIH cohorts without HIV. CVID is often thought of as an antibody problem, but in fact most patients also have low CD4/CD8 ratio and other cellular abnormalities. We review IDNET to categorize nine molecular immunodeficiencies including two subcategories of CVID into low, normal, or high ratios. Finally, we present two new cases in the literature of patients with recurrent infection and discuss how viewing the cases through the "lens" of CD4/CD8 ratio and IHG can facilitate clinical decisions. CONCLUSIONS Emerging data suggests at least some immunodeficiencies can be grouped by how abnormal their CD4/CD8 ratio or IHG. This represents a clinically available biomarker that can be tracked to see if the condition is worsening or not.
Collapse
Affiliation(s)
- Justine Hung
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bryan Vonasek
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Daniel Rosenberg
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tri Vo
- Department of Pediatrics, Michigan State University, East Lansing, MI 48842, USA
| | - Rob Striker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
6
|
Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I's of immunity and aging: immunosenescence, inflammaging, and immune resilience. FRONTIERS IN AGING 2024; 5:1490302. [PMID: 39478807 PMCID: PMC11521913 DOI: 10.3389/fragi.2024.1490302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
As we age, our immune system's ability to effectively respond to pathogens declines, a phenomenon known as immunosenescence. This age-related deterioration affects both innate and adaptive immunity, compromising immune function and leading to chronic inflammation that accelerates aging. Immunosenescence is characterized by alterations in immune cell populations and impaired functionality, resulting in increased susceptibility to infections, diminished vaccine efficacy, and higher prevalence of age-related diseases. Chronic low-grade inflammation further exacerbates these issues, contributing to a decline in overall health and resilience. This review delves into the characteristics of immunosenescence and examines the various intrinsic and extrinsic factors contributing to immune aging and how the hallmarks of aging and cell fates can play a crucial role in this process. Additionally, it discusses the impact of sex, age, social determinants, and gut microbiota health on immune aging, illustrating the complex interplay of these factors in altering immune function. Furthermore, the concept of immune resilience is explored, focusing on the metrics for assessing immune health and identifying strategies to enhance immune function. These strategies include lifestyle interventions such as diet, regular physical activity, stress management, and the use of gerotherapeutics and other approaches. Understanding and mitigating the effects of immunosenescence are crucial for developing interventions that support robust immune responses in aged individuals.
Collapse
Affiliation(s)
- Marianna V. Wrona
- Columbia University in the City of New York, New York, NY, United States
| | - Rituparna Ghosh
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Kaitlyn Coll
- Florida International University, Miami, FL, United States
| | - Connor Chun
- Bronx High School of Science, New York, NY, United States
| | - Matthew J. Yousefzadeh
- Columbia University in the City of New York, New York, NY, United States
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
7
|
Holtz A, Van Weyenbergh J, Hong SL, Cuypers L, O'Toole Á, Dudas G, Gerdol M, Potter BI, Ntoumi F, Mapanguy CCM, Vanmechelen B, Wawina-Bokalanga T, Van Holm B, Menezes SM, Soubotko K, Van Pottelbergh G, Wollants E, Vermeersch P, Jacob AS, Maes B, Obbels D, Matheeussen V, Martens G, Gras J, Verhasselt B, Laffut W, Vael C, Goegebuer T, van der Kant R, Rousseau F, Schymkowitz J, Serrano L, Delgado J, Wenseleers T, Bours V, André E, Suchard MA, Rambaut A, Dellicour S, Maes P, Durkin K, Baele G. Emergence of the B.1.214.2 SARS-CoV-2 lineage with an Omicron-like spike insertion and a unique upper airway immune signature. BMC Infect Dis 2024; 24:1139. [PMID: 39390446 PMCID: PMC11468156 DOI: 10.1186/s12879-024-09967-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
We investigate the emergence, mutation profile, and dissemination of SARS-CoV-2 lineage B.1.214.2, first identified in Belgium in January 2021. This variant, featuring a 3-amino acid insertion in the spike protein similar to the Omicron variant, was speculated to enhance transmissibility or immune evasion. Initially detected in international travelers, it substantially transmitted in Central Africa, Belgium, Switzerland, and France, peaking in April 2021. Our travel-aware phylogeographic analysis, incorporating travel history, estimated the origin to the Republic of the Congo, with primary European entry through France and Belgium, and multiple smaller introductions during the epidemic. We correlate its spread with human travel patterns and air passenger data. Further, upon reviewing national reports of SARS-CoV-2 outbreaks in Belgian nursing homes, we found this strain caused moderately severe outcomes (8.7% case fatality ratio). A distinct nasopharyngeal immune response was observed in elderly patients, characterized by 80% unique signatures, higher B- and T-cell activation, increased type I IFN signaling, and reduced NK, Th17, and complement system activation, compared to similar outbreaks. This unique immune response may explain the variant's epidemiological behavior and underscores the need for nasal vaccine strategies against emerging variants.
Collapse
Affiliation(s)
- Andrew Holtz
- Lyssavirus Epidemiology and Neuropathology Unit, Institut Pasteur, Université Paris Cité, Paris, France.
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium.
| | - Samuel L Hong
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Lize Cuypers
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Áine O'Toole
- Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Gytis Dudas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Barney I Potter
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Francine Ntoumi
- Fondation Congolaise Pour La Recherche Médicale, Brazzaville, Republic of Congo
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Claujens Chastel Mfoutou Mapanguy
- Fondation Congolaise Pour La Recherche Médicale, Brazzaville, Republic of Congo
- Faculty of Sciences and Techniques, University Marien Ngouabi, Brazzaville, Republic of Congo
| | - Bert Vanmechelen
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Tony Wawina-Bokalanga
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Bram Van Holm
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Soraya Maria Menezes
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | | | | | - Elke Wollants
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Pieter Vermeersch
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Ann-Sophie Jacob
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Jessa Hospital, Hasselt, Belgium
- Hasselt University, Hasselt, Belgium
- Limburg Clinical Research Center, Hasselt, Belgium
| | | | - Veerle Matheeussen
- Department of Laboratory Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Laboratory of Medical Biochemistry and Laboratory of Medical Microbiology, University of Antwerp, Wilrijk, Belgium
| | - Geert Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| | - Jérémie Gras
- Institut de Pathologie Et de Génétique, Gosselies, Belgium
| | - Bruno Verhasselt
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Wim Laffut
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Carl Vael
- Department of Laboratory Medicine, KLINA General Hospital, Brasschaat, AZ, Belgium
| | | | - Rob van der Kant
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Luis Serrano
- Center for Genomic Regulation, Barcelona Institute for Science and Technology, 08003, Barcelona, Spain
- Universitat Pompeu Fabra, 08002, Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats, 08010, Barcelona, Spain
| | - Javier Delgado
- Center for Genomic Regulation, Barcelona Institute for Science and Technology, 08003, Barcelona, Spain
| | | | - Vincent Bours
- Department of Medical Genetics, CHU Liege, Liege, Belgium
| | - Emmanuel André
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Marc A Suchard
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Andrew Rambaut
- Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Simon Dellicour
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Université Libre de Bruxelles, Brussels, Belgium
| | - Piet Maes
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Keith Durkin
- Laboratory of Human Genetics, GIGA Research Institute, Liège, Belgium
| | - Guy Baele
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Woolley G, Kroll K, Hoffman K, Ward A, Corneli A, Mudrak SV, Qureshi MU, Okeke NL, Chan C, Jones AAD, Tomaras GD, Reeves RK. The Climate Change Burden on Immune Health: Are Persons Living with HIV More at Risk? AIDS Res Hum Retroviruses 2024; 40:549-554. [PMID: 38753709 PMCID: PMC11535442 DOI: 10.1089/aid.2024.0050] [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] [Indexed: 05/18/2024] Open
Abstract
Climate change poses one of the most significant modern threats to overall human health,especially for vulnerable populations including persons living with HIV (PLWH). In this perspective, we specifically explore the concept of immune resilience in human health and how climate change phenomena - including extreme weather events, food insecurity, pollution, and emerging diseases - may exacerbate immune dysfunction and comorbidities faced by PLWH and hinder access to HIV treatment and prevention services. Multidisciplinary, collaborative efforts are urgently needed to quantify these impacts, develop mitigation strategies, and strengthen policies and funding to bolster immune resilience for PLWH in the face of accelerating climate change.
Collapse
Affiliation(s)
- Griffin Woolley
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Kyle Kroll
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Kate Hoffman
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Ashley Ward
- Nicholas Institute for Energy, Environment & Sustainability, Duke University, Durham, North Carolina, USA
| | - Amy Corneli
- Department of Population Health Sciences, Duke University, Durham, North Carolina, USA
- Center for AIDS Research, Duke University, Durham, North Carolina, USA
| | - Sarah V. Mudrak
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - M. Umar Qureshi
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - N. Lance Okeke
- Center for AIDS Research, Duke University, Durham, North Carolina, USA
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | | | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Center for AIDS Research, Duke University, Durham, North Carolina, USA
| | - R. Keith Reeves
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Center for AIDS Research, Duke University, Durham, North Carolina, USA
| |
Collapse
|
9
|
Tripodi D, Dominici R, Sacco D, Pozzobon C, Spiti S, Falbo R, Brambilla P, Mascagni P, Leoni V. Antibody Response against SARS-CoV-2 after mRNA Vaccine in a Cohort of Hospital Healthy Workers Followed for 17 Months. Vaccines (Basel) 2024; 12:506. [PMID: 38793757 PMCID: PMC11125999 DOI: 10.3390/vaccines12050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
The assessment of antibody response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is of critical importance to verify the protective efficacy of available vaccines. Hospital healthcare workers play an essential role in the care and treatment of patients and were particularly at risk of contracting the SARS-CoV-2 infection during the pandemic. The vaccination protocol introduced in our hospital protected the workers and contributed to the containment of the infection' s spread and transmission, although a reduction in vaccine efficacy against symptomatic and breakthrough infections in vaccinated individuals was observed over time. Here, we present the results of a longitudinal and prospective analysis of the anti-SARS-CoV-2 antibodies at multiple time points over a 17-month period to determine how circulating antibody levels change over time following natural infection and vaccination for SARS-CoV-2 before (T0-T4) and after the spread of the omicron variant (T5-T6), analyzing the antibody response of 232 healthy workers at the Pio XI hospital in Desio. A General Estimating Equation model indicated a significant association of the antibody response with time intervals and hospital area, independent of age and sex. Specifically, a similar pattern of antibody response was observed between the surgery and administrative departments, and a different pattern with higher peaks of average antibody response was observed in the emergency and medical departments. Furthermore, using a logistic model, we found no differences in contracting SARS-CoV-2 after the third dose based on the hospital department. Finally, analysis of antibody distribution following the spread of the omicron variant, subdividing the cohort of positive individuals into centiles, highlighted a cut-off of 550 BAU/mL and showed that subjects with antibodies below this are more susceptible to infection than those with a concentration above the established cut-off value.
Collapse
Affiliation(s)
- Domenico Tripodi
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto Dominici
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Davide Sacco
- Department of Brain and Behavioural Sciences, Università degli Studi di Pavia, 27100 Pavia, Italy;
- Laboratory of Medical Genetics, Centro Diagnostico Italiano, 20100 Milan, Italy
| | - Claudia Pozzobon
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Simona Spiti
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Rosanna Falbo
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
| | - Paolo Brambilla
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Paolo Mascagni
- Clinical Unit of Occupational Health, Desio Hospital, ASST Brianza, 20832 Desio, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
10
|
Kempaiah P, Libertin CR, Chitale RA, Naeyma I, Pleqi V, Sheele JM, Iandiorio MJ, Hoogesteijn AL, Caulfield TR, Rivas AL. Decoding Immuno-Competence: A Novel Analysis of Complete Blood Cell Count Data in COVID-19 Outcomes. Biomedicines 2024; 12:871. [PMID: 38672225 PMCID: PMC11048687 DOI: 10.3390/biomedicines12040871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND While 'immuno-competence' is a well-known term, it lacks an operational definition. To address this omission, this study explored whether the temporal and structured data of the complete blood cell count (CBC) can rapidly estimate immuno-competence. To this end, one or more ratios that included data on all monocytes, lymphocytes and neutrophils were investigated. MATERIALS AND METHODS Longitudinal CBC data collected from 101 COVID-19 patients (291 observations) were analyzed. Dynamics were estimated with several approaches, which included non-structured (the classic CBC format) and structured data. Structured data were assessed as complex ratios that capture multicellular interactions among leukocytes. In comparing survivors with non-survivors, the hypothesis that immuno-competence may exhibit feedback-like (oscillatory or cyclic) responses was tested. RESULTS While non-structured data did not distinguish survivors from non-survivors, structured data revealed immunological and statistical differences between outcomes: while survivors exhibited oscillatory data patterns, non-survivors did not. In survivors, many variables (including IL-6, hemoglobin and several complex indicators) showed values above or below the levels observed on day 1 of the hospitalization period, displaying L-shaped data distributions (positive kurtosis). In contrast, non-survivors did not exhibit kurtosis. Three immunologically defined data subsets included only survivors. Because information was based on visual patterns generated in real time, this method can, potentially, provide information rapidly. DISCUSSION The hypothesis that immuno-competence expresses feedback-like loops when immunological data are structured was not rejected. This function seemed to be impaired in immuno-suppressed individuals. While this method rapidly informs, it is only a guide that, to be confirmed, requires additional tests. Despite this limitation, the fact that three protective (survival-associated) immunological data subsets were observed since day 1 supports many clinical decisions, including the early and personalized prognosis and identification of targets that immunomodulatory therapies could pursue. Because it extracts more information from the same data, structured data may replace the century-old format of the CBC.
Collapse
Affiliation(s)
- Prakasha Kempaiah
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Rohit A. Chitale
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Islam Naeyma
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
| | - Vasili Pleqi
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Michelle J. Iandiorio
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | | | - Thomas R. Caulfield
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ariel L. Rivas
- Center for Global Health, Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
11
|
Black CA, Benavides R, Bandy SM, Dallas SD, Gawrys G, So W, Moreira AG, Aguilar S, Quidilla K, Smelter DF, Reveles KR, Frei CR, Koeller JM, Lee GC. Diverse Role of blaCTX-M and Porins in Mediating Ertapenem Resistance among Carbapenem-Resistant Enterobacterales. Antibiotics (Basel) 2024; 13:185. [PMID: 38391571 PMCID: PMC10885879 DOI: 10.3390/antibiotics13020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Among carbapenem-resistant Enterobacterales (CRE) are diverse mechanisms, including those that are resistant to meropenem but susceptible to ertapenem, adding further complexity to the clinical landscape. This study investigates the emergence of ertapenem-resistant, meropenem-susceptible (ErMs) Escherichia coli and Klebsiella pneumoniae CRE across five hospitals in San Antonio, Texas, USA, from 2012 to 2018. The majority of the CRE isolates were non-carbapenemase producers (NCP; 54%; 41/76); 56% of all NCP isolates had an ErMs phenotype. Among ErMs strains, E. coli comprised the majority (72%). ErMs strains carrying blaCTX-M had, on average, 9-fold higher copies of blaCTX-M than CP-ErMs strains as well as approximately 4-fold more copies than blaCTX-M-positive but ertapenem- and meropenem-susceptible (EsMs) strains (3.7 vs. 0.9, p < 0.001). Notably, carbapenem hydrolysis was observed to be mediated by strains harboring blaCTX-M with and without a carbapenemase(s). ErMs also carried more mobile genetic elements, particularly IS26 composite transposons, than EsMs (37 vs. 0.2, p < 0.0001). MGE- ISVsa5 was uniquely more abundant in ErMs than either EsMs or ErMr strains, with over 30 more average ISVsa5 counts than both phenotype groups (p < 0.0001). Immunoblot analysis demonstrated the absence of OmpC expression in NCP-ErMs E. coli, with 92% of strains lacking full contig coverage of ompC. Overall, our findings characterize both collaborative and independent efforts between blaCTX-M and OmpC in ErMs strains, indicating the need to reappraise the term "non-carbapenemase (NCP)", particularly for strains highly expressing blaCTX-M. To improve outcomes for CRE-infected patients, future efforts should focus on mechanisms underlying the emerging ErMs subphenotype of CRE strains to develop technologies for its rapid detection and provide targeted therapeutic strategies.
Collapse
Affiliation(s)
- Cody A. Black
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Raymond Benavides
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Sarah M. Bandy
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Steven D. Dallas
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- University Health System, San Antonio, TX 78229, USA
| | - Gerard Gawrys
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- University Health System, San Antonio, TX 78229, USA
| | - Wonhee So
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Alvaro G. Moreira
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Samantha Aguilar
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- University Health System, San Antonio, TX 78229, USA
| | - Kevin Quidilla
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Dan F. Smelter
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kelly R. Reveles
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Christopher R. Frei
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- University Health System, San Antonio, TX 78229, USA
| | - Jim M. Koeller
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Grace C. Lee
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (C.A.B.)
- Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| |
Collapse
|
12
|
Wilcox DR, Rudmann EA, Ye E, Noori A, Magdamo C, Jain A, Alabsi H, Foy B, Triant VA, Robbins GK, Westover MB, Das S, Mukerji SS. Cognitive concerns are a risk factor for mortality in people with HIV and coronavirus disease 2019. AIDS 2023; 37:1565-1571. [PMID: 37195278 PMCID: PMC10355333 DOI: 10.1097/qad.0000000000003595] [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: 12/05/2022] [Accepted: 05/03/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Data supporting dementia as a risk factor for coronavirus disease 2019 (COVID-19) mortality relied on ICD-10 codes, yet nearly 40% of individuals with probable dementia lack a formal diagnosis. Dementia coding is not well established for people with HIV (PWH), and its reliance may affect risk assessment. METHODS This retrospective cohort analysis of PWH with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PCR positivity includes comparisons to people without HIV (PWoH), matched by age, sex, race, and zipcode. Primary exposures were dementia diagnosis, by International Classification of Diseases (ICD)-10 codes, and cognitive concerns, defined as possible cognitive impairment up to 12 months before COVID-19 diagnosis after clinical review of notes from the electronic health record. Logistic regression models assessed the effect of dementia and cognitive concerns on odds of death [odds ratio (OR); 95% CI (95% confidence interval)]; models adjusted for VACS Index 2.0. RESULTS Sixty-four PWH were identified out of 14 129 patients with SARS-CoV-2 infection and matched to 463 PWoH. Compared with PWoH, PWH had a higher prevalence of dementia (15.6% vs. 6%, P = 0.01) and cognitive concerns (21.9% vs. 15.8%, P = 0.04). Death was more frequent in PWH ( P < 0.01). Adjusted for VACS Index 2.0, dementia [2.4 (1.0-5.8), P = 0.05] and cognitive concerns [2.4 (1.1-5.3), P = 0.03] were associated with increased odds of death. In PWH, the association between cognitive concern and death trended towards statistical significance [3.92 (0.81-20.19), P = 0.09]; there was no association with dementia. CONCLUSION Cognitive status assessments are important for care in COVID-19, especially among PWH. Larger studies should validate findings and determine long-term COVID-19 consequences in PWH with preexisting cognitive deficits.
Collapse
Affiliation(s)
- Douglas R. Wilcox
- Department of Neurology, Massachusetts General Hospital
- Department of Neurology, Brigham and Women's Hospital
- Department of Neurology, Harvard Medical School
| | - Emily A. Rudmann
- Neuroimmunology and Neuro-Infectious Diseases Division, Department of Neurology, Massachusetts General Hospital, Boston
- Division of Infectious Diseases, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Charlestown
| | - Elissa Ye
- Department of Neurology, Massachusetts General Hospital
| | - Ayush Noori
- Department of Neurology, Massachusetts General Hospital
| | - Colin Magdamo
- Department of Neurology, Massachusetts General Hospital
| | - Aayushee Jain
- Department of Neurology, Massachusetts General Hospital
| | - Haitham Alabsi
- Department of Neurology, Massachusetts General Hospital
- Department of Neurology, Harvard Medical School
| | - Brody Foy
- Center for Systems Biology, Massachusetts General Hospital, and Department of Systems Biology, Harvard Medical School
| | - Virginia A. Triant
- Division of Infectious Diseases
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - M. Brandon Westover
- Department of Neurology, Massachusetts General Hospital
- Department of Neurology, Harvard Medical School
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital
- Department of Neurology, Harvard Medical School
| | - Shibani S. Mukerji
- Neuroimmunology and Neuro-Infectious Diseases Division, Department of Neurology, Massachusetts General Hospital, Boston
- Division of Infectious Diseases, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Charlestown
| |
Collapse
|
13
|
Ahuja SK, Manoharan MS, Lee GC, McKinnon LR, Meunier JA, Steri M, Harper N, Fiorillo E, Smith AM, Restrepo MI, Branum AP, Bottomley MJ, Orrù V, Jimenez F, Carrillo A, Pandranki L, Winter CA, Winter LA, Gaitan AA, Moreira AG, Walter EA, Silvestri G, King CL, Zheng YT, Zheng HY, Kimani J, Blake Ball T, Plummer FA, Fowke KR, Harden PN, Wood KJ, Ferris MT, Lund JM, Heise MT, Garrett N, Canady KR, Abdool Karim SS, Little SJ, Gianella S, Smith DM, Letendre S, Richman DD, Cucca F, Trinh H, Sanchez-Reilly S, Hecht JM, Cadena Zuluaga JA, Anzueto A, Pugh JA, Agan BK, Root-Bernstein R, Clark RA, Okulicz JF, He W. Immune resilience despite inflammatory stress promotes longevity and favorable health outcomes including resistance to infection. Nat Commun 2023; 14:3286. [PMID: 37311745 PMCID: PMC10264401 DOI: 10.1038/s41467-023-38238-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/17/2023] [Indexed: 06/15/2023] Open
Abstract
Some people remain healthier throughout life than others but the underlying reasons are poorly understood. Here we hypothesize this advantage is attributable in part to optimal immune resilience (IR), defined as the capacity to preserve and/or rapidly restore immune functions that promote disease resistance (immunocompetence) and control inflammation in infectious diseases as well as other causes of inflammatory stress. We gauge IR levels with two distinct peripheral blood metrics that quantify the balance between (i) CD8+ and CD4+ T-cell levels and (ii) gene expression signatures tracking longevity-associated immunocompetence and mortality-associated inflammation. Profiles of IR metrics in ~48,500 individuals collectively indicate that some persons resist degradation of IR both during aging and when challenged with varied inflammatory stressors. With this resistance, preservation of optimal IR tracked (i) a lower risk of HIV acquisition, AIDS development, symptomatic influenza infection, and recurrent skin cancer; (ii) survival during COVID-19 and sepsis; and (iii) longevity. IR degradation is potentially reversible by decreasing inflammatory stress. Overall, we show that optimal IR is a trait observed across the age spectrum, more common in females, and aligned with a specific immunocompetence-inflammation balance linked to favorable immunity-dependent health outcomes. IR metrics and mechanisms have utility both as biomarkers for measuring immune health and for improving health outcomes.
Collapse
Affiliation(s)
- Sunil K Ahuja
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Muthu Saravanan Manoharan
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Grace C Lee
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Justin A Meunier
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Nathan Harper
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Alisha M Smith
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Marcos I Restrepo
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Anne P Branum
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Matthew J Bottomley
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX1 2JD, UK
- Oxford Kidney Unit, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Fabio Jimenez
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Andrew Carrillo
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Lavanya Pandranki
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Caitlyn A Winter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Lauryn A Winter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Alvaro A Gaitan
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Alvaro G Moreira
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Elizabeth A Walter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Guido Silvestri
- Department of Pathology, Emory University School of Medicine & Emory National Primate Research Center, Atlanta, GA, 30322, USA
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- National Resource Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Hong-Yi Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- National Resource Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Joshua Kimani
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - T Blake Ball
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Keith R Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Paul N Harden
- Oxford Kidney Unit, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX1 2JD, UK
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Mark T Heise
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Kristen R Canady
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Susan J Little
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sara Gianella
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Davey M Smith
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Scott Letendre
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
| | - Douglas D Richman
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, 07100, Italy
| | - Hanh Trinh
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Sandra Sanchez-Reilly
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Joan M Hecht
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Jose A Cadena Zuluaga
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Antonio Anzueto
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jacqueline A Pugh
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Brian K Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | | | - Robert A Clark
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Jason F Okulicz
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Department of Medicine, Infectious Diseases Service, Brooke Army Medical Center, San Antonio, TX, 78234, USA
| | - Weijing He
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| |
Collapse
|
14
|
Karagiannis TT, Dowrey TW, Villacorta-Martin C, Montano M, Reed E, Belkina AC, Andersen SL, Perls TT, Monti S, Murphy GJ, Sebastiani P. Multi-modal profiling of peripheral blood cells across the human lifespan reveals distinct immune cell signatures of aging and longevity. EBioMedicine 2023; 90:104514. [PMID: 37005201 PMCID: PMC10114155 DOI: 10.1016/j.ebiom.2023.104514] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/27/2023] [Accepted: 02/22/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Age-related changes in immune cell composition and functionality are associated with multimorbidity and mortality. However, many centenarians delay the onset of aging-related disease suggesting the presence of elite immunity that remains highly functional at extreme old age. METHODS To identify immune-specific patterns of aging and extreme human longevity, we analyzed novel single cell profiles from the peripheral blood mononuclear cells (PBMCs) of a random sample of 7 centenarians (mean age 106) and publicly available single cell RNA-sequencing (scRNA-seq) datasets that included an additional 7 centenarians as well as 52 people at younger ages (20-89 years). FINDINGS The analysis confirmed known shifts in the ratio of lymphocytes to myeloid cells, and noncytotoxic to cytotoxic cell distributions with aging, but also identified significant shifts from CD4+ T cell to B cell populations in centenarians suggesting a history of exposure to natural and environmental immunogens. We validated several of these findings using flow cytometry analysis of the same samples. Our transcriptional analysis identified cell type signatures specific to exceptional longevity that included genes with age-related changes (e.g., increased expression of STK17A, a gene known to be involved in DNA damage response) as well as genes expressed uniquely in centenarians' PBMCs (e.g., S100A4, part of the S100 protein family studied in age-related disease and connected to longevity and metabolic regulation). INTERPRETATION Collectively, these data suggest that centenarians harbor unique, highly functional immune systems that have successfully adapted to a history of insults allowing for the achievement of exceptional longevity. FUNDING TK, SM, PS, GM, SA, TP are supported by NIH-NIAUH2AG064704 and U19AG023122. MM and PS are supported by NIHNIA Pepper center: P30 AG031679-10. This project is supported by the Flow Cytometry Core Facility at BUSM. FCCF is funded by the NIH Instrumentation grant: S10 OD021587.
Collapse
Affiliation(s)
- Tanya T Karagiannis
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA.
| | - Todd W Dowrey
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Monty Montano
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Boston Pepper Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric Reed
- Data Intensive Study Center, Tufts University, Boston, MA, USA
| | - Anna C Belkina
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Stacy L Andersen
- Department of Medicine, Geriatrics Section, Boston University School of Medicine, Boston, MA, USA
| | - Thomas T Perls
- Department of Medicine, Geriatrics Section, Boston University School of Medicine, Boston, MA, USA
| | - Stefano Monti
- Bioinformatics Program, Boston University, Boston, MA, USA; Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - George J Murphy
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA; Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA; Department of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
15
|
Coronavirus disease 2019 vaccine effectiveness among a population-based cohort of people with HIV. AIDS 2023; 37:365-366. [PMID: 36541649 DOI: 10.1097/qad.0000000000003439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Moran CA, Oliver NT, Szabo B, Collins LF, Nguyen MLT, Shah NS, Moanna A, Colasanti JA, Cantos VD, Armstrong WS, Sheth AN, Ofotokun I, Kelley CF, Marconi VC, Lahiri CD. The association between comorbidities and coronavirus disease 2019 hospitalization among people with HIV differs by age. AIDS 2023; 37:71-81. [PMID: 36111530 PMCID: PMC9742289 DOI: 10.1097/qad.0000000000003386] [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] [Indexed: 01/25/2023]
Abstract
OBJECTIVES To determine whether factors associated with coronavirus disease 2019 (COVID-19) hospitalization among people with HIV (PWH) differ by age stratum. DESIGN Retrospective cohort study. METHODS All adult PWH with a positive SARS-CoV-2 PCR in a public safety-net health system between 1 March 2020 and 28 February 2021 and a Veterans Affairs Medical Center between 1 1 March 2020 and 15 November 2020 in Atlanta, Georgia were included. We performed multivariable logistic regression to determine demographic and clinical factors associated with COVID-19 hospitalization overall and stratified by age less than 50 and at least 50 years. RESULTS Three hundred and sixty-five PWH (mean age 49 years, 74% cisgender male, 82% black) were included. Ninety-six percent were on antiretroviral therapy (ART), 87% had CD4 + T-cell count at least 200 cells/μl, and 89% had HIV-1 RNA less than 200 copies/ml. Overall, age [adjusted odds ratio (aOR) 95% confidence interval (CI) 1.07 (1.04-1.10)], later date of SARS-CoV-2 infection [aOR 0.997 (0.995-1.00)], heart disease [aOR 2.27 (1.06-4.85)], and history of hepatitis C virus (HCV) [aOR 2.59 (1.13-5.89)] were associated with COVID-19 hospitalization. Age-adjusted comorbidity burden was associated with 30% increased risk of hospitalization [aOR 1.30 (1.11-1.54)]. Among 168 PWH less than 50 years old, older age [aOR 1.09 (1.01-1.18)] and no ART use [aOR 40.26 (4.12-393.62)] were associated with hospitalization; age-adjusted comorbidity burden was not ( P = 0.25). Among 197 PWH at least 50, older age [aOR 1.10 (1.04-1.16)], heart disease [aOR 2.45 (1.04-5.77)], history of HCV [aOR 3.52 (1.29-9.60)], and age-adjusted comorbidity burden [aOR 1.36 (1.12-1.66)] were associated with hospitalization. CONCLUSION Comorbidity burden is more strongly associated with COVID-19 hospitalization among older, rather than younger, PWH. These findings may have important implications for risk-stratifying COVID-19 therapies and booster recommendations in PWH.
Collapse
Affiliation(s)
- Caitlin A Moran
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Nora T Oliver
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Atlanta Veterans Affairs Medical Center, Decatur
| | | | - Lauren F Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Minh Ly T Nguyen
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - N Sarita Shah
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Atlanta Veterans Affairs Medical Center, Decatur
- Department of Epidemiology
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | - Abeer Moanna
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Atlanta Veterans Affairs Medical Center, Decatur
| | - Jonathan A Colasanti
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | - Valeria D Cantos
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Wendy S Armstrong
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Anandi N Sheth
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Ighovwerha Ofotokun
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Colleen F Kelley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| | - Vincent C Marconi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
- Atlanta Veterans Affairs Medical Center, Decatur
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | - Cecile D Lahiri
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
- Grady Health System, Atlanta
| |
Collapse
|
17
|
McColl ER, Croyle MA, Zamboni WC, Honer WG, Heise M, Piquette-Miller M, Goralski KB. COVID-19 Vaccines and the Virus: Impact on Drug Metabolism and Pharmacokinetics. Drug Metab Dispos 2023; 51:130-141. [PMID: 36273826 PMCID: PMC11022893 DOI: 10.1124/dmd.122.000934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/07/2022] [Accepted: 09/30/2022] [Indexed: 01/08/2023] Open
Abstract
This article reports on an American Society of Pharmacology and Therapeutics, Division of Drug Metabolism and Disposition symposium held at Experimental Biology on April 2, 2022, in Philadelphia. As of July 2022, over 500 million people have been infected with SARS-CoV-2 (the virus causing COVID-19) and over 12 billion vaccine doses have been administered. Clinically significant interactions between viral infections and hepatic drug metabolism were first recognized over 40 years ago during a cluster of pediatric theophylline toxicity cases attributed to reduced hepatic drug metabolism amid an influenza B outbreak. Today, a substantive body of research supports that the activated innate immune response generally decreases hepatic cytochrome P450 activity. The interactions extend to drug transporters and other organs and have the potential to impact drug absorption, distribution, metabolism, and excretion (ADME). Based on this knowledge, altered ADME is predicted with SARS-CoV-2 infection or vaccination. The report begins with a clinical case exploring the possibility of SARS-CoV-2 vaccination increasing clozapine levels. This is followed by discussions of how SARS-CoV-2 infection or vaccines alter the metabolism and disposition of complex drugs, such as nanoparticles and biologics and small molecule therapies. The review concludes with a discussion of the effects of viral infections on placental amino acid transport and their potential to impact fetal development. The session improved our understanding of the impact of emerging viral infections and vaccine technologies on drug metabolism and disposition, which will help mitigate drug toxicity and improve drug and vaccine safety and effectiveness. SIGNIFICANCE STATEMENT: Altered pharmacokinetics of small molecule and complex molecule drugs and fetal brain distribution of amino acids following SARS-CoV-2 infection or immunization are possible. The proposed mechanisms involve decreased liver cytochrome P450 metabolism of small molecules, enhanced innate immune system metabolism of complex molecules, and altered placental and fetal blood-brain barrier amino acid transport, respectively. Future research is needed to understand the effects of these interactions on adverse drug responses, drug and vaccine safety, and effectiveness and fetal neurodevelopment.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Maria A Croyle
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William C Zamboni
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William G Honer
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Mark Heise
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Kerry B Goralski
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| |
Collapse
|
18
|
Moreira A, Tovar M, Smith AM, Lee GC, Meunier JA, Cheema Z, Moreira A, Winter C, Mustafa SB, Seidner S, Findley T, Garcia JGN, Thébaud B, Kwinta P, Ahuja SK. Development of a peripheral blood transcriptomic gene signature to predict bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L76-L87. [PMID: 36472344 PMCID: PMC9829478 DOI: 10.1152/ajplung.00250.2022] [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: 08/04/2022] [Revised: 09/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung disease of extreme prematurity, yet mechanisms that associate with or identify neonates with increased susceptibility for BPD are largely unknown. Combining artificial intelligence with gene expression data is a novel approach that may assist in better understanding mechanisms underpinning chronic lung disease and in stratifying patients at greater risk for BPD. The objective of this study is to develop an early peripheral blood transcriptomic signature that can predict preterm neonates at risk for developing BPD. Secondary analysis of whole blood microarray data from 97 very low birth weight neonates on day of life 5 was performed. BPD was defined as positive pressure ventilation or oxygen requirement at 28 days of age. Participants were randomly assigned to a training (70%) and testing cohort (30%). Four gene-centric machine learning models were built, and their discriminatory abilities were compared with gestational age or birth weight. This study adheres to the transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD) statement. Neonates with BPD (n = 62 subjects) exhibited a lower median gestational age (26.0 wk vs. 30.0 wk, P < 0.01) and birth weight (800 g vs. 1,280 g, P < 0.01) compared with non-BPD neonates. From an initial pool (33,252 genes/patient), 4,523 genes exhibited a false discovery rate (FDR) <1%. The area under the receiver operating characteristic curve (AUC) for predicting BPD utilizing gestational age or birth weight was 87.8% and 87.2%, respectively. The machine learning models, using a combination of five genes, revealed AUCs ranging between 85.8% and 96.1%. Pathways integral to T cell development and differentiation were associated with BPD. A derived five-gene whole blood signature can accurately predict BPD in the first week of life.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Miriam Tovar
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Alisha M Smith
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Grace C Lee
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Justin A Meunier
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Zoya Cheema
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Axel Moreira
- Division of Critical Care, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Caitlyn Winter
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Shamimunisa B Mustafa
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Steven Seidner
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Tina Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, Texas
| | - Joe G N Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Przemko Kwinta
- Neonatal Intensive Care Unit, Department of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Sunil K Ahuja
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
19
|
Rao H, Song X, Lei J, Lu P, Zhao G, Kang X, Zhang D, Zhang T, Ren Y, Peng C, Li Y, Pei J, Cao Z. Ibrutinib Prevents Acute Lung Injury via Multi-Targeting BTK, FLT3 and EGFR in Mice. Int J Mol Sci 2022; 23:13478. [PMID: 36362264 PMCID: PMC9657648 DOI: 10.3390/ijms232113478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 09/12/2023] Open
Abstract
Ibrutinib has potential therapeutic or protective effects against viral- and bacterial-induced acute lung injury (ALI), likely by modulating the Bruton tyrosine kinase (BTK) signaling pathway. However, ibrutinib has multi-target effects. Moreover, immunity and inflammation targets in ALI treatment are poorly defined. We investigated whether the BTK-, FLT3-, and EGFR-related signaling pathways mediated the protective effects of ibrutinib on ALI. The intratracheal administration of poly I:C or LPS after ibrutinib administration in mice was performed by gavage. The pathological conditions of the lungs were assessed by micro-CT and HE staining. The levels of neutrophils, lymphocytes, and related inflammatory factors in the lungs were evaluated by ELISA, flow cytometry, immunohistochemistry, and immunofluorescence. Finally, the expression of proteins associated with the BTK-, FLT3-, and EGFR-related signaling pathways were evaluated by Western blotting. Ibrutinib (10 mg/kg) protected against poly I:C-induced (5 mg/kg) and LPS-induced (5 mg/kg) lung inflammation. The wet/dry weight ratio (W/D) and total proteins in the bronchoalveolar lavage fluid (BALF) were markedly reduced after ibrutinib (10 mg/kg) treatment, relative to the poly I:C- and LPS-treated groups. The levels of ALI indicators (NFκB, IL-1β, IL-6, TNF-α, IFN-γ, neutrophils, and lymphocytes) were significantly reduced after treatment. Accordingly, ibrutinib inhibited the poly I:C- and LPS-induced BTK-, FLT3-, and EGFR-related pathway activations. Ibrutinib inhibited poly I:C- and LPS-induced acute lung injury, and this may be due to its ability to suppress the BTK-, FLT3-, and EGFR-related signaling pathways. Therefore, ibrutinib is a potential protective agent for regulating immunity and inflammation in poly I:C- and LPS-induced ALI.
Collapse
Affiliation(s)
- Huanan Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaominting Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jieting Lei
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peng Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Guiying Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin Kang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Duanna Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yali Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
20
|
Wu Q, Pennini ME, Bergmann JN, Kozak ML, Herring K, Sciarretta KL, Armstrong KL. Applying lessons learned from COVID-19 therapeutic trials to improve future ALI/ARDS trials. Open Forum Infect Dis 2022; 9:ofac381. [PMID: 35983268 PMCID: PMC9379817 DOI: 10.1093/ofid/ofac381] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Host-directed therapeutics targeting immune dysregulation are considered the most promising approach to address the unmet clinical need for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) related to coronavirus disease 2019 (COVID-19). To better understand the current clinical study landscape and gaps in treating hospitalized patients with severe or critical COVID-19, we identified COVID-19 trials developing host-directed therapies registered at ClinicalTrials.gov and discussed the factors contributing to the success vs failure of these studies. We have learned, instead of the one-size-fits-all approach, future clinical trials evaluating a targeted immunomodulatory agent in heterogeneous patients with ALI/ARDS due to COVID-19 or other infectious diseases can use immune-based biomarkers in addition to clinical and demographic characteristics to improve patient stratification and inform clinical decision-making. Identifying distinct patient subgroups based on immune profiles across the disease trajectory, regardless of the causative pathogen, may accelerate evaluating host-directed therapeutics in trials of ALI/ARDS and related conditions (eg, sepsis).
Collapse
Affiliation(s)
- Qun Wu
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Meghan E Pennini
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Julie N Bergmann
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Marina L Kozak
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kristen Herring
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Sciarretta
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Armstrong
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| |
Collapse
|
21
|
Şahin EA, Mavi D, Kara E, Sönmezer MÇ, İnkaya AÇ, Ünal S. Integrase inhibitor-based regimens are related to favorable systemic inflammatory index and platecrit scores in people living with HIV (PLWH) up to 2 years. Postgrad Med 2022; 134:635-640. [PMID: 35671079 DOI: 10.1080/00325481.2022.2085931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Despite the advances in antiretroviral treatment (ART), persistent inflammation remained a challenge. We analyzed the inflammatory-score changes through 2-years in people living with HIV (PLWH) treated with different antiretroviral regimes. METHODS This study was conducted in Hacettepe University HIV/AIDS Treatment and Research Center. PLWH diagnosed between 2014 and 2020 were included. Inflammatory and metabolic markers (CD4/CD8 ratio, C-reactive protein (CRP), Systemic Inflammatory Index (SII), Neutrophil-Lymphocyte Ratio (NLR), Mean Platelet Volume (MPV), Platecrite (PCT), and Low-Density Lipoprotein/High-Density Lipoprotein (LDL/HDL), Platelet-to-Lymphocyte Ratio (PLR) and ARTs were captured from database through 2-years from the diagnosis. The 2-year change (Δ) in markers was calculated and compared by ART type (backbone and 3rd agent). Mann-Whitney-U test and T-test were used for statistical analysis. RESULTS This study included 205 PLWH; 175 (85.4%) were male, and the mean age was 38.98 (±10.88) years. The number of PLWH with suppressed viremia (<40 HIV-RNA copies/ml) was 164 (80%) at the end of the second year. MPV increased significantly higher among PLWH receiving ABC/3TC compared to PLWH receiving TDF/FTC (p < 0.05). The CD4:CD8 ratio increased, and SII, NLR, LDL/HDL ratios decreased significantly among PLWH treated with integrase strand transfer inhibitors (INSTI) compared with protease inhibitors (PI) and Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs) (p < 0.05). CONCLUSIONS Integrase inhibitor treatment is related to favorable inflammatory marker profile among PLWH in the 2-year follow-up. A favorable inflammatory profile may, in turn, contribute to the prevention of non-communicable diseases (NCD) among PLWH. This study showed that simple, easy-to-calculate markers could be implemented to define ongoing inflammation among PLWH under suppressive ART.
Collapse
Affiliation(s)
| | - Deniz Mavi
- Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Emre Kara
- Faculty of Pharmacy, Department of Clinical Pharmacy, Hacettepe University, Ankara, Turkey
| | - Meliha Çağla Sönmezer
- Faculty of Medicine, Department of Infectious Diseases, Hacettepe University, Ankara, Turkey
| | - Ahmet Çağkan İnkaya
- Faculty of Medicine, Department of Infectious Diseases, Hacettepe University, Ankara, Turkey
| | - Serhat Ünal
- Faculty of Medicine, Department of Infectious Diseases, Hacettepe University, Ankara, Turkey
| |
Collapse
|
22
|
Lee GC, Moreira AG, Hinojosa C, Benavides R, Winter C, Anderson AC, Chen CJ, Borsa N, Hastings G, Black CA, Bandy SM, Shaffer A, Restrepo MI, Ahuja SK. Metformin Attenuates Inflammatory Responses and Enhances Antibody Production in an Acute Pneumonia Model of Streptococcus pneumoniae. FRONTIERS IN AGING 2022; 3:736835. [PMID: 35821804 PMCID: PMC9261336 DOI: 10.3389/fragi.2022.736835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 04/06/2022] [Indexed: 01/09/2023]
Abstract
Metformin may potentially reverse various age-related conditions; however, it is unclear whether metformin can also mitigate or delay the deterioration of immunological resilience that occurs in the context of infections that are commonly observed in older persons. We examined whether metformin promotes the preservation of immunological resilience in an acute S. pneumoniae (SPN) infection challenge in young adult mice. Mice were fed metformin (MET-alone) or standard chow (controls-alone) for 10 weeks prior to receiving intratracheal inoculation of SPN. A subset of each diet group received pneumococcal conjugate vaccine at week 6 (MET + PCV and control + PCV). Compared to controls-alone, MET-alone had significantly less infection-associated morbidity and attenuated inflammatory responses during acute SPN infection. Metformin lowered the expression of genes in the lungs related to inflammation as well as shorter lifespan in humans. This was accompanied by significantly lower levels of pro-inflammatory cytokines (e.g., IL6). MET + PCV vs. control + PCV manifested enhanced SPN anticapsular IgM and IgG levels. The levels of SPN IgM production negatively correlated with expression levels of genes linked to intestinal epithelial structure among MET + PCV vs. control + PCV groups. Correspondingly, the gut microbial composition of metformin-fed mice had a significantly higher abundance in the Verrucomicrobia, Akkermansia muciniphila, a species previously associated with beneficial effects on intestinal integrity and longevity. Together, these findings indicate metformin's immunoprotective potential to protect against infection-associated declines in immunologic resilience.
Collapse
Affiliation(s)
- Grace C. Lee
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
- Pharmacotherapy Education and Research Center, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- The Foundation for Advancing Veterans’ Health Research, San Antonio, TX, United States
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Alvaro G. Moreira
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, United States
- Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Cecilia Hinojosa
- Department Pulmonary Diseases and Critical Care Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Raymond Benavides
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
- Pharmacotherapy Education and Research Center, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Caitlyn Winter
- The Foundation for Advancing Veterans’ Health Research, San Antonio, TX, United States
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, United States
- Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Audrey C. Anderson
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Chang-Jui Chen
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Noemi Borsa
- Department Pulmonary Diseases and Critical Care Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Respiratory Unit and Adult Cystic Fibrosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabrielyd Hastings
- Department Pulmonary Diseases and Critical Care Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Cody A. Black
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
- Pharmacotherapy Education and Research Center, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Sarah M. Bandy
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
- Pharmacotherapy Education and Research Center, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexander Shaffer
- Department Pulmonary Diseases and Critical Care Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Marcos I. Restrepo
- Department Pulmonary Diseases and Critical Care Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Sunil K. Ahuja
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, United States
- South Texas Veterans Health Care System, San Antonio, TX, United States
- Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
23
|
Rasmussen TA, Ahuja SK, Kuwanda L, Vjecha MJ, Hudson F, Lal L, Rhodes A, Chang J, Palmer S, Auberson-Munderi P, Mugerwa H, Wood R, Badal-Faesen S, Pillay S, Mngqibisa R, LaRosa A, Hildago J, Petoumenos K, Chiu C, Lutaakome J, Kitonsa J, Kabaswaga E, Pala P, Ganoza C, Fisher K, Chang C, Lewin SR, Wright EJ. Antiretroviral Initiation at ≥800 CD4+ Cells/mm3 Associated With Lower Human Immunodeficiency Virus Reservoir Size. Clin Infect Dis 2022; 75:1781-1791. [PMID: 35396591 PMCID: PMC9662177 DOI: 10.1093/cid/ciac249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Identifying factors that determine the frequency of latently infected CD4+ T cells on antiretroviral therapy (ART) may inform strategies for human immunodeficiency virus (HIV) cure. We investigated the role of CD4+ count at ART initiation for HIV persistence on ART. METHODS Among participants of the Strategic Timing of Antiretroviral Treatment Study, we enrolled people with HIV (PWH) who initiated ART with CD4+ T-cell counts of 500-599, 600-799, or ≥ 800 cells/mm3. After 36-44 months on ART, the levels of total HIV-DNA, cell-associated unspliced HIV-RNA (CA-US HIV-RNA), and two-long terminal repeat HIV-DNA in CD4+ T cells were quantified and plasma HIV-RNA was measured by single-copy assay. We measured T-cell expression of Human Leucocyte Antigen-DR Isotype (HLA-DR), programmed death-1, and phosphorylated signal transducer and activator of transcription-5 (pSTAT5). Virological and immunological measures were compared across CD4+ strata. RESULTS We enrolled 146 PWH, 36 in the 500-599, 60 in the 600-799, and 50 in the ≥ 800 CD4 strata. After 36-44 months of ART, total HIV-DNA, plasma HIV-RNA, and HLA-DR expression were significantly lower in PWH with CD4+ T-cell count ≥ 800 cells/mm3 at ART initiation compared with 600-799 or 500-599 cells/mm3. The median level of HIV-DNA after 36-44 months of ART was lower by 75% in participants initiating ART with ≥ 800 vs 500-599 cells/mm3 (median [interquartile range]: 16.3 [7.0-117.6] vs 68.4 [13.7-213.1] copies/million cells, respectively). Higher pSTAT5 expression significantly correlated with lower levels of HIV-DNA and CA-US HIV-RNA. Virological measures were significantly lower in females. CONCLUSIONS Initiating ART with a CD4+ count ≥ 800 cells/mm3 compared with 600-799 or 500-599 cells/mm3 was associated with achieving a substantially smaller HIV reservoir on ART.
Collapse
Affiliation(s)
- Thomas A Rasmussen
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Department of Infectious Diseases, Aarhus University Hospital, AarhusDenmark
| | - Sunil K Ahuja
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Locadiah Kuwanda
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Michael J Vjecha
- Institute for Clinical Research, Inc., Veterans Affairs Medical Center, Washington D.C., USA
| | - Fleur Hudson
- MRC Clinical Trials Unit at UCL, London UK Uganda Virus Research Institute/MRC, London, United Kingdom,LSHTM Uganda Research Unit, HIV Intervention Programme, Entebbe, Uganda
| | | | - Ajantha Rhodes
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Judy Chang
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, Australia
| | | | | | - Robin Wood
- The Desmond Tutu HIV Centre, Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sharlaa Badal-Faesen
- Clinical HIV Research Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Sandy Pillay
- Enhancing Care Foundation, Department of Research and Post-graduate Support, Durban University of Technology, Durban, South Africa
| | - Rosie Mngqibisa
- Enhancing Care Foundation, Department of Research and Post-graduate Support, Durban University of Technology, Durban, South Africa
| | | | | | - Kathy Petoumenos
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Chris Chiu
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Joseph Lutaakome
- LSHTM Uganda Research Unit, HIV Intervention Programme, Entebbe, Uganda,Uganda Virus Research Institute/MRC, Entebbe, Uganda
| | - Jonathan Kitonsa
- LSHTM Uganda Research Unit, HIV Intervention Programme, Entebbe, Uganda,Uganda Virus Research Institute/MRC, Entebbe, Uganda
| | | | | | - Carmela Ganoza
- Asociación Civil Impacta Salud y Educación, Lima, Perú,Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Katie Fisher
- Centre for Virus Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, Australia
| | - Christina Chang
- The Kirby Institute, University of New South Wales, Sydney, Australia,Centre for the AIDS Programme of Research in South Africa, Durban, South Africa,Central Clinical School, Monash University, Infectious Diseases, Melbourne, Australia,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | | | - Edwina J Wright
- Correspondence: E. Wright, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, 85 Commercial Rd, 3004 Melbourne, Australia ()
| |
Collapse
|
24
|
Mukherjee PK, Efferth T, Das B, Kar A, Ghosh S, Singha S, Debnath P, Sharma N, Bhardwaj PK, Haldar PK. Role of medicinal plants in inhibiting SARS-CoV-2 and in the management of post-COVID-19 complications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153930. [PMID: 35114450 PMCID: PMC8730822 DOI: 10.1016/j.phymed.2022.153930] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 05/07/2023]
Abstract
BACKGROUND The worldwide corona virus disease outbreak, generally known as COVID-19 pandemic outbreak resulted in a major health crisis globally. The morbidity and transmission modality of COVID-19 appear more severe and uncontrollable. The respiratory failure and following cardiovascular complications are the main pathophysiology of this deadly disease. Several therapeutic strategies are put forward for the development of safe and effective treatment against SARS-CoV-2 virus from the pharmacological view point but till date there are no specific treatment regimen developed for this viral infection. PURPOSE The present review emphasizes the role of herbs and herbs-derived secondary metabolites in inhibiting SARS-CoV-2 virus and also for the management of post-COVID-19 related complications. This approach will foster and ensure the safeguards of using medicinal plant resources to support the healthcare system. Plant-derived phytochemicals have already been reported to prevent the viral infection and to overcome the post-COVID complications like parkinsonism, kidney and heart failure, liver and lungs injury and mental problems. In this review, we explored mechanistic approaches of herbal medicines and their phytocomponenets as antiviral and post-COVID complications by modulating the immunological and inflammatory states. STUDY DESIGN Studies related to diagnosis and treatment guidelines issued for COVID-19 by different traditional system of medicine were included. The information was gathered from pharmacological or non-pharmacological interventions approaches. The gathered information sorted based on therapeutic application of herbs and their components against SARSCoV-2 and COVID-19 related complications. METHODS A systemic search of published literature was conducted from 2003 to 2021 using different literature database like Google Scholar, PubMed, Science Direct, Scopus and Web of Science to emphasize relevant articles on medicinal plants against SARS-CoV-2 viral infection and Post-COVID related complications. RESULTS Collected published literature from 2003 onwards yielded with total 625 articles, from more than 18 countries. Among these 625 articles, more than 95 medicinal plants and 25 active phytomolecules belong to 48 plant families. Reports on the therapeutic activity of the medicinal plants belong to the Lamiaceae family (11 reports), which was found to be maximum reported from 4 different countries including India, China, Australia, and Morocco. Other reports on the medicinal plant of Asteraceae (7 reports), Fabaceae (8 reports), Piperaceae (3 reports), Zingiberaceae (3 reports), Ranunculaceae (3 reports), Meliaceae (4 reports) were found, which can be explored for the development of safe and efficacious products targeting COVID-19. CONCLUSION Keeping in mind that the natural alternatives are in the priority for the management and prevention of the COVID-19, the present review may help to develop an alternative approach for the management of COVID-19 viral infection and post-COVID complications from a mechanistic point of view.
Collapse
Affiliation(s)
- Pulok K Mukherjee
- Institute of Bioresources and Sustainable Development, Imphal-795001, India; School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Bhaskar Das
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India
| | - Amit Kar
- Institute of Bioresources and Sustainable Development, Imphal-795001, India
| | - Suparna Ghosh
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India
| | - Seha Singha
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India
| | - Pradip Debnath
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India
| | - Nanaocha Sharma
- Institute of Bioresources and Sustainable Development, Imphal-795001, India
| | | | - Pallab Kanti Haldar
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata -700 032, India
| |
Collapse
|
25
|
Du L, Xu C, Zeng Z, Chen F, Tang K, Liang Y, Guo Y. Exploration of induced sputum BIRC3 levels and clinical implications in asthma. BMC Pulm Med 2022; 22:86. [PMID: 35287655 PMCID: PMC8922789 DOI: 10.1186/s12890-022-01887-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/10/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Baculoviral IAP repeat-containing 3 (BIRC3) which encodes a member of the IAP family of proteins upregulated in the asthma expression profile dataset. However, there was few research on studying the clinical implication of BIRC3 in asthma. OBJECTIVE To validate BIRC3 expression and its clinical implications in induced sputum of asthma. METHODS Based on the GSE76262 (118 asthma cases and 21 healthy controls) dataset, differentially expressed genes were screened using R software. Subsequently, BIRC3 mRNA and protein were clinically verified in induced sputum samples through quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Besides, the correlations between BIRC3 expression and asthmatic eosinophilic/allergic inflammation indicators (FeNO, IgE, and EOS%), pulmonary function (FEV1, FEV1% pred, FVC% pred, and FEV1/FVC), and inflammatory cytokines (IL-4, IL-5, IL-13, IL-25, IL-10, IL-33, and TSLP) were analyzed. Finally, BIRC3 mRNA was detected in human primary bronchial epithelial cells stimulated by cytokines (IL-4 or IL-13). RESULTS BIRC3 was screened as a candidate gene in the GSE76262, which was highly expressed in asthma. Highly expressed BIRC3 was positively correlated with eosinophilic and allergic indicators, including FeNO, blood eosinophil, and serum IgE. Moreover, BIRC3 protein was positively associated with inflammation cytokines, like IL-4, IL-5, IL-13, IL-25, IL-10, IL-33, and TSLP, while negatively correlated with FEV1, FEV1%pred, FVC% pred, and FEV1/FVC. Furthermore, the expression of BIRC3 could be induced in primary bronchial epithelial cells treated by cytokines IL-4 or IL-13. CONCLUSIONS BIRC3 significantly increased in induced sputum of asthma and positively correlated with airway eosinophilic and peripheral blood allergic inflammation, type 2 cytokines, and airway obstruction. Increased BIRC3 might be involved in the pathogenesis of asthma by affecting the eosinophilic and allergic inflammation.
Collapse
Affiliation(s)
- Lijuan Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Changyi Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Fengjia Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Kun Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Yuxia Liang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| | - Yubiao Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
26
|
Montano M, Oursler KK, Xu K, Sun YV, Marconi VC. Biological ageing with HIV infection: evaluating the geroscience hypothesis. THE LANCET. HEALTHY LONGEVITY 2022; 3:e194-e205. [PMID: 36092375 PMCID: PMC9454292 DOI: 10.1016/s2666-7568(21)00278-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although people with HIV are living longer, as they age they remain disproportionately burdened with multimorbidity that is exacerbated in resource-poor settings. The geroscience hypothesis postulates that a discrete set of between five and ten hallmarks of biological ageing drive multimorbidity, but these processes have not been systematically examined in the context of people with HIV. We examine four major hallmarks of ageing (macromolecular damage, senescence, inflammation, and stem-cell dysfunction) as gerodrivers in the context of people with HIV. As a counterbalance, we introduce healthy ageing, physiological reserve, intrinsic capacity, and resilience as promoters of geroprotection that counteract gerodrivers. We discuss emerging geroscience-based diagnostic biomarkers and therapeutic strategies, and provide examples based on recent advances in cellular senescence, and other, non-pharmacological approaches. Finally, we present a conceptual model of biological ageing in the general population and in people with HIV that integrates gerodrivers and geroprotectors as modulators of homoeostatic reserves and organ function over the lifecourse.
Collapse
|
27
|
Marconi VC, Krishnan V, Ely EW, Montano M. Immune health grades: Finding resilience in the COVID-19 pandemic and beyond. J Allergy Clin Immunol 2022; 149:565-568. [PMID: 34740606 PMCID: PMC8560746 DOI: 10.1016/j.jaci.2021.10.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Vincent C Marconi
- Emory University School of Medicine and Rollins School of Public Health, Atlanta Veterans Affairs Medical Center, Atlanta, Ga.
| | | | - E Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship Center, Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine at Vanderbilt University Medical Center, Nashville, Tenn; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center, Nashville, Tenn
| | | |
Collapse
|
28
|
PrabhuDas M, Fuldner R, Farber D, Kuchel GA, Mannick J, Nikolich-Zugich J, Sen R, Turner J. Research and resource needs for understanding host immune responses to SARS-CoV-2 and COVID-19 vaccines during aging. NATURE AGING 2021; 1:1073-1077. [PMID: 36908301 PMCID: PMC9997058 DOI: 10.1038/s43587-021-00156-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
On 16 and 17 March 2021, the National Institute of Allergy and Infectious Diseases and the National Institute of Aging convened a virtual workshop to discuss developments in SARS-CoV-2 research pertaining to immune responses in older adults, COVID-19 vaccines in both aged animals and older individuals, and to gain some perspective on the critical knowledge gaps that need addressing to establish scientific priorities for future research studies.
Collapse
Affiliation(s)
- Mercy PrabhuDas
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rebecca Fuldner
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Donna Farber
- Department of Microbiology and Immmunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - George A. Kuchel
- University of Connecticut Center on Aging, UConn Health, Farmington, CT, USA
| | | | - Janko Nikolich-Zugich
- Department of Immunobiology, Arizona Center on Aging, University of Arizona, Tucson, AZ, USA
| | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, MD, USA
| | - Joanne Turner
- Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|