1
|
Crine V, Papenberg G, Johansson J, Boraxbekk CJ, Wåhlin A, Lindenberger U, Lövdén M, Riklund K, Bäckman L, Nyberg L, Karalija N. Associations between inflammation and striatal dopamine D2-receptor availability in aging. J Neuroinflammation 2025; 22:24. [PMID: 39885603 DOI: 10.1186/s12974-025-03355-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Normal brain aging is associated with dopamine decline, which has been linked to age-related cognitive decline. Factors underlying individual differences in dopamine integrity at older ages remain, however, unclear. Here we aimed at investigating: (i) whether inflammation is associated with levels and 5-year changes of in vivo dopamine D2-receptor (DRD2) availability, (ii) if DRD2-inflammation associations differ between men and women, and (iii) whether inflammation and cerebral small-vessel disease (white-matter lesions) serve as two independent predictors of DRD2 availability. METHODS Analyses were performed in a sample of healthy adults > 60 years assessed at two measurement occasions separated by 5 years. At both occasions, DRD2 availability was estimated by 11C-raclopride PET, and white-matter lesions by MRI. Inflammation was assessed by two C-reactive protein-associated DNA methylation scores at study baseline. RESULTS Individuals with higher DNA methylation scores at baseline showed reduced striatal DRD2 availability. An interaction was found between DNA methylation scores and sex in relation to striatal DRD2 availability, such that associations were found in men but not in women. DNA methylation scores at study entrance were not significantly associated with 5-year striatal DRD2 decline rates. No significant association was found between DNA methylation scores and white-matter lesions, but higher scores as well as higher lesion burden were independently associated with reduced striatal DRD2 availability in men. CONCLUSIONS These findings suggest negative associations between one proxy of inflammation and DRD2 availability in older adults, selectively for men who had higher DNA methylation scores. Future studies should investigate other inflammatory markers in relation to dopamine integrity.
Collapse
Affiliation(s)
- Vanessa Crine
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institute and Stockholm University, Stockholm, Sweden
| | - Jarkko Johansson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Carl-Johan Boraxbekk
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Sports Medicine Copenhagen (ISMC), Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anders Wåhlin
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, Umeå, Sweden
| | - Ulman Lindenberger
- Center for Lifeorgdivision Psychology, Max Planck Institute for Human Development, Berlin, Germany
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany and London, UK
| | - Martin Lövdén
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
| | - Katrine Riklund
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Lars Bäckman
- Aging Research Center, Karolinska Institute and Stockholm University, Stockholm, Sweden
| | - Lars Nyberg
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Nina Karalija
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden.
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.
| |
Collapse
|
2
|
Chen M, Yuan L, Chen B, Chang H, Luo J, Zhang H, Chen Z, Kong J, Yi Y, Bai M, Dong M, Zhou H, Jiang H. SLC29A1 and SLC29A2 are human nicotinamide cell membrane transporters. Nat Commun 2025; 16:1181. [PMID: 39885119 DOI: 10.1038/s41467-025-56402-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Nicotinamide (NAM), a main precursor of NAD+, is essential for cellular fuel respiration, energy production, and other cellular processes. Transporters for other precursors of NAD+ such as nicotinic acid and nicotinamide mononucleotide (NMN) have been identified, but the cellular transporter of nicotinamide has not been elucidated. Here, we demonstrate that equilibrative nucleoside transporter 1 and 2 (ENT1 and 2, encoded by SLC29A1 and 2) drive cellular nicotinamide uptake and establish nicotinamide metabolism homeostasis. In addition, ENT1/2 exhibits a strong capacity to change the cellular metabolite composition and the transcript, especially those related to nicotinamide. We further observe that ENT1/2 regulates cellular respiration and senescence, contributing by altering the NAD+ pool level and mitochondrial status. Changes to cellular respiration, mitochondrial status and senescence by ENT1/2 knockdown are reversed by NMN supplementation. Together, ENT1 and ENT2 act as both cellular nicotinamide-level keepers and nicotinamide biological regulators through their NAM transport functions.
Collapse
Affiliation(s)
- Mingyang Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Luexiang Yuan
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hui Chang
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Luo
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hengbin Zhang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Zhongjian Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, China
| | - Jiao Kong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Yaodong Yi
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Mengru Bai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Minlei Dong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Huidi Jiang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| |
Collapse
|
3
|
Baker C, Kim M, Benayoun BA. A researcher's guide to studying sex differences in immune aging. Trends Mol Med 2025:S1471-4914(25)00005-X. [PMID: 39884873 DOI: 10.1016/j.molmed.2025.01.005] [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: 10/03/2024] [Revised: 12/11/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Sex differences in immune system aging significantly impact disease susceptibility and vaccine responses among older adults, but with notable disparities between men and women. This area has gained importance because vaccines can exhibit differential efficacy by sex in aging populations, underscoring the need for sex-specific strategies. As the global population ages, understanding these sex-based immune differences is crucial for developing targeted interventions for age-related diseases. Addressing these disparities requires robust preclinical models that mimic human immune aging to uncover mechanisms and inform personalized approaches. In this review we assess the translational potential of preclinical mouse models in studying sex differences in immune aging, and emphasize the urgency of sex-specific interventions to improve health outcomes in older adults.
Collapse
Affiliation(s)
- Clayton Baker
- Leonard Davis School of Gerontology, University of Southern California (USC), Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA
| | - Minhoo Kim
- Leonard Davis School of Gerontology, University of Southern California (USC), Los Angeles, CA 90089, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California (USC), Los Angeles, CA 90089, USA; Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA 90089, USA; Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA 90089, USA; Epigenetics and Gene Regulation, USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA; USC Stem Cell Initiative, Los Angeles, CA 90089, USA.
| |
Collapse
|
4
|
Li X, Hou M, Zhang F, Ji Z, Cai Y, Shi Y. Per- and Polyfluoroalkyl Substances and Female Health Concern: Gender-based Accumulation Differences, Adverse Outcomes, and Mechanisms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1469-1486. [PMID: 39803974 DOI: 10.1021/acs.est.4c08701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The deleterious health implications of perfluoroalkyl and polyfluoroalkyl substances (PFAS) are widely recognized. Females, in contrast to males, exhibit unique pathways for PFAS exposure and excretion, leading to complex health outcomes. The health status of females is largely influenced by hormone-related processes. PFAS have been reported to be associated with various aspects of female health, including reproductive system disorders and pregnancy-related diseases. In this article, we provide insights into the correlations between PFAS and female-prevalent diseases. Current epidemiological and toxicological evidence has demonstrated that the adverse effects of PFAS on the health of the female reproductive system are primarily attributed to the disruption of the hypothalamic-pituitary-gonadal (HPG) axis and hormonal homeostasis. However, these findings do not sufficiently elucidate the intricate associations between PFAS and specific diseases. Furthermore, autoimmune disorders, another category that is more prevalent in women compared to men, require additional investigation. Immune biomarkers pertinent to autoimmune disorders have been observed to be influenced by PFAS exposure, although epidemiological evidence is insufficient to substantiate these relations. Further thorough exploration encompassing epidemiological and toxicological studies is essential to elucidating the inherent influence of PFAS on human pathologies. Additionally, comprehensive investigations into female health issues beyond their reproductive functions is essential.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minmin Hou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Zhang
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Zhengquan Ji
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Yaqi Cai
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yali Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Riemann L, Gutierrez R, Odak I, Barros-Martins J, Roesner LM, Leon Lara X, Falk C, Schulz TF, Hansen G, Werfel T, Förster R. Integrative deep immune profiling of the elderly reveals systems-level signatures of aging, sex, smoking, and clinical traits. EBioMedicine 2025; 112:105558. [PMID: 39862806 DOI: 10.1016/j.ebiom.2025.105558] [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: 10/07/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Aging increases disease susceptibility and reduces vaccine responsiveness, highlighting the need to better understand the aging immune system and its clinical associations. Studying the human immune system, however, remains challenging due to its complexity and significant inter-individual variability. METHODS We conducted an immune profiling study of 550 elderly participants (≥60 years) and 100 young controls (20-40 years) from the RESIST Senior Individuals (SI) cohort. Extensive demographic, clinical, and laboratory data were collected. Multi-color spectral flow cytometry and 48-plex plasma cytokine assays were used for deep immune phenotyping. Data were analyzed using unsupervised clustering and multi-dataset integration approaches. FINDINGS We studied 97 innate and adaptive immune cell populations, revealing intricate age- and sex-related changes in the elderly immune system. Our large sample size allowed detection of even subtle changes in cytokines and immune cell clusters. Integrative analysis combining clinical, laboratory, and immunological data revealed systems-level aging signatures, including shifts in specific immune cell subpopulations and cytokine concentrations (e.g., HGF and CCL27). Additionally, we identified unique immune signatures associated with smoking, obesity, and diseases such as osteoporosis, heart failure, and gout. INTERPRETATION This study provides one of the most comprehensive immune profiles of elderly individuals, uncovering high-resolution immune changes associated with aging. Our findings highlight clinically relevant immune signatures that enhance our understanding of aging-related diseases and could guide future research into new treatments, offering translational insights into human health and aging. FUNDING Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy-EXC 2155-project number 390874280.
Collapse
Affiliation(s)
- Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
| | | | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany; The Tisch Cancer Institute, Icahn School of Medicine, New York, USA
| | - Joana Barros-Martins
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Microbiology and Immunology, Columbia University Medical Center, New York, USA
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Ximena Leon Lara
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine Falk
- Institute of Transplantation Immunology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Thomas F Schulz
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Center of Lung Research (DZL), BREATH, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany.
| |
Collapse
|
6
|
Yakovenko I, Mihai IS, Selinger M, Rosenbaum W, Dernstedt A, Gröning R, Trygg J, Carroll L, Forsell M, Henriksson J. Telomemore enables single-cell analysis of cell cycle and chromatin condensation. Nucleic Acids Res 2025; 53:gkaf031. [PMID: 39878215 PMCID: PMC11775621 DOI: 10.1093/nar/gkaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/15/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Single-cell RNA-seq methods can be used to delineate cell types and states at unprecedented resolution but do little to explain why certain genes are expressed. Single-cell ATAC-seq and multiome (ATAC + RNA) have emerged to give a complementary view of the cell state. It is however unclear what additional information can be extracted from ATAC-seq data besides transcription factor binding sites. Here, we show that ATAC-seq telomere-like reads counter-inituively cannot be used to infer telomere length, as they mostly originate from the subtelomere, but can be used as a biomarker for chromatin condensation. Using long-read sequencing, we further show that modern hyperactive Tn5 does not duplicate 9 bp of its target sequence, contrary to common belief. We provide a new tool, Telomemore, which can quantify nonaligning subtelomeric reads. By analyzing several public datasets and generating new multiome fibroblast and B-cell atlases, we show how this new readout can aid single-cell data interpretation. We show how drivers of condensation processes can be inferred, and how it complements common RNA-seq-based cell cycle inference, which fails for monocytes. Telomemore-based analysis of the condensation state is thus a valuable complement to the single-cell analysis toolbox.
Collapse
Affiliation(s)
- Iryna Yakovenko
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Ionut Sebastian Mihai
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Industrial Doctoral School, Umeå University, Umeå, Sweden
| | - Martin Selinger
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Department of Chemistry, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - William Rosenbaum
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Andy Dernstedt
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Remigius Gröning
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Johan Trygg
- Department of Chemistry, Umeå University, Linnaeus väg 10, Umeå universitet, 901 87, Umeå, Sweden
- Sartorius Corporate Research, Östra Strandgatan 24, 903 33, Umeå, Sweden
| | - Laura Carroll
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Integrated Science Lab (IceLab), Umeå University, Naturvetarhuset, Universitetsvägen, 901 87, Umeå, Sweden
| | - Mattias Forsell
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Johan Henriksson
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Integrated Science Lab (IceLab), Umeå University, Naturvetarhuset, Universitetsvägen, 901 87, Umeå, Sweden
| |
Collapse
|
7
|
Amodio G, Giacomini G, Boeri L, Raffo M, Cilio S, Pozzi E, Belladelli F, Negri F, Ferrara AM, d'Arma A, Santoni de Sio FR, Pagliardini L, Papaleo E, Ventimiglia E, Alfano M, Montorsi F, Salonia A, Gregori S. Specific types of male infertility are correlated with T cell exhaustion or senescence signatures. Nat Commun 2025; 16:971. [PMID: 39856063 PMCID: PMC11759947 DOI: 10.1038/s41467-025-56193-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The association between male infertility and health status has yet to be unraveled. Here, by combining multiparameter phenotyping and scRNA-seq, we delineate the immune status of infertile men both at the semen and systemic levels. We first observe that young infertile men have a pro-inflammatory milieu with increased frequency of myeloid cells and inflammatory mediators in the seminal fluid and the peripheral blood, which are immune alterations typically observed in healthy elderly men. Transcriptomic profiling confirms the upregulation of genes associated with the interferon-gamma and -alpha responses in peripheral blood T cells of infertile men with oligo-astheno-teratozoospermia or non-obstructive azoospermia, with distinct T cell signatures of exhaustion and senescence discriminating the two infertile conditions. These findings provide evidence that subtypes of male infertility are characterized by specific immune signatures and unravel the potential link between infertility and the risk of developing secondary diseases.
Collapse
Affiliation(s)
- Giada Amodio
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giorgia Giacomini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Boeri
- IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Department of Urology, Milan, Italy
| | - Massimiliano Raffo
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Simone Cilio
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, Urology Unit, University of Naples "Federico II", Naples, Italy
| | - Edoardo Pozzi
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Federico Belladelli
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Fausto Negri
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Anna Maria Ferrara
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia d'Arma
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Luca Pagliardini
- Reproductive Sciences Laboratory, Obstetrics and Gynaecology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Enrico Papaleo
- Reproductive Sciences Laboratory, Obstetrics and Gynaecology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Eugenio Ventimiglia
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy.
- University Vita-Salute San Raffaele, Milan, Italy.
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
8
|
Zhang C, Hao T, Bortoluzzi A, Chen MH, Wu X, Wang J, Ermel R, Kim Y, Chen S, Chen W. Sex-dependent differences in hematopoietic stem cell aging and leukemogenic potential. Oncogene 2025; 44:64-78. [PMID: 39487323 PMCID: PMC11706783 DOI: 10.1038/s41388-024-03197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Sex influences many biological outcomes, but how sex affects hematopoietic stem cell (HSC) aging and hematological disorders is poorly understood. The widespread use of young animal models to study age-related diseases further complicates these matters. Using aged and long-lived BALB/c mouse models, we discovered that aging mice exhibit sex-dependent disparities, mirroring aging humans, in developing myeloid skewing, anemia, and leukemia. These disparities are underlined by sex-differentiated HSC aging characteristics across the population, single-cell, and molecular levels. The HSC population expanded significantly with aging and longevity in males, but this occurred to a much lesser degree in aging females that instead expanded committed progenitors. Aging male HSCs are more susceptible to BCR-ABL1 transformation with faster development of chronic myeloid leukemia (CML) than female HSCs. Additionally, the loss of the aging regulator Sirt1 inhibited CML development in aging male but not female mice. Our results showed for the first time that sex-differentiated HSC aging impacts hematopoiesis, leukemogenesis, and certain gene functions. This discovery provides insights into understanding age-dependent hematological diseases and sex-targeted strategies for the treatment and prevention of certain blood disorders and cancer.
Collapse
MESH Headings
- Animals
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Female
- Male
- Mice
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Cellular Senescence
- Aging/pathology
- Aging/physiology
- Mice, Inbred BALB C
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- Hematopoiesis
- Sex Characteristics
- Humans
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Sex Factors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
Collapse
Affiliation(s)
- Chunxiao Zhang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Amgen, Thousand Oaks, CA, USA
| | - Taisen Hao
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Bristol Myers Squibb, Seattle, WA, USA
| | - Alessia Bortoluzzi
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Min-Hsuan Chen
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Jinhui Wang
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Richard Ermel
- Center for Comparative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Young Kim
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Shiuan Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - WenYong Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
9
|
Angarola BL, Sharma S, Katiyar N, Kang HG, Nehar-Belaid D, Park S, Gott R, Eryilmaz GN, LaBarge MA, Palucka K, Chuang JH, Korstanje R, Ucar D, Anczukόw O. Comprehensive single-cell aging atlas of healthy mammary tissues reveals shared epigenomic and transcriptomic signatures of aging and cancer. NATURE AGING 2025; 5:122-143. [PMID: 39587369 PMCID: PMC11754115 DOI: 10.1038/s43587-024-00751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/16/2024] [Indexed: 11/27/2024]
Abstract
Aging is the greatest risk factor for breast cancer; however, how age-related cellular and molecular events impact cancer initiation is unknown. In this study, we investigated how aging rewires transcriptomic and epigenomic programs of mouse mammary glands at single-cell resolution, yielding a comprehensive resource for aging and cancer biology. Aged epithelial cells exhibit epigenetic and transcriptional changes in metabolic, pro-inflammatory and cancer-associated genes. Aged stromal cells downregulate fibroblast marker genes and upregulate markers of senescence and cancer-associated fibroblasts. Among immune cells, distinct T cell subsets (Gzmk+, memory CD4+, γδ) and M2-like macrophages expand with age. Spatial transcriptomics reveals co-localization of aged immune and epithelial cells in situ. Lastly, we found transcriptional signatures of aging mammary cells in human breast tumors, suggesting possible links between aging and cancer. Together, these data uncover that epithelial, immune and stromal cells shift in proportions and cell identity, potentially impacting cell plasticity, aged microenvironment and neoplasia risk.
Collapse
Affiliation(s)
| | | | - Neerja Katiyar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hyeon Gu Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Giray N Eryilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| | - Olga Anczukόw
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| |
Collapse
|
10
|
Pangrazzi L, Meryk A. Molecular and Cellular Mechanisms of Immunosenescence: Modulation Through Interventions and Lifestyle Changes. BIOLOGY 2024; 14:17. [PMID: 39857248 PMCID: PMC11760833 DOI: 10.3390/biology14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Immunosenescence, the age-related decline in immune function, is a complex biological process with profound implications for health and longevity. This phenomenon, characterized by alterations in both innate and adaptive immunity, increases susceptibility to infections, reduces vaccine efficacy, and contributes to the development of age-related diseases. At the cellular level, immunosenescence manifests as decreased production of naive T and B cells, accumulation of memory and senescent cells, thymic involution, and dysregulated cytokine production. Recent advances in molecular biology have shed light on the underlying mechanisms of immunosenescence, including telomere attrition, epigenetic alterations, mitochondrial dysfunction, and changes in key signaling pathways such as NF-κB and mTOR. These molecular changes lead to functional impairments in various immune cell types, altering their proliferative capacity, differentiation, and effector functions. Emerging research suggests that lifestyle factors may modulate the rate and extent of immunosenescence at both cellular and molecular levels. Physical activity, nutrition, stress management, and sleep patterns have been shown to influence immune cell function, inflammatory markers, and oxidative stress in older adults. This review provides a comprehensive analysis of the molecular and cellular mechanisms underlying immunosenescence and explores how lifestyle interventions may impact these processes. We will examine the current understanding of immunosenescence at the genomic, epigenomic, and proteomic levels, and discuss how various lifestyle factors can potentially mitigate or partially reverse aspects of immune aging. By integrating recent findings from immunology, gerontology, and molecular biology, we aim to elucidate the intricate interplay between lifestyle and immune aging at the molecular level, potentially informing future strategies for maintaining immune competence in aging populations.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Institute for Biomedical Aging Research, Faculty of Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
11
|
Chen F, Chen Y, Liang H, Pan X, Wang Y, Shen L, Hu Y. Non-linear effects of age in reporting of adverse events following influenza immunization in Zhejiang, China. BMC Infect Dis 2024; 24:1457. [PMID: 39716104 DOI: 10.1186/s12879-024-10385-1] [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: 12/18/2023] [Accepted: 12/20/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Previous evidence had suggested age and sex affect the reporting rate of adverse events following immunization (AEFI), but with little exploration of potential their non-linear and interaction effects on AEFIs. Examining these non-linear effects could be beneficial for identifying high-risk populations. METHODS Using AEFI records and vaccination data from national passive surveillance system of adverse event following immunization and Zhejiang provincial immunization information system in the 2021-2022 influenza season, respectively. The effects of age and sex on AEFIs were analyzed through the generalized additive model (logistic regression with a smooth term) to estimate non-linear characteristics after adjusting for other co-variables (adopted significance level p < 0.05). RESULTS There were 1,259,975 influenza vaccine doses administered and 1304 AEFI records reported during the 2021-2022 influenza season, with a reporting rate of 10.35/10,000 doses. The odds of reporting an AEFI increased from 6 months of age, peaking at about 54 years of age, then gradually declined. The odds of females experiencing AEFIs are higher than that of males. The data model indicated clear effects of age, sex, and their interaction (p < 0.01) on reporting rate of AEFI. Concomitant vaccination and vaccine type were also the impact factors for reporting rate of AEFI. CONCLUSION This study revealed a non-linear property in age and the AEFI odds, with a significant interaction and higher reporting rate in females. In addition, the odds of AEFI increased with co-administration compared to separate vaccination.
Collapse
Affiliation(s)
- Fuxing Chen
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Yaping Chen
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Hui Liang
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Xuejiao Pan
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Ying Wang
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Lingzhi Shen
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China
| | - Yu Hu
- Institute of Immunization and Prevention, Zhejiang Center for Disease Control and Prevention, No. 3399 Binsheng Road, Binjiang District, Hangzhou, P.R. China.
| |
Collapse
|
12
|
Accardi G, Calabrò A, Caldarella R, Caruso C, Ciaccio M, Di Simone M, Ligotti ME, Meraviglia S, Zarcone R, Candore G, Aiello A. Immune-Inflammatory Response in Lifespan-What Role Does It Play in Extreme Longevity? A Sicilian Semi- and Supercentenarians Study. BIOLOGY 2024; 13:1010. [PMID: 39765677 PMCID: PMC11673833 DOI: 10.3390/biology13121010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025]
Abstract
Studying models of healthy aging and exceptional longevity is crucial to understanding a possible longevity signature, as most show resistance to age-related diseases. In particular, semi- and supercentenarians are a highly selected group, having survived significant adversities, including the Spanish flu and COVID-19 pandemics, indicating distinctive immune system characteristics. This paper analyzes the inflammatory scores (INFLA-score, Systemic Inflammation Response Index (SIRI)) and Aging-Related Immune Phenotype (ARIP) indicators calculated from the dataset of the DESIGN project, including 249 participants aged 19-111 years, aiming to understand the immune-inflammatory (IMFLAM) role in achieving longevity. Statistical analyses, including linear regression and one-way ANOVA, were performed to explore the correlations between these parameters and age. Both INFLA-score and SIRI showed a significant increase with age. However, no statistical differences were found when comparing the values of semi- and supercentenarians to other age groups, which are similar to adults and lower than younger centenarians. Regarding ARIP values, it is noteworthy that when comparing the CD8+ Naïve/Effector scores between groups, no significant differences were observed between the semi- and supercentenarian group and the other groups. These results support the idea that the control of IMFLAM response can promote extreme longevity.
Collapse
Affiliation(s)
- Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
| | - Rosalia Caldarella
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (R.C.); (M.C.)
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
| | - Marcello Ciaccio
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (R.C.); (M.C.)
- Section of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (M.D.S.); (S.M.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Mattia Emanuela Ligotti
- ISMETT-IRCCS Mediterranean Institute for Transplants and Highly Specialised Therapies, 90127 Palermo, Italy;
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (M.D.S.); (S.M.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Rosa Zarcone
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (R.C.); (M.C.)
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy; (G.A.); (A.C.); (R.Z.); (A.A.)
| |
Collapse
|
13
|
Menezes AA, Shah ZA. A Review of the Consequences of Gut Microbiota in Neurodegenerative Disorders and Aging. Brain Sci 2024; 14:1224. [PMID: 39766423 PMCID: PMC11726757 DOI: 10.3390/brainsci14121224] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/15/2025] Open
Abstract
Age-associated alterations in the brain lead to cognitive deterioration and neurodegenerative disorders (NDDs). This review with a particular focus on Alzheimer's disease (AD), emphasizes the burgeoning significance of the gut microbiota (GMB) in neuroinflammation and its impact on the gut-brain axis (GBA), a communication conduit between the gut and the central nervous system (CNS). Changes in the gut microbiome, including diminished microbial diversity and the prevalence of pro-inflammatory bacteria, are associated with AD pathogenesis. Promising therapies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, may restore gut health and enhance cognitive performance. Clinical data remain insufficient, necessitating further research to elucidate causes, enhance therapy, and consider individual variances. This integrative approach may yield innovative therapies aimed at the GMB to improve cognitive function and brain health in older people.
Collapse
Affiliation(s)
| | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
14
|
Chen L, Wang D, Liu W, Zhou S, Gu Q, Zhou T. Immunomodulation of exopolysaccharide produced by Lacticaseibacillus rhamnosus ZFM216 in cyclophosphamide-induced immunosuppressed mice by modulating gut microbiota. Int J Biol Macromol 2024; 283:137619. [PMID: 39551322 DOI: 10.1016/j.ijbiomac.2024.137619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
This study investigated the immunoregulatory activity of exopolysaccharides (EPS) produced by Lacticaseibacillus rhamnosus ZFM216 in immunosuppressed mice induced by cyclophosphamide (CTX). The results showed that EPS treatment effectively improved the body weight, immune organ index and splenic lymphocyte proliferation. EPS also mitigated the damage of immune organs, restored intestinal morphology, and regulated the levels of serum hemolysin and cytokines (e.g. TNF-α, INF-γ and IL-10). EPS promoted the release of NO, TNF-α, IL-1β, and IL-6 in RAW 264.7 cells, however, such effect was inhibited in the presence of inhibitors of TLR4 and MAPKs signaling pathways-related proteins, confirming that EPS achieved the immunomodulation by activating these two signaling pathways. Additionally, EPS, as a prebiotic, effectively improved the diversity of microbial communities, regulated the relative abundance of dominant microbial communities, restored CTX-induced gut microbiota dysbiosis, and promoted the production of short chain fatty acids (SCFAs) in the gut of mice. Thus, immunoregulatory effect of EPS could be attributed to its good ability to modulate the gut microbiota. EPS produced by L. rhamnosus ZFM216 has promising application as an ingredient of functional foods due to its potent probiotic effect.
Collapse
Affiliation(s)
- Liang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Xiasha, Hangzhou, Zhejiang, 310018, PR China
| | - Dong Wang
- Zhejiang Chemtrue Bio-Pharm Co., Ltd. Xiasha, Hangzhou, Zhejiang, 310018, PR China
| | - Wei Liu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, PR China
| | - Shaobo Zhou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Xiasha, Hangzhou, Zhejiang, 310018, PR China; School of Science, Faculty of Engineering and Science, University of Greenwich, Central Avenue, Chatham ME4 4TB, UK
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Xiasha, Hangzhou, Zhejiang, 310018, PR China
| | - Tao Zhou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Xiasha, Hangzhou, Zhejiang, 310018, PR China.
| |
Collapse
|
15
|
Arosio B, Picca A. The biological roots of the sex-frailty paradox. Exp Gerontol 2024; 198:112619. [PMID: 39490699 DOI: 10.1016/j.exger.2024.112619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aging is a dynamic process that requires a continuous response and adaptation to internal and external stimuli over the life course. This eventually results in people aging differently and women aging differently than men. The "gender paradox" describes how women experience greater longevity than men, although linked with higher rates of disability and poor health status. Recently, the concept of frailty has been incorporated into this paradox giving rise to the "sex-frailty paradox" which describes how women are frailer because they manifest worse health status but, at the same time, appear less susceptible to death than men of the same age. However, very little is known about the biological roots of this sex-related difference in frailty. Inflamm-aging, the chronic low-grade inflammatory state associated with age, plays a key pathophysiological role in several age-related diseases/conditions, including Alzheimer's disease (AD), for which women have a higher lifetime risk than men. Interestingly, inflamm-aging develops at a different rate in women compared to men, with features that could play a critical role in the development of AD in women. According to this view, a continuum between aging and age-related diseases that probably lacks clear boundaries can be envisioned in which several shared biological mechanisms that progress at different pace may lead to different aging trajectories in women than in men. It, therefore, becomes urgent to consider a holistic approach in the study of aging, and decline it from a gender medicine perspective also considering the biological roots of the sex-frailty paradox.
Collapse
Affiliation(s)
- Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
16
|
Jin Z, Liu X, Guo H, Chen S, Zhu X, Pan S, Wu Y. Sex-specific modulating role of social support in the associations between oxidative stress, inflammation, and telomere length in older adults. J Behav Med 2024; 47:1040-1051. [PMID: 39179728 DOI: 10.1007/s10865-024-00515-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
Telomere length, a biomarker of human aging, is related to adverse health outcomes. Growing evidence indicates that oxidative stress and inflammation contributes to telomere shortening, whereas social support may protect from telomere shortening. Despite sex differences in telomere length and social support, little is known about whether there are sex differences in the relationship between oxidative stress/inflammation and telomere length, and sex-specific moderating roles of social support in older adults. Using data from the National Health and Nutrition Examination Survey (NHANES) 1999-2002, this study assessed whether the associations between oxidative stress/inflammation and telomere length vary with sex and explored social support as a moderator in these associations among 2289 older adults. Oxidative stress was measured based on serum Gamma-glutamyl transferase (GGT), and inflammation was measured based on C-reactive protein (CRP). After adjusting for the covariates, GGT was significantly associated with telomere length in females only (β = - 0.037, 95% CI = - 0.070, - 0.005), while CRP was associated with telomere length in males only (β = - 0.019, 95% CI = - 0.035, - 0.002). Moreover, high social support mitigated the negative association between GGT and telomere length, which was more evident in females. Furthermore, social support moderated the association between CRP and telomere length in males aged 70 and above. Our findings indicated that biological mechanisms related to telomere length may vary with sex, while social support plays a sex-specific moderating role.
Collapse
Affiliation(s)
- Zhou Jin
- Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, School of Mental Health and The Affiliated Wenzhou Kangning Hospital, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xuejian Liu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, School of Mental Health and The Affiliated Wenzhou Kangning Hospital, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Haonan Guo
- Department of Sociology, Faculty of Social Science, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Sixuan Chen
- Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, School of Mental Health and The Affiliated Wenzhou Kangning Hospital, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xianghe Zhu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, School of Mental Health and The Affiliated Wenzhou Kangning Hospital, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Sipei Pan
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yili Wu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, School of Mental Health and The Affiliated Wenzhou Kangning Hospital, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
17
|
Touil H, Luquez T, Comandante-Lou N, Lee AJ, Fujita M, Habeck C, Kroshilina A, Hegewisch-Solloa E, McInvale J, Zuroff L, Isnard S, Walker E, Zhang L, Routy JP, Zhang Y, Mace EM, Klotz L, Wiendl H, Xia Z, Bar-Or A, Menon V, Stern Y, De Jager PL. Relation of CMV and brain atrophy to trajectories of immunosenescence in diverse populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.614568. [PMID: 39416188 PMCID: PMC11482892 DOI: 10.1101/2024.10.07.614568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Immunosenescence (ISC), the aging of the immune system, has largely been studied in populations of European descent. Here, circulating immune cell cytometric data from African-American, Hispanic, and non-Hispanic White participants were generated. Known and novel age effects were identified using either a meta-analysis approach or a parallel genetic approach. Most results are consistent across the three populations, but some cell populations display evidence of heterogeneity, such as a PD-L1 + CD56 + NK cell subset. The study estimated "Immunological Age" (IA) during physiologic aging. While we found no relation of IA to Multiple Sclerosis, IA is associated with entorhinal cortex atrophy, a presymptomatic feature of Alzheimer's disease, linking neurodegeneration and peripheral immunity. ISC trajectories were also inferred, highlighting age, CMV status, and genetic ancestry as key influences. Our assessment offers reference ISC trajectories for personalization of assessments of immune function over the life course in diverse populations.
Collapse
|
18
|
Shen X, Wang C, Zhou X, Zhou W, Hornburg D, Wu S, Snyder MP. Nonlinear dynamics of multi-omics profiles during human aging. NATURE AGING 2024; 4:1619-1634. [PMID: 39143318 PMCID: PMC11564093 DOI: 10.1038/s43587-024-00692-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
Aging is a complex process associated with nearly all diseases. Understanding the molecular changes underlying aging and identifying therapeutic targets for aging-related diseases are crucial for increasing healthspan. Although many studies have explored linear changes during aging, the prevalence of aging-related diseases and mortality risk accelerates after specific time points, indicating the importance of studying nonlinear molecular changes. In this study, we performed comprehensive multi-omics profiling on a longitudinal human cohort of 108 participants, aged between 25 years and 75 years. The participants resided in California, United States, and were tracked for a median period of 1.7 years, with a maximum follow-up duration of 6.8 years. The analysis revealed consistent nonlinear patterns in molecular markers of aging, with substantial dysregulation occurring at two major periods occurring at approximately 44 years and 60 years of chronological age. Distinct molecules and functional pathways associated with these periods were also identified, such as immune regulation and carbohydrate metabolism that shifted during the 60-year transition and cardiovascular disease, lipid and alcohol metabolism changes at the 40-year transition. Overall, this research demonstrates that functions and risks of aging-related diseases change nonlinearly across the human lifespan and provides insights into the molecular and biological pathways involved in these changes.
Collapse
Affiliation(s)
- Xiaotao Shen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Singapore, Singapore
| | - Chuchu Wang
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Xin Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, USA
| | - Wenyu Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Si Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, USA.
| |
Collapse
|
19
|
Chang ST, Chuang YF, Li AH, Fan YT, Liao MR, Chen IY, Hung RW, Yang TO, Chiu YL. Age-dependent immune profile in healthy individuals: an original study, systematic review and meta-analysis. Immun Ageing 2024; 21:75. [PMID: 39472926 PMCID: PMC11520839 DOI: 10.1186/s12979-024-00480-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND The circulatory peripheral immune system is the most convenient approach for determining an individual's immune status. Due to various reasons, while previous studies have addressed the critical impact of age, most individual studies did not analyze immunosenescence in a systemic manner, which complicates the possibility of building a reference range for age-dependent immune profiles for effective immune monitoring. To address this gap, this study analyzed a group of healthy individuals to establish age-specific reference ranges of the healthy circulatory immune profile, and a systematic review and meta-analysis were conducted to validate the findings and create generalizable immune cell reference ranges. RESULTS Our study recruited a total of 363 healthy Taiwanese adults (median age 42 years [IQR 30, 62], age range 21 to 87 years, 43.3% male), including 158 under 40 years old, 127 between 40-64 years old, and 78 over 64 years old. Significant age-related alterations were observed in both adaptive and innate immune cell subsets. CD8 + T cells decreased and CD4/CD8 ratio increased, with notable increases in NK cells. CD4 + T cells were less impacted by aging, while CD8 + T cells significantly lost CD28 and increased CD31 expression with age. A clear reverse trend in naïve and memory subsets of CD4 + and CD8 + T cells was observed. Detailed reference ranges for immune cell subsets in healthy Taiwanese adults were established. A systematic review included 7,425 adults and a meta-analysis of 12 eligible studies confirmed our findings in Taiwan, enhancing generalizability. CONCLUSIONS Combined with previous studies and original data through a systematic review and meta-analysis, we highlighted and quantified significant immune profile differences between older and younger individuals. The sex and age-specific reference ranges for peripheral immune cell subsets can serve as a basis for effective immune monitoring of various aging-related illnesses.
Collapse
Affiliation(s)
- Syuan-Ting Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Fang Chuang
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Ai-Hsien Li
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
- Health Management Center, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Yang-Teng Fan
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| | - Man-Ru Liao
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
- Health Management Center, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - I-Yu Chen
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Ruo-Wei Hung
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Tienyu Owen Yang
- Science Officer, Cancer Epidemiology Unit, Nuffield, Department of Population Health , University of Oxford, Richard Doll Building, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Yen-Ling Chiu
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei, Taiwan.
| |
Collapse
|
20
|
Chhabra Y, Fane ME, Pramod S, Hüser L, Zabransky DJ, Wang V, Dixit A, Zhao R, Kumah E, Brezka ML, Truskowski K, Nandi A, Marino-Bravante GE, Carey AE, Gour N, Maranto DA, Rocha MR, Harper EI, Ruiz J, Lipson EJ, Jaffee EM, Bibee K, Sunshine JC, Ji H, Weeraratna AT. Sex-dependent effects in the aged melanoma tumor microenvironment influence invasion and resistance to targeted therapy. Cell 2024; 187:6016-6034.e25. [PMID: 39243764 PMCID: PMC11580838 DOI: 10.1016/j.cell.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
There is documented sex disparity in cutaneous melanoma incidence and mortality, increasing disproportionately with age and in the male sex. However, the underlying mechanisms remain unclear. While biological sex differences and inherent immune response variability have been assessed in tumor cells, the role of the tumor-surrounding microenvironment, contextually in aging, has been overlooked. Here, we show that skin fibroblasts undergo age-mediated, sex-dependent changes in their proliferation, senescence, ROS levels, and stress response. We find that aged male fibroblasts selectively drive an invasive, therapy-resistant phenotype in melanoma cells and promote metastasis in aged male mice by increasing AXL expression. Intrinsic aging in male fibroblasts mediated by EZH2 decline increases BMP2 secretion, which in turn drives the slower-cycling, highly invasive, and therapy-resistant melanoma cell phenotype, characteristic of the aged male TME. Inhibition of BMP2 activity blocks the emergence of invasive phenotypes and sensitizes melanoma cells to BRAF/MEK inhibition.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Mitchell E Fane
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sneha Pramod
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laura Hüser
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Daniel J Zabransky
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Agrani Dixit
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ruzhang Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Edwin Kumah
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Megan L Brezka
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kevin Truskowski
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Asmita Nandi
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gloria E Marino-Bravante
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Naina Gour
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Devon A Maranto
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Murilo R Rocha
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth I Harper
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Justin Ruiz
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Evan J Lipson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA; The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kristin Bibee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joel C Sunshine
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Di Rosa M, Sabbatinelli J, Giuliani A, Carella M, Magro D, Biscetti L, Soraci L, Spannella F, Fedecostante M, Lenci F, Tortato E, Pimpini L, Burattini M, Cecchini S, Cherubini A, Bonfigli AR, Capalbo M, Procopio AD, Balistreri CR, Olivieri F. Inflammation scores based on C-reactive protein and albumin predict mortality in hospitalized older patients independent of the admission diagnosis. Immun Ageing 2024; 21:67. [PMID: 39385197 PMCID: PMC11463076 DOI: 10.1186/s12979-024-00471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Systemic inflammation significantly increases the risk of short- and long-term mortality in geriatric hospitalized patients. To predict mortality in older patients with various age-related diseases and infections, including COVID-19, inflammatory biomarkers such as the C-reactive protein (CRP) to albumin ratio (CAR), and related scores and indexes, i.e. Glasgow Prognostic Score (GPS), modified GPS (mGPS), and high sensitivity (hs)-mGPS, have been increasingly utilized. Despite their easy affordability and widespread availability, these biomarkers are predominantly assessed for clinical purposes rather than predictive applications, leading to their underutilization in hospitalized older patients. In this study, we investigated the association of CAR, GPS, mGPS, and hs-mGPS with short-term mortality in 3,206 geriatric hospitalized patients admitted for acute conditions, irrespective of admission diagnosis. We observed that unit increases of CAR, and the highest classes of GPS, mGPS, and hs-mGPS were significantly associated with a two- to threefold increased risk of death, even adjusting the risk for different confounding variables. Interestingly, a hs-mGPS of 2 showed the highest effect size. Furthermore, gender analysis indicated a stronger association between all CRP-albumin based parameters and mortality in men, underscoring the gender-specific relevance of inflammation-based circulating parameters in mortality prediction. In conclusion, scores based on serum CRP and albumin levels offer additional guidance for the stratification of in-hospital mortality risk in older patients by providing additional information on the degree of systemic inflammation.
Collapse
Affiliation(s)
- Mirko Di Rosa
- Centre for Biostatistics and Applied Geriatric Clinical Epidemiology, IRCCS INRCA, Ancona, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy.
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy.
| | - Angelica Giuliani
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Bari Institute, Bari, Italy
| | - Miriam Carella
- Complex Operative Unit of Clinical Pathology, ARNAS Civico Di Cristina e Benfratelli Hospitals, Palermo, Italy
| | - Daniele Magro
- Cellular, Molecular and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | | | - Luca Soraci
- Unit of Geriatric Medicine, IRCCS INRCA, Cosenza, Italy
| | - Francesco Spannella
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Internal Medicine and Geriatrics, IRCCS INRCA, Ancona, Italy
| | - Massimiliano Fedecostante
- Geriatria, Accettazione Geriatrica e Centro Di Ricerca Per L'invecchiamento, IRCCS INRCA, Ancona, Italy
| | - Federica Lenci
- Unit of Nephrology and Dialysis, IRCCS INRCA, Ancona, Italy
| | | | | | | | - Sara Cecchini
- Diagnostic Imaging, Clinical and Interventional Radiology, IRCCS INRCA, Osimo, Italy
| | - Antonio Cherubini
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Geriatria, Accettazione Geriatrica e Centro Di Ricerca Per L'invecchiamento, IRCCS INRCA, Ancona, Italy
| | | | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Carmela Rita Balistreri
- Cellular, Molecular and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
22
|
Harris RM, Whitfield T, Blanton LV, Skaletsky H, Blumen K, Hyland P, McDermott E, Summers K, Hughes JF, Jackson E, Teglas P, Liu B, Chan YM, Page DC. Independent effects of testosterone, estradiol, and sex chromosomes on gene expression in immune cells of trans- and cisgender individuals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617275. [PMID: 39416170 PMCID: PMC11482753 DOI: 10.1101/2024.10.08.617275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The origins of sex differences in human disease are elusive, in part because of difficulties in separating the effects of sex hormones and sex chromosomes. To separate these variables, we examined gene expression in four groups of trans- or cisgender individuals: XX individuals treated with exogenous testosterone (n=21), XY treated with exogenous estradiol (n=13), untreated XX (n=20), and untreated XY (n=15). We performed single-cell RNA-sequencing of 358,426 peripheral blood mononuclear cells. Across the autosomes, 8 genes responded with a significant change in expression to testosterone, 34 to estradiol, and 32 to sex chromosome complement with no overlap between the groups. No sex-chromosomal genes responded significantly to testosterone or estradiol, but X-linked genes responded to sex chromosome complement in a remarkably stable manner across cell types. Through leveraging a four-state study design, we successfully separated the independent actions of testosterone, estradiol, and sex chromosome complement on genome-wide gene expression in humans.
Collapse
Affiliation(s)
- Rebecca M. Harris
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Whitehead Institute, Cambridge, MA 02142, USA
| | | | | | - Helen Skaletsky
- Whitehead Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | - Kai Blumen
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Phoebe Hyland
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Em McDermott
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kiana Summers
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | | | | | - Bingrun Liu
- Whitehead Institute, Cambridge, MA 02142, USA
| | - Yee-Ming Chan
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - David C. Page
- Whitehead Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
23
|
Johnson VJ, Luster MI, Maier A, Boles C, Miller EW, Arrieta DE. Application and interpretation of immunophenotyping data in safety and risk assessment. FRONTIERS IN TOXICOLOGY 2024; 6:1409365. [PMID: 39430110 PMCID: PMC11486759 DOI: 10.3389/ftox.2024.1409365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024] Open
Abstract
The use of immunophenotyping during immunotoxicity investigations was first popularized in the 1980 s and has since become more integrated into diagnostic and non-clinical assessments. The data provided from immunophenotyping can serve as an initial source of information to guide decisions for additional, more advanced, immunotoxicity testing as well as for human health safety and risk assessment of drugs and chemicals. However, comprehensive guidance describing applications of immunophenotyping data in immunotoxicity investigations is lacking, particularly among regulatory bodies. Therefore, a critical examination is needed for the appropriate interpretations and potential misinterpretations of such data during the assessment of drug safety and chemical risk. As such, the current uses and implications of immunophenotyping data in human health safety and risk assessments has been evaluated to provide additional context for the application of current methodologies and guidelines. In addition, case studies are presented to highlight the challenges of interpreting immunophenotyping results along with incorporating the findings into immunotoxicity investigations. Based on the analyses of current approaches and methodologies, a decision flow is presented for use of immunophenotyping data during risk informed decision making.
Collapse
Affiliation(s)
- Victor J. Johnson
- Burleson Research Technologies, Inc., Morrisville, NC, United States
| | | | - Andrew Maier
- Stantec ChemRisk, Cincinnati, OH, United States
- Integral Consulting, Inc., Cincinnati, OH, United States
| | - Corey Boles
- Stantec ChemRisk, Raleigh, NC, United States
- Insight Exposure and Risk Sciences Group, Raleigh, NC, United States
| | | | - Daniel E. Arrieta
- Chevron Phillips Chemical Company LP, The Woodlands, TX, United States
| |
Collapse
|
24
|
Huang Y, Li H, Liang R, Chen J, Tang Q. The influence of sex-specific factors on biological transformations and health outcomes in aging processes. Biogerontology 2024; 25:775-791. [PMID: 39001953 PMCID: PMC11374838 DOI: 10.1007/s10522-024-10121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
The aging process demonstrates notable differences between males and females, which are key factors in disease susceptibility and lifespan. The differences in sex chromosomes are fundamental to the presence of sex bias in organisms. Moreover, sex-specific epigenetic modifications and changes in sex hormone levels impact the development of immunity differently during embryonic development and beyond. Mitochondria, telomeres, homeodynamic space, and intestinal flora are intricately connected to sex differences in aging. These elements can have diverse effects on men and women, resulting in unique biological transformations and health outcomes as they grow older. This review explores how sex interacts with these elements and shapes the aging process.
Collapse
Affiliation(s)
- Yongyin Huang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Hongyu Li
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Qiang Tang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China.
| |
Collapse
|
25
|
Yolmo P, Rahimi S, Chenard S, Conseil G, Jenkins D, Sachdeva K, Emon I, Hamilton J, Xu M, Rangachari M, Michaud E, Mansure JJ, Kassouf W, Berman DM, Siemens DR, Koti M. Atypical B Cells Promote Cancer Progression and Poor Response to Bacillus Calmette-Guérin in Non-Muscle Invasive Bladder Cancer. Cancer Immunol Res 2024; 12:1320-1339. [PMID: 38916567 PMCID: PMC11443217 DOI: 10.1158/2326-6066.cir-23-1114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/03/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Poor response to Bacillus Calmette-Guérin (BCG) immunotherapy remains a major barrier in the management of patients with non-muscle invasive bladder cancer (NMIBC). Multiple factors are associated with poor outcomes, including biological aging and female sex. More recently, it has emerged that a B-cell-infiltrated pretreatment immune microenvironment of NMIBC tumors can influence the response to intravesically administered BCG. The mechanisms underlying the roles of B cells in NMIBC are poorly understood. Here, we show that B-cell-dominant tertiary lymphoid structures (TLSs), a hallmark feature of the chronic mucosal immune response, are abundant and located close to the epithelial compartment in pretreatment tumors from BCG non-responders. Digital spatial proteomic profiling of whole tumor sections from male and female patients with NMIBC who underwent treatment with intravesical BCG, revealed higher expression of immune exhaustion-associated proteins within the tumor-adjacent TLSs in both responders and non-responders. Chronic local inflammation, induced by the N-butyl-N-(4-hydroxybutyl) nitrosamine carcinogen, led to TLS formation with recruitment and differentiation of the immunosuppressive atypical B-cell (ABC) subset within the bladder microenvironment, predominantly in aging female mice compared to their male counterparts. Depletion of ABCs simultaneous to BCG treatment delayed cancer progression in female mice. Our findings provide evidence indicating a role for ABCs in BCG response and will inform future development of therapies targeting the B-cell-exhaustion axis.
Collapse
Affiliation(s)
- Priyanka Yolmo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Sadaf Rahimi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Stephen Chenard
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Gwenaëlle Conseil
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Danielle Jenkins
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Kartik Sachdeva
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Isaac Emon
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Jake Hamilton
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Minqi Xu
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Manu Rangachari
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Eva Michaud
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - Jose J Mansure
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - Wassim Kassouf
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - David M Berman
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - David R Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| |
Collapse
|
26
|
Fritz García JHG, Keller Valsecchi CI, Basilicata MF. Sex as a biological variable in ageing: insights and perspectives on the molecular and cellular hallmarks. Open Biol 2024; 14:240177. [PMID: 39471841 PMCID: PMC11521605 DOI: 10.1098/rsob.240177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 11/01/2024] Open
Abstract
Sex-specific differences in lifespan and ageing are observed in various species. In humans, women generally live longer but are frailer and suffer from different age-related diseases compared to men. The hallmarks of ageing, such as genomic instability, telomere attrition or loss of proteostasis, exhibit sex-specific patterns. Sex chromosomes and sex hormones, as well as the epigenetic regulation of the inactive X chromosome, have been shown to affect lifespan and age-related diseases. Here we review the current knowledge on the biological basis of sex-biased ageing. While our review is focused on humans, we also discuss examples of model organisms such as the mouse, fruit fly or the killifish. Understanding these molecular differences is crucial as the elderly population is expected to double worldwide by 2050, making sex-specific approaches in the diagnosis, treatment, therapeutic development and prevention of age-related diseases a pressing need.
Collapse
Affiliation(s)
| | | | - M. Felicia Basilicata
- Institute of Molecular Biology (IMB), Mainz, Germany
- University Medical Center (UMC), Mainz, Germany
| |
Collapse
|
27
|
Huang Y, Huang X, Ling L, Liu C, Chen S, Zhou J, Du L, Xiong J. Prospective Reassessment of the Association Between Pro-Inflammatory Factors and Prognosis After on-Pump Cardiac Surgery. KARDIOLOGIIA 2024; 64:87-95. [PMID: 39392271 DOI: 10.18087/cardio.2024.9.n2682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/08/2024] [Indexed: 10/12/2024]
Abstract
AIM Elevated levels of pro-inflammatory factors in plasma have been linked to worse prognosis after on-pump cardiac surgery, yet interventions that reduce the levels in patients have failed to improve prognosis. Therefore, we explored whether levels of pro-inflammatory factors are associated with prognosis of patients after valve surgery with cardiopulmonary bypass. MATERIAL AND METHODS 244 patients were prospectively enrolled into observational study. Levels of tumor necrosis factor (TNF) - α, interleukin-8 and neutrophil elastase were measured once before and several times after cardiopulmonary bypass. The levels were compared between patients who experienced in-hospital adverse events or not, and between patients who experienced major adverse cardiac or cerebrovascular events (MACCEs) during three-year follow-up or not. RESULTS Of the 244 patients enrolled, in-hospital adverse events occurred in 38 (15.6 %); of the 237 patients who completed follow-up, MACCEs occurred in 30 (12.7 %). Surgery led to significant increases in levels of all three pro-inflammatory factors, with levels returning to pre-bypass baseline on arrival in the intensive care unit (TNF-α), 4 h after arrival (interleukin-8) or 20 h after arrival (neutrophil elastase). However, pre- and post-bypass levels of all three factors did not differ significantly between patients who experienced adverse events in-hospital or not, or between patients who experienced MACCEs during follow-up or not. CONCLUSIONS Levels of TNF-α, interleukin-8 and neutrophil elastase may not be associated with poor prognosis after cardiopulmonary bypass. This may help explain why "cytokine clearance" strategies fail to improve clinical outcomes after on-pump cardiac surgery.
Collapse
Affiliation(s)
- Yingting Huang
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Xunbei Huang
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Liqin Ling
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Chaonan Liu
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Si Chen
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Jing Zhou
- Sichuan University, West China Hospital, Department of Laboratory Medicine
| | - Lei Du
- Sichuan University, West China Hospital, Department of Anesthesiology
| | - Jiyue Xiong
- Sichuan University, West China Hospital, Department of Anesthesiology
| |
Collapse
|
28
|
Al-Diab O, Sünkel C, Blanc E, Catar RA, Ashraf MI, Zhao H, Wang P, Rinschen MM, Fritsche-Guenther R, Grahammer F, Bachmann S, Beule D, Kirwan JA, Rajewsky N, Huber TB, Gürgen D, Kusch A. Sex-specific molecular signature of mouse podocytes in homeostasis and in response to pharmacological challenge with rapamycin. Biol Sex Differ 2024; 15:72. [PMID: 39278930 PMCID: PMC11404044 DOI: 10.1186/s13293-024-00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/30/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Sex differences exist in the prevalence and progression of major glomerular diseases. Podocytes are the essential cell-type in the kidney which maintain the physiological blood-urine barrier, and pathological changes in podocyte homeostasis are critical accelerators of impairment of kidney function. However, sex-specific molecular signatures of podocytes under physiological and stress conditions remain unknown. This work aimed at identifying sexual dimorphic molecular signatures of podocytes under physiological condition and pharmacologically challenged homeostasis with mechanistic target of rapamycin (mTOR) inhibition. mTOR is a crucial regulator involved in a variety of physiological and pathological stress responses in the kidney and inhibition of this pathway may therefore serve as a general stress challenger to get fundamental insights into sex differences in podocytes. METHODS The genomic ROSAmT/mG-NPHS2 Cre mouse model was used which allows obtaining highly pure podocyte fractions for cell-specific molecular analyses, and vehicle or pharmacologic treatment with the mTOR inhibitor rapamycin was performed for 3 weeks. Subsequently, deep RNA sequencing and proteomics were performed of the isolated podocytes to identify intrinsic sex differences. Studies were supplemented with metabolomics from kidney cortex tissues. RESULTS Although kidney function and morphology remained normal in all experimental groups, RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways. Interestingly, rapamycin abolished prominent sex-specific clustering of podocyte gene expression and induced major changes only in male transcriptome. Several sex-biased transcription factors could be identified as possible upstream regulators of these sexually dimorphic responses. Concordant to transcriptomics, metabolomic changes were more prominent in males. Remarkably, high number of previously reported kidney disease genes showed intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. CONCLUSIONS Our results highlight remarkable intrinsic sex-differences and sex-specific response patterns towards pharmacological challenged podocyte homeostasis which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. This work provides rationale and an in-depth database for novel targets to be tested in specific kidney disease models to advance with sex-specific treatment strategies.
Collapse
Affiliation(s)
- Ola Al-Diab
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christin Sünkel
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Rusan Ali Catar
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pinchao Wang
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Markus M Rinschen
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Raphaela Fritsche-Guenther
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jennifer A Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Dennis Gürgen
- Experimental Pharmacology & Oncology Berlin-Buch GmbH, 13125 Berlin-Buch, Germany
| | - Angelika Kusch
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- BIH Biomedical Innovation Academy (BIA), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
29
|
Blanton LV, San Roman AK, Wood G, Buscetta A, Banks N, Skaletsky H, Godfrey AK, Pham TT, Hughes JF, Brown LG, Kruszka P, Lin AE, Kastner DL, Muenke M, Page DC. Stable and robust Xi and Y transcriptomes drive cell-type-specific autosomal and Xa responses in vivo and in vitro in four human cell types. CELL GENOMICS 2024; 4:100628. [PMID: 39111319 PMCID: PMC11480847 DOI: 10.1016/j.xgen.2024.100628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024]
Abstract
Recent in vitro studies of human sex chromosome aneuploidy showed that the Xi ("inactive" X) and Y chromosomes broadly modulate autosomal and Xa ("active" X) gene expression. We tested these findings in vivo. Linear modeling of CD4+ T cells and monocytes from individuals with one to three X chromosomes and zero to two Y chromosomes revealed 82 sex-chromosomal and 344 autosomal genes whose expression changed significantly with Xi and/or Y dosage in vivo. Changes in sex-chromosomal expression were remarkably constant in vivo and in vitro; autosomal responses to Xi and/or Y dosage were largely cell-type specific (∼2.6-fold more variation than sex-chromosomal responses). Targets of the sex-chromosomal transcription factors ZFX and ZFY accounted for a significant fraction of these autosomal responses both in vivo and in vitro. We conclude that the human Xi and Y transcriptomes are surprisingly robust and stable, yet they modulate autosomal and Xa genes in a cell-type-specific fashion.
Collapse
Affiliation(s)
| | | | - Geryl Wood
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ashley Buscetta
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Banks
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helen Skaletsky
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | | | - Thao T Pham
- Whitehead Institute, Cambridge, MA 02142, USA
| | | | - Laura G Brown
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | - Paul Kruszka
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela E Lin
- Genetics Unit, MassGeneral for Children, Boston, MA 02114, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David C Page
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
30
|
McLamb F, Feng Z, Vu JP, Griffin L, Vasquez MF, Bozinovic G. Lagging Brain Gene Expression Patterns of Drosophila melanogaster Young Adult Males Confound Comparisons Between Sexes. Mol Neurobiol 2024:10.1007/s12035-024-04427-7. [PMID: 39196495 DOI: 10.1007/s12035-024-04427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
Many species, including fruit flies (Drosophila melanogaster), are sexually dimorphic. Phenotypic variation in morphology, physiology, and behavior can affect development, reproduction, health, and aging. Therefore, designating sex as a variable and sex-blocking should be considered when designing experiments. The brain regulates phenotypes throughout the lifespan by balancing survival and reproduction, and sex-specific development at each life stage is likely. Changes in morphology and physiology are governed by differential gene expression, a quantifiable molecular marker for age- and sex-specific variations. We assessed the fruit fly brain transcriptome at three adult ages for gene expression signatures of sex, age, and sex-by-age: 6698 genes were differentially expressed between sexes, with the most divergence at 3 days. Between ages, 31.1% of 6084 differentially expressed genes (1890 genes) share similar expression patterns from 3 to 7 days in females, and from 7 to 14 days in males. Most of these genes (90.5%, 1712) were upregulated and enriched for chemical stimulus detection and/or cilium regulation. Our data highlight an important delay in male brain gene regulation compared to females. Because significant delays in expression could confound comparisons between sexes, studies of sexual dimorphism at phenotypically comparable life stages rather than chronological age should be more biologically relevant.
Collapse
Affiliation(s)
- Flannery McLamb
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, USA
| | - Zuying Feng
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA
| | - Jeanne P Vu
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA
- Graduate School of Public Health, San Diego State University, San Diego, CA, USA
| | - Lindsey Griffin
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, USA
| | - Miguel F Vasquez
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, USA
| | - Goran Bozinovic
- Boz Life Science Research and Teaching Institute, La Jolla, CA, USA.
- Graduate School of Public Health, San Diego State University, San Diego, CA, USA.
- Center for Life in Extreme Environments, Portland State University, Portland, OR, USA.
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
31
|
Pan PH, Luo CW, Ting WC, Shiu BH, Huang JY, Tsai SCS, Lin FCF. Impact of Ascending HPV Infection on Colorectal Cancer Risk: Evidence from a Nationwide Study. Microorganisms 2024; 12:1746. [PMID: 39338421 PMCID: PMC11434182 DOI: 10.3390/microorganisms12091746] [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: 07/22/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent and escalating health issue in Taiwan. This nationwide study delves into the relationship between Human Papillomavirus (HPV) infection and CRC risk, employing population datasets from 2007 to 2017. Cox regression analyses revealed a statistically significant hazard ratio (HR) of 1.73 (95% CI: 1.63-1.83) for CRC in HPV-positive patients, indicating a considerably elevated risk compared to non-infected individuals. Further, stratification by sex showed males with HPV have a higher CRC risk (HR = 1.49, 95% CI: 1.40-1.58) compared to females. Age-related analysis uncovered a progressive increase in CRC risk with advancing age (HR = 34.69 for over 80 years). The study of specific CRC subtypes showed varying risks: HR = 1.74 for the colon, HR = 1.64 for the rectum, and a notably higher HR = 4.72 for the anus. Comorbid conditions such as hypertension (HR = 1.26), diabetes mellitus (HR = 1.32), and abnormal liver function (HR = 1.18) also correlate with significantly increased CRC risks. These findings suggest that HPV is a significant risk factor for CRC, with disparities in risk based on anatomical location, demographic characteristics, and comorbidities, highlighting the need for intervention strategies and targeted prevention.
Collapse
Affiliation(s)
- Pin-Ho Pan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Ci-Wen Luo
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan
| | - Wen-Chien Ting
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Bei-Hao Shiu
- Division of Colorectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Jing-Yang Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Stella Chin-Shaw Tsai
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Superintendent Office, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan
- College of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Frank Cheau-Feng Lin
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
32
|
Noh JY, Han HW, Kim DM, Giles ED, Farnell YZ, Wright GA, Sun Y. Innate immunity in peripheral tissues is differentially impaired under normal and endotoxic conditions in aging. Front Immunol 2024; 15:1357444. [PMID: 39221237 PMCID: PMC11361940 DOI: 10.3389/fimmu.2024.1357444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic low-grade inflammation is a hallmark of aging, aka "inflammaging", which is linked to a wide range of age-associated diseases. Immune dysfunction increases disease susceptibility, and increases morbidity and mortality of aging. Innate immune cells, including monocytes, macrophages and neutrophils, are the first responders of host defense and the key mediators of various metabolic and inflammatory insults. Currently, the understanding of innate immune programming in aging is largely fragmented. Here we investigated the phenotypic and functional properties of innate immune cells in various peripheral tissues of young and aged mice under normal and endotoxic conditions. Under the steady state, aged mice showed elevated pro-inflammatory monocytes/macrophages in peripheral blood, adipose tissue, liver, and colon. Under lipopolysaccharide (LPS)-induced inflammatory state, the innate immune cells of aged mice showed a different response to LPS stimulus than that of young mice. LPS-induced immune responses displayed differential profiles in different tissues and cell types. In the peripheral blood, when responding to LPS, the aged mice showed higher neutrophils, but lower pro-inflammatory monocytes than that in young mice. In the peritoneal fluid, while young mice exhibited significantly elevated pro-inflammatory neutrophils and macrophages in response to LPS, aged mice exhibited decreased pro-inflammatory neutrophils and variable cytokine responses in macrophages. In the adipose tissue, LPS induced less infiltrated neutrophils but more infiltrated macrophages in old mice than young mice. In the liver, aged mice showed a more robust increase of pro-inflammatory macrophages compared to that in young mice under LPS stimulation. In colon, macrophages showed relatively mild response to LPS in both young and old mice. We have further tested bone-marrow derived macrophages (BMDM) from young and aged mice, we found that BMDM from aged mice have impaired polarization, displaying higher expression of pro-inflammatory markers than those from young mice. These data collectively suggest that innate immunity in peripheral tissues is impaired in aging, and the dysregulation of immunity is tissue- and cell-dependent. Our findings in the rodent model underscore the complexity of aging immunity. Further investigation is needed to determine whether the immune profile observed in aged mice is applicable in age-associated diseases in humans.
Collapse
Affiliation(s)
- Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
| | - Yuhua Z. Farnell
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Gus A. Wright
- Department of Veterinary Pathobiology, Texas A&M University,
College Station, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
33
|
Patrick R, Naval-Sanchez M, Deshpande N, Huang Y, Zhang J, Chen X, Yang Y, Tiwari K, Esmaeili M, Tran M, Mohamed AR, Wang B, Xia D, Ma J, Bayliss J, Wong K, Hun ML, Sun X, Cao B, Cottle DL, Catterall T, Barzilai-Tutsch H, Troskie RL, Chen Z, Wise AF, Saini S, Soe YM, Kumari S, Sweet MJ, Thomas HE, Smyth IM, Fletcher AL, Knoblich K, Watt MJ, Alhomrani M, Alsanie W, Quinn KM, Merson TD, Chidgey AP, Ricardo SD, Yu D, Jardé T, Cheetham SW, Marcelle C, Nilsson SK, Nguyen Q, White MD, Nefzger CM. The activity of early-life gene regulatory elements is hijacked in aging through pervasive AP-1-linked chromatin opening. Cell Metab 2024; 36:1858-1881.e23. [PMID: 38959897 DOI: 10.1016/j.cmet.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/28/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
A mechanistic connection between aging and development is largely unexplored. Through profiling age-related chromatin and transcriptional changes across 22 murine cell types, analyzed alongside previous mouse and human organismal maturation datasets, we uncovered a transcription factor binding site (TFBS) signature common to both processes. Early-life candidate cis-regulatory elements (cCREs), progressively losing accessibility during maturation and aging, are enriched for cell-type identity TFBSs. Conversely, cCREs gaining accessibility throughout life have a lower abundance of cell identity TFBSs but elevated activator protein 1 (AP-1) levels. We implicate TF redistribution toward these AP-1 TFBS-rich cCREs, in synergy with mild downregulation of cell identity TFs, as driving early-life cCRE accessibility loss and altering developmental and metabolic gene expression. Such remodeling can be triggered by elevating AP-1 or depleting repressive H3K27me3. We propose that AP-1-linked chromatin opening drives organismal maturation by disrupting cell identity TFBS-rich cCREs, thereby reprogramming transcriptome and cell function, a mechanism hijacked in aging through ongoing chromatin opening.
Collapse
Affiliation(s)
- Ralph Patrick
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Marina Naval-Sanchez
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Nikita Deshpande
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Yifei Huang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Jingyu Zhang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Xiaoli Chen
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Ying Yang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Kanupriya Tiwari
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Mohammadhossein Esmaeili
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Amin R Mohamed
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Binxu Wang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Di Xia
- Genome Innovation Hub, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Jun Ma
- Genome Innovation Hub, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Jacqueline Bayliss
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kahlia Wong
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Michael L Hun
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Xuan Sun
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin Cao
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Tara Catterall
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Hila Barzilai-Tutsch
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Institut NeuroMyoGène, University Claude Bernard Lyon 1, 69008 Lyon, France
| | - Robin-Lee Troskie
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Zhian Chen
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Andrea F Wise
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sheetal Saini
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ye Mon Soe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Snehlata Kumari
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Anne L Fletcher
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Konstantin Knoblich
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia; Research Centre for Health Sciences, Taif University, Taif, Saudi Arabia
| | - Walaa Alsanie
- Department of Clinical Laboratories Sciences, Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia; Research Centre for Health Sciences, Taif University, Taif, Saudi Arabia
| | - Kylie M Quinn
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Tobias D Merson
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ann P Chidgey
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Surgery, Cabrini Monash University, Malvern, VIC 3144, Australia
| | - Seth W Cheetham
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Christophe Marcelle
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Institut NeuroMyoGène, University Claude Bernard Lyon 1, 69008 Lyon, France
| | - Susan K Nilsson
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Melanie D White
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Christian M Nefzger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
34
|
van der Heiden M, Shetty S, Bijvank E, Beckers L, Cevirgel A, van Sleen Y, Tcherniaeva I, Ollinger T, Burny W, van Binnendijk RS, van Houten MA, Buisman AM, Rots NY, van Beek J, van Baarle D. Multiple vaccine comparison in the same adults reveals vaccine-specific and age-related humoral response patterns: an open phase IV trial. Nat Commun 2024; 15:6603. [PMID: 39097574 PMCID: PMC11297912 DOI: 10.1038/s41467-024-50760-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/18/2024] [Indexed: 08/05/2024] Open
Abstract
Vaccine responsiveness is often reduced in older adults. Yet, our lack of understanding of low vaccine responsiveness hampers the development of effective vaccination strategies to reduce the impact of infectious diseases in the ageing population. Young-adult (25-49 y), middle-aged (50-64 y) and older-adult ( ≥ 65 y) participants of the VITAL clinical trials (n = 315, age-range: 28-98 y), were vaccinated with an annual (2019-2020) quadrivalent influenza (QIV) booster vaccine, followed by a primary 13-valent pneumococcal-conjugate (PCV13) vaccine (summer/autumn 2020) and a primary series of two SARS-CoV-2 mRNA-1273 vaccines (spring 2021). This unique setup allowed investigation of humoral responsiveness towards multiple vaccines within the same individuals over the adult age-range. Booster QIV vaccination induced comparable H3N2 hemagglutination inhibition (HI) titers in all age groups, whereas primary PCV13 and mRNA-1273 vaccination induced lower antibody concentrations in older as compared to younger adults (primary endpoint). The persistence of humoral responses, towards the 6 months timepoint, was shorter in older adults for all vaccines (secondary endpoint). Interestingly, highly variable vaccine responder profiles overarching multiple vaccines were observed. Yet, approximately 10% of participants, mainly comprising of older male adults, were classified as low responders to multiple vaccines. This study aids the identification of risk groups for low vaccine responsiveness and hence supports targeted vaccination strategies. Trial number: NL69701.041.19, EudraCT: 2019-000836-24.
Collapse
Affiliation(s)
- Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Sudarshan Shetty
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Elske Bijvank
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Lisa Beckers
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Alper Cevirgel
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Irina Tcherniaeva
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | | | - Rob S van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marianne A van Houten
- Spaarne Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Pediatrics, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Anne-Marie Buisman
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Nynke Y Rots
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
35
|
Andrew MK. Commentary on: Cost-effectiveness of pneumococcal vaccination and of programs to increase its uptake in U.S. older adults. J Am Geriatr Soc 2024; 72:2299-2302. [PMID: 39007431 DOI: 10.1111/jgs.19079] [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: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024]
Abstract
See related articles by Wateska et al.
Collapse
Affiliation(s)
- Melissa K Andrew
- Department of Medicine (Geriatrics), Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
36
|
Yamamoto C, Kobashi Y, Kawamura T, Nishikawa Y, Saito H, Oguro F, Zhao T, Takita M, Sawano T, Ozaki A, Abe T, Ito N, Kaneko Y, Nakayama A, Wakui M, Kodama T, Tsubokura M. Group of longitudinal adverse event patterns after the fourth dose of COVID-19 vaccination with a latent class analysis. Front Public Health 2024; 12:1406315. [PMID: 39139673 PMCID: PMC11320210 DOI: 10.3389/fpubh.2024.1406315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Vaccination has been implemented as a useful measure to combat the COVID-19 pandemic. However, there is a tendency for individuals to avoid vaccination due to the possibility of adverse events, making it important to investigate the relationship between COVID-19 vaccines and their adverse events. This study explored longitudinal adverse event patterns and factors that influence adverse events following the second to fourth doses of the COVID-19 vaccine through a latent class analysis. Methods Participants were recruited from the Fukushima Prefecture and included individuals who had completed four doses of the COVID-19 mRNA vaccine. This study utilized data from questionnaire surveys and blood collection conducted between September 2021 and November 2022. In the questionnaire, factors such as sex, age, medical history, medication, type of vaccine administered, and adverse events following vaccination were recorded. Additionally, in the blood data, serological tests [IgG(S)] and cellular immune responses (T-spot) were measured. Descriptive statistics, latent class analysis, multivariable logistic regression, and multiple regression analyses were performed to identify the longitudinal adverse event patterns and influencing factors. By analyzing adverse events over time, we identified two distinct groups: those less prone to experiencing adverse events (Group 1) and those more susceptible (Group 2) to latent class analysis. Results A total of 1,175 participants were included after excluding those without any adverse events. The median age of the participants in Group 1 was 70 years, and in Group 2 it was 51 years. The proportion of female participants was 298 in Group 1 and 353 in Group 2. Patients in Group 2 were significantly younger (p < 0.001) and more likely to be female (p < 0.001) than those in Group 1. Furthermore, the median IgG(S) value after the fourth vaccination was 3,233 AU/mL in Group 1 and 4,059.39 AU/mL in Group 2. The median T-spot value was 15.4 in Group 1 and 28.5 in Group 2. Group 2 showed significantly higher IgG(S) and T-spot values after the fourth vaccination (p < 0.001). Discussion Our findings suggest that factors other than age, particularly sex and a history of allergies, significantly influence the likelihood of experiencing adverse events. Groups categorized by latent class analysis for longitudinal adverse events are expected to be valuable for optimizing vaccination strategies and formulating public health measures.
Collapse
Affiliation(s)
- Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Takeshi Kawamura
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Hiroaki Saito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of Internal Medicine, Soma Central Hospital, Soma, Fukushima, Japan
| | - Fumiya Oguro
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
| | - Tianchen Zhao
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Toyoaki Sawano
- Department of Surgery, Jyoban Hospital, Iwaki, Fukushima, Japan
| | - Akihiko Ozaki
- Department of Breast and Thyroid Surgery, Jyoban Hospital, Iwaki, Fukushima, Japan
| | - Toshiki Abe
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Naomi Ito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Minato-ku, Tokyo, Japan
| | - Aya Nakayama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Fukushima, Japan
- Minamisoma Municipal General Hospital, Minamisoma, Fukushima, Japan
| |
Collapse
|
37
|
Hahn EE, Stiller J, Alexander MR, Grealy A, Taylor JM, Jackson N, Frere CH, Holleley CE. Century-old chromatin architecture revealed in formalin-fixed vertebrates. Nat Commun 2024; 15:6378. [PMID: 39075073 PMCID: PMC11286846 DOI: 10.1038/s41467-024-50668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/18/2024] [Indexed: 07/31/2024] Open
Abstract
Gene expression is regulated by changes in chromatin architecture intrinsic to cellular differentiation and as an active response to environmental stimuli. Chromatin dynamics are a major driver of phenotypic diversity, regulation of development, and manifestation of disease. Remarkably, we know little about the evolutionary dynamics of chromatin reorganisation through time, data essential to characterise the impact of environmental stress during the ongoing biodiversity extinction crisis (20th-21st century). Linking the disparate fields of chromatin biology and museum science through their common use of the preservative formaldehyde (a constituent of formalin), we have generated historical chromatin profiles in museum specimens up to 117 years old. Historical chromatin profiles are reproducible, tissue-specific, sex-specific, and environmental condition-dependent in vertebrate specimens. Additionally, we show that over-fixation modulates differential chromatin accessibility to enable semi-quantitative estimates of relative gene expression in vertebrates and a yeast model. Our approach transforms formalin-fixed biological collections into an accurate, comprehensive, and global record of environmental impact on gene expression and phenotype.
Collapse
Affiliation(s)
- Erin E Hahn
- National Research Collections Australia, Commonwealth Scientific Industrial Research Organisation, Canberra, ACT 2601, Australia
| | - Jiri Stiller
- Agriculture and Food, Commonwealth Scientific Industrial Research Organisation, St Lucia, Queensland, 4067, Australia
| | - Marina R Alexander
- National Research Collections Australia, Commonwealth Scientific Industrial Research Organisation, Canberra, ACT 2601, Australia
| | - Alicia Grealy
- National Research Collections Australia, Commonwealth Scientific Industrial Research Organisation, Canberra, ACT 2601, Australia
| | - Jennifer M Taylor
- Agriculture and Food, Commonwealth Scientific Industrial Research Organisation, St Lucia, Queensland, 4067, Australia
| | - Nicola Jackson
- School of the Environment, University of Queensland, St Lucia, Queensland, 4067, Australia
| | - Celine H Frere
- School of the Environment, University of Queensland, St Lucia, Queensland, 4067, Australia
| | - Clare E Holleley
- National Research Collections Australia, Commonwealth Scientific Industrial Research Organisation, Canberra, ACT 2601, Australia.
| |
Collapse
|
38
|
Janssen LLG, van Leeuwen-Kerkhoff N, Westers TM, de Gruijl TD, van de Loosdrecht AA. The immunoregulatory role of monocytes and thrombomodulin in myelodysplastic neoplasms. Front Oncol 2024; 14:1414102. [PMID: 39132505 PMCID: PMC11310157 DOI: 10.3389/fonc.2024.1414102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) are clonal disorders of the myeloid lineage leading to peripheral blood cytopenias. Dysregulation of innate immunity is hypothesized to be a potent driver of MDS. A recent study revealed increased thrombomodulin (TM) expression on classical monocytes in MDS, which was associated with prolonged survival. TM is a receptor with immunoregulatory capacities, however, its exact role in MDS development remains to be elucidated. In this review we focus on normal monocyte biology and report on the involvement of monocytes in myeloid disease entities with a special focus on MDS. Furthermore, we delve into the current knowledge on TM and its function in monocytes in health and disease and explore the role of TM-expressing monocytes as driver, supporter or epiphenomenon in the MDS bone marrow environment.
Collapse
Affiliation(s)
- Luca L. G. Janssen
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Nathalie van Leeuwen-Kerkhoff
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Theresia M. Westers
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Institute for Immunity and Infectious Diseases, Amsterdam, Netherlands
| | - Arjan A. van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands
| |
Collapse
|
39
|
Korst CLBM, O’Neill C, Bruins WSC, Cosovic M, Twickler I, Verkleij CPM, Le Clerre D, Themeli M, Chion-Sotinel I, Zweegman S, Galetto R, Mutis T, van de Donk NWCJ. Preclinical activity of allogeneic SLAMF7-specific CAR T-cells (UCARTCS1) in multiple myeloma. J Immunother Cancer 2024; 12:e008769. [PMID: 39060023 PMCID: PMC11284884 DOI: 10.1136/jitc-2023-008769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Autologous BCMA-specific CAR T-cell therapies have substantial activity in multiple myeloma (MM). However, due to logistical limitations and BCMAlow relapses, there is a need for alternatives. UCARTCS1 cells are 'off-the-shelf' allogeneic CAR T-cells derived from healthy donors targeting SLAMF7 (CS1), which is highly expressed in MM cells. In this study, we evaluated the preclinical activity of UCARTCS1 in MM cell lines, in bone marrow (BM) samples obtained from MM patients and in an MM mouse model. METHODS Luciferase-transduced MM cell lines were incubated with UCARTCS1 cells or control (non-transduced, SLAMF7/TCRαβ double knock-out) T-cells at different effector to target ratios for 24 hours. MM cell lysis was assessed by bioluminescence. Anti-MM activity of UCARTCS1 was also evaluated in 29 BM samples obtained from newly diagnosed patients (n=10), daratumumab-naïve relapsed/refractory patients (n=10) and daratumumab-refractory patients (n=9) in 24-hour flow cytometry-based cytotoxicity assays. Finally, UCARTCS1 activity was assessed in mouse xenograft models. RESULTS UCARTCS1 cells induced potent CAR-mediated, and dose-dependent lysis of both MM cell lines and primary MM cells. There was no difference in ex vivo activity of UCARTCS1 between heavily pretreated and newly diagnosed patients. In addition, efficacy of UCARTCS1 was not affected by SLAMF7 expression level on MM cells, proportion of tumor cells, or frequency of regulatory T-cells in BM samples obtained from MM patients. UCARTCS1 treatment eliminated SLAMF7+ non-malignant immune cells in a dose-dependent manner, however lysis of normal cells was less pronounced compared to that of MM cells. Additionally, durable anti-MM responses were observed with UCARTCS1 in an MM xenograft model. CONCLUSIONS These results demonstrate that UCARTCS1 has potent anti-MM activity against MM cell lines and primary MM cells, as well as in an MM xenograft model and support the evaluation of UCARTCS1 in patients with advanced MM.
Collapse
Affiliation(s)
- Charlotte L B M Korst
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Chloe O’Neill
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Wassilis S C Bruins
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Meliha Cosovic
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Inoka Twickler
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Maria Themeli
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Wang TW, Nakanishi M. Immune surveillance of senescence: potential application to age-related diseases. Trends Cell Biol 2024:S0962-8924(24)00121-1. [PMID: 39025762 DOI: 10.1016/j.tcb.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
Several lines of evidence suggest that the age-dependent accumulation of senescent cells leads to chronic tissue microinflammation, which in turn contributes to age-related pathologies. In general, senescent cells can be eliminated by the host's innate and adaptive immune surveillance system, including macrophages, NK cells, and T cells. Impaired immune surveillance leads to the accumulation of senescent cells and accelerates the aging process. Recently, senescent cells, like cancer cells, have been shown to express certain types of immune checkpoint proteins as well as non-classical immune-tolerant MHC variants, leading to immune escape from surveillance systems. Thus, immune checkpoint blockade (ICB) may be a promising strategy to enhance immune surveillance of senescence, leading to the amelioration of some age-related diseases and tissue dysfunction.
Collapse
Affiliation(s)
- Teh-Wei Wang
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
41
|
Franco-Enzástiga Ú, Inturi NN, Natarajan K, Mwirigi JM, Mazhar K, Schlachetzki JC, Schumacher M, Price TJ. Epigenomic landscape of the human dorsal root ganglion: sex differences and transcriptional regulation of nociceptive genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587047. [PMID: 38586055 PMCID: PMC10996669 DOI: 10.1101/2024.03.27.587047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Gene expression is influenced by chromatin architecture via controlled access of regulatory factors to DNA. To better understand gene regulation in the human dorsal root ganglion (hDRG) we used bulk and spatial transposase-accessible chromatin technology followed by sequencing (ATAC-seq). Using bulk ATAC-seq, we detected that in females diverse differentially accessible chromatin regions (DARs) mapped to the X chromosome and in males to autosomal genes. EGR1/3 and SP1/4 transcription factor binding motifs were abundant within DARs in females, and JUN, FOS and other AP-1 factors in males. To dissect the open chromatin profile in hDRG neurons, we used spatial ATAC-seq. The neuron cluster showed higher chromatin accessibility in GABAergic, glutamatergic, and interferon-related genes in females, and in Ca2+- signaling-related genes in males. Sex differences in transcription factor binding sites in neuron-proximal barcodes were consistent with the trends observed in bulk ATAC-seq data. We validated that EGR1 expression is biased to female hDRG compared to male. Strikingly, XIST, the long-noncoding RNA responsible for X inactivation, hybridization signal was found to be highly dispersed in the female neuronal but not non-neuronal nuclei suggesting weak X inactivation in female hDRG neurons. Our findings point to baseline epigenomic sex differences in the hDRG that likely underlie divergent transcriptional responses that determine mechanistic sex differences in pain.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Nikhil N. Inturi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Khadija Mazhar
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Johannes C.M. Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, California, 94143 USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
42
|
Booijink R, Ramachandran P, Bansal R. Implications of innate immune sexual dimorphism for MASLD pathogenesis and treatment. Trends Pharmacol Sci 2024; 45:614-627. [PMID: 38853100 DOI: 10.1016/j.tips.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
43
|
Han J, Rindone AN, Elisseeff JH. Immunoengineering Biomaterials for Musculoskeletal Tissue Repair across Lifespan. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311646. [PMID: 38416061 PMCID: PMC11239302 DOI: 10.1002/adma.202311646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Musculoskeletal diseases and injuries are among the leading causes of pain and morbidity worldwide. Broad efforts have focused on developing pro-regenerative biomaterials to treat musculoskeletal conditions; however, these approaches have yet to make a significant clinical impact. Recent studies have demonstrated that the immune system is central in orchestrating tissue repair and that targeting pro-regenerative immune responses can improve biomaterial therapeutic outcomes. However, aging is a critical factor negatively affecting musculoskeletal tissue repair and immune function. Hence, understanding how age affects the response to biomaterials is essential for improving musculoskeletal biomaterial therapies. This review focuses on the intersection of the immune system and aging in response to biomaterials for musculoskeletal tissue repair. The article introduces the general impacts of aging on tissue physiology, the immune system, and the response to biomaterials. Then, it explains how the adaptive immune system guides the response to injury and biomaterial implants in cartilage, muscle, and bone and discusses how aging impacts these processes in each tissue type. The review concludes by highlighting future directions for the development and translation of personalized immunomodulatory biomaterials for musculoskeletal tissue repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Alexandra N. Rindone
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine; Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| |
Collapse
|
44
|
Trombetta CM, Accardi G, Aiello A, Calabrò A, Caruso C, Ligotti ME, Marchi S, Montomoli E, Neto MM, Temperton N, Candore G. Centenarians, semi and supercentenarians, COVID-19 and Spanish flu: a serological assessment to gain insight into the resilience of older centenarians to COVID-19. Immun Ageing 2024; 21:44. [PMID: 38937774 PMCID: PMC11210044 DOI: 10.1186/s12979-024-00450-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Although it is well known that the older people have been the most susceptible to COVID-19, there are conflicting data on the susceptibility of centenarians. Two epidemiological study have shown that older centenarians (> 101 years old at the time of the 2020 pandemic peak) are more resilient than the remaining centenarians, suggesting that this resilience might be linked to the 1918 Spanish Flu pandemic. To gain insight into this matter, specifically whether the resilience of older centenarians to SARS-CoV-2 infection is linked to the Spanish Flu they had been affected by, we conducted a retrospective serological study. This study examined serum samples from 33 centenarians, encompassing semi- (aged > 104 < 110 years, N = 7) and supercentenarians (aged > 109 years, N = 4), born between 1905 and 1922, against both SARS-CoV-2 and 1918 H1N1 pseudotype virus. RESULTS Anamnestic and laboratory data suggest that SARS-CoV-2 infection occurred in 8 centenarians. The infection appeared to have been asymptomatic or mild, and hospitalization was not required, despite 3 out of 8 being between 109 and 110 years old. The levels of anti-spike antibodies in centenarians infected and/or vaccinated were higher, although not significantly, than those produced by a random sample of seventy-year-old individuals used as controls. All centenarians had antibody levels against the 1918 H1N1 virus significantly higher (almost 50 times) than those observed in the quoted group of seventy-year-old subjects, confirming the key role in maintaining immunological memory from a priming that occurred over 100 years ago. Centenarians whose blood was collected prior to the pandemic outbreak demonstrated neutralising antibodies against the 1918 H1N1 virus, but all these subjects tested negative for SARS-CoV-2. CONCLUSION This retrospective study shows that older centenarians are quite resilient to COVID-19, as they are capable of producing good levels of neutralising antibodies and experiencing mild or asymptomatic disease. This could be attributed to the 1918 Spanish flu pandemic through mechanisms other than the presence of cross-reactive antibodies between the 1918 H1N1 virus and SARS-CoV-2. Another possibility is that the association is purely temporal, solely correlated with the advanced age of resilient centenarians compared to those born after 1918, since older centenarians are known to have better control of immune-inflammatory responses.
Collapse
Affiliation(s)
- Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi Research srl, Siena, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Department of Research, ISMETT-IRCCS Mediterranean Institute forTransplants and Highly Specialized Therapies, Palermo, Italy
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi Research srl, Siena, Italy
- VisMederi srl, Siena, Italy
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Kent, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Kent, UK
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
45
|
Cho HM, Choe SH, Lee JR, Park HR, Ko MG, Lee YJ, Lee HY, Park SH, Park SJ, Kim YH, Huh JW. Transcriptome analysis of cynomolgus macaques throughout their lifespan reveals age-related immune patterns. NPJ AGING 2024; 10:30. [PMID: 38902280 PMCID: PMC11189941 DOI: 10.1038/s41514-024-00158-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Despite the different perspectives by diverse research sectors spanning several decades, aging research remains uncharted territory for human beings. Therefore, we investigated the transcriptomic characteristics of eight male healthy cynomolgus macaques, and the annual sampling was designed with two individuals in four age groups. As a laboratory animal, the macaques were meticulously shielded from all environmental factors except aging. The results showed recent findings of certain immune response and the age-associated network of primate immunity. Three important aging patterns were identified and each gene clusters represented a different immune response. The increased expression pattern was predominantly associated with innate immune cells, such as Neutrophils and NK cells, causing chronic inflammation with aging whereas the other two decreased patterns were associated with adaptive immunity, especially "B cell activation" affecting antibody diversity of aging. Furthermore, the hub gene network of the patterns reflected transcriptomic age and correlated with human illness status, aiding in future human disease prediction. Our macaque transcriptome profiling results offer systematic insights into the age-related immunological features of primates.
Collapse
Affiliation(s)
- Hyeon-Mu Cho
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Se-Hee Choe
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Ja-Rang Lee
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56216, Republic of Korea
| | - Hye-Ri Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Min-Gyeong Ko
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Yun-Jung Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Hwal-Yong Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Sung Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea.
| |
Collapse
|
46
|
Boetto C, Frouin A, Henches L, Auvergne A, Suzuki Y, Patin E, Bredon M, Chiu A, Consortium MI, Sankararaman S, Zaitlen N, Kennedy SP, Quintana-Murci L, Duffy D, Sokol H, Aschard H. MANOCCA: a robust and computationally efficient test of covariance in high-dimension multivariate omics data. Brief Bioinform 2024; 25:bbae272. [PMID: 38856173 PMCID: PMC11163461 DOI: 10.1093/bib/bbae272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/16/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024] Open
Abstract
Multivariate analysis is becoming central in studies investigating high-throughput molecular data, yet, some important features of these data are seldom explored. Here, we present MANOCCA (Multivariate Analysis of Conditional CovAriance), a powerful method to test for the effect of a predictor on the covariance matrix of a multivariate outcome. The proposed test is by construction orthogonal to tests based on the mean and variance and is able to capture effects that are missed by both approaches. We first compare the performances of MANOCCA with existing correlation-based methods and show that MANOCCA is the only test correctly calibrated in simulation mimicking omics data. We then investigate the impact of reducing the dimensionality of the data using principal component analysis when the sample size is smaller than the number of pairwise covariance terms analysed. We show that, in many realistic scenarios, the maximum power can be achieved with a limited number of components. Finally, we apply MANOCCA to 1000 healthy individuals from the Milieu Interieur cohort, to assess the effect of health, lifestyle and genetic factors on the covariance of two sets of phenotypes, blood biomarkers and flow cytometry-based immune phenotypes. Our analyses identify significant associations between multiple factors and the covariance of both omics data.
Collapse
Affiliation(s)
- Christophe Boetto
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Arthur Frouin
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Léo Henches
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Antoine Auvergne
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Yuka Suzuki
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Etienne Patin
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, 25-28 rue Dr Roux, 75015 Paris, France
| | - Marius Bredon
- Sorbonne Université, INSERM, Centre de recherche Saint-Antoine, CRSA, Microbiota, Gut and Inflammation Laboratory, Hôpital Saint-Antoine (UMR S938) Sorbonne Université, 27 rue Chaligny, 75012 Paris, France
| | - Alec Chiu
- Department of Human Genetics, University California Los Angeles, 695 Charles E. Young Drive South, Box 708822, Los Angeles, CA 90095-7088, United States
| | | | - Sriram Sankararaman
- Department of Human Genetics, University California Los Angeles, 695 Charles E. Young Drive South, Box 708822, Los Angeles, CA 90095-7088, United States
| | - Noah Zaitlen
- Department of Human Genetics, University California Los Angeles, 695 Charles E. Young Drive South, Box 708822, Los Angeles, CA 90095-7088, United States
| | - Sean P Kennedy
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Lluis Quintana-Murci
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, 25-28 rue Dr Roux, 75015 Paris, France
- Chair of Human Genomics and Evolution, Collège de France, 11 Pl. Marcelin Berthelot, 75005 Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de recherche Saint-Antoine, CRSA, Microbiota, Gut and Inflammation Laboratory, Hôpital Saint-Antoine (UMR S938) Sorbonne Université, 27 rue Chaligny, 75012 Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, 184 rue du Faubourg Saint-Antoine, 75571 PARIS Cedex 12, France
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, 184 rue du faubourg Saint-Antoine, 75012 Paris, France
- INRAE Micalis & AgroParisTech, UMR1319, Micalis & AgroParisTech, 4 avenue Jean Jaurès, 78352 Jouy en Josas, France
| | - Hugues Aschard
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, 25-28 rue du Dr Roux, 75015 Paris, France
- Department of Epidemiology, Harvard TH Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| |
Collapse
|
47
|
Huret C, Ferrayé L, David A, Mohamed M, Valentin N, Charlotte F, Savignac M, Goodhardt M, Guéry JC, Rougeulle C, Morey C. Altered X-chromosome inactivation predisposes to autoimmunity. SCIENCE ADVANCES 2024; 10:eadn6537. [PMID: 38701219 PMCID: PMC11068014 DOI: 10.1126/sciadv.adn6537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024]
Abstract
In mammals, males and females show marked differences in immune responses. Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation (XCI), the epigenetic mechanism ensuring the silencing of one X in females, may participate in these sex biases. We perturbed the expression of the trigger of XCI, the noncoding RNA Xist, in female mice. This resulted in reactivation of genes on the inactive X, including members of the Toll-like receptor 7 (TLR7) signaling pathway, in monocyte/macrophages and dendritic and B cells. Consequently, female mice spontaneously developed inflammatory signs typical of lupus, including anti-nucleic acid autoantibodies, increased frequencies of age-associated and germinal center B cells, and expansion of monocyte/macrophages and dendritic cells. Mechanistically, TLR7 signaling is dysregulated in macrophages, leading to sustained expression of target genes upon stimulation. These findings provide a direct link between maintenance of XCI and female-biased autoimmune manifestations and highlight altered XCI as a cause of autoimmunity.
Collapse
Affiliation(s)
- Christophe Huret
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Léa Ferrayé
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Paul Sabatier, Toulouse, France
| | - Antoine David
- Université Paris Cité, INSERM UMRS 976, Institut de Recherche Saint Louis, F-75010, Paris, France
| | - Myriame Mohamed
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Nicolas Valentin
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Frédéric Charlotte
- Sorbonne University, Department of Pathological Anatomy and Cytology, Hôpital Pitié-Salpêtrière Charles Foix, F-75013, Paris, France
| | - Magali Savignac
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Paul Sabatier, Toulouse, France
| | - Michele Goodhardt
- Université Paris Cité, INSERM UMRS 976, Institut de Recherche Saint Louis, F-75010, Paris, France
| | - Jean-Charles Guéry
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Paul Sabatier, Toulouse, France
| | - Claire Rougeulle
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Céline Morey
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| |
Collapse
|
48
|
Stankiewicz LN, Rossi FMV, Zandstra PW. Rebuilding and rebooting immunity with stem cells. Cell Stem Cell 2024; 31:597-616. [PMID: 38593798 DOI: 10.1016/j.stem.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
Advances in modern medicine have enabled a rapid increase in lifespan and, consequently, have highlighted the immune system as a key driver of age-related disease. Immune regeneration therapies present exciting strategies to address age-related diseases by rebooting the host's primary lymphoid tissues or rebuilding the immune system directly via biomaterials or artificial tissue. Here, we identify important, unanswered questions regarding the safety and feasibility of these therapies. Further, we identify key design parameters that should be primary considerations guiding technology design, including timing of application, interaction with the host immune system, and functional characterization of the target patient population.
Collapse
Affiliation(s)
- Laura N Stankiewicz
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
49
|
Tsitkov S, Valentine K, Kozareva V, Donde A, Frank A, Lei S, E Van Eyk J, Finkbeiner S, Rothstein JD, Thompson LM, Sareen D, Svendsen CN, Fraenkel E. Disease related changes in ATAC-seq of iPSC-derived motor neuron lines from ALS patients and controls. Nat Commun 2024; 15:3606. [PMID: 38697975 PMCID: PMC11066062 DOI: 10.1038/s41467-024-47758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), like many other neurodegenerative diseases, is highly heritable, but with only a small fraction of cases explained by monogenic disease alleles. To better understand sporadic ALS, we report epigenomic profiles, as measured by ATAC-seq, of motor neuron cultures derived from a diverse group of 380 ALS patients and 80 healthy controls. We find that chromatin accessibility is heavily influenced by sex, the iPSC cell type of origin, ancestry, and the inherent variance arising from sequencing. Once these covariates are corrected for, we are able to identify ALS-specific signals in the data. Additionally, we find that the ATAC-seq data is able to predict ALS disease progression rates with similar accuracy to methods based on biomarkers and clinical status. These results suggest that iPSC-derived motor neurons recapitulate important disease-relevant epigenomic changes.
Collapse
Affiliation(s)
- Stanislav Tsitkov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey Valentine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Velina Kozareva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aneesh Donde
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steve Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Dhruv Sareen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
50
|
Tadount F, Kiely M, Assi A, Rafferty E, Sadarangani M, MacDonald SE, Quach C. Sex Differences in the Immunogenicity and Efficacy of Seasonal Influenza Vaccines: A Meta-analysis of Randomized Controlled Trials. Open Forum Infect Dis 2024; 11:ofae222. [PMID: 38737434 PMCID: PMC11088355 DOI: 10.1093/ofid/ofae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/19/2024] [Indexed: 05/14/2024] Open
Abstract
Background Sex impacts individuals' response to vaccination. However, most vaccine studies do not report these differences disaggregated by sex. The aim of this study was to assess sex differences in the immunogenicity and efficacy of influenza vaccine. Methods We performed a meta-analysis using phase 3 randomized controlled trial data conducted between 2010 and 2018. Using hemagglutination inhibition antibody titers for each strain, differences in geometric mean ratios (GMRs) were calculated by sex. Risk ratios (RRs) comparing seroconversion proportions were pooled for females and males using random-effects models. Vaccine efficacy (VE) was assessed. Data were analyzed by age group (18-64 vs ≥65 years). Results A total of 33 092 healthy adults from 19 studies were included for immunogenicity analysis, and 6740 from 1 study for VE. Whereas no sex differences in immunogenicity were found in adults <65 years old, older females had a significantly greater chance to seroconvert compared to older males for all strains: RRH1N1 = 1.17 [95% confidence interval {CI}, 1.12-1.23]; RRH3N2 = 1.09 [95% CI, 1.05-1.14]; RRVictoria = 1.23 [95% CI, 1.14-1.31]; RRYamagata = 1.22 [95% CI, 1.14-1.30]. GMRs were also higher in older females for all strains compared to older males. VE in preventing laboratory-confirmed influenza was higher in older females compared to older males with VEs of 27.32% (95% CI, 1.15%-46.56%) and 6.06% (95% CI, -37.68% to 35.90%), respectively. Conclusions Our results suggest a higher immunogenicity and VE in females compared to males in older adults. These differences in immunogenicity and VE support the disaggregation of vaccine data by sex in clinical trials and observational studies. Clinical Trials Registration CRD42018112260.
Collapse
Affiliation(s)
- Fazia Tadount
- Sainte-Justine Hospital Health and Research Center, Montreal, Canada
- Département de Microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montreal, Canada
| | - Marilou Kiely
- Département de Microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montreal, Canada
- Institut national de santé publique du Québec, Québec, Canada
| | - Ali Assi
- Faculty of Nursing and School of Public Health, University of Alberta, Edmonton, Canada
| | - Ellen Rafferty
- Faculty of Nursing and Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Shannon E MacDonald
- Faculty of Nursing and School of Public Health, University of Alberta, Edmonton, Canada
| | - Caroline Quach
- Sainte-Justine Hospital Health and Research Center, Montreal, Canada
- Département de Microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montreal, Canada
- Département de Pédiaterie, Faculté de médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|