51
|
Kashima Y, Mizutani T, Nakayama-Hosoya K, Moriyama S, Matsumura T, Yoshimura Y, Sasaki H, Horiuchi H, Miyata N, Miyazaki K, Tachikawa N, Takahashi Y, Suzuki T, Sugano S, Matano T, Kawana-Tachikawa A, Suzuki Y. Multimodal single-cell analyses of peripheral blood mononuclear cells of COVID-19 patients in Japan. Sci Rep 2023; 13:1935. [PMID: 36732528 PMCID: PMC9893982 DOI: 10.1038/s41598-023-28696-9] [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: 09/06/2022] [Accepted: 01/22/2023] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2 continues to spread worldwide. Patients with COVID-19 show distinct clinical symptoms. Although many studies have reported various causes for the diversity of symptoms, the underlying mechanisms are not fully understood. Peripheral blood mononuclear cells from COVID-19 patients were collected longitudinally, and single-cell transcriptome and T cell receptor repertoire analysis was performed. Comparison of molecular features and patients' clinical information revealed that the proportions of cells present, and gene expression profiles differed significantly between mild and severe cases; although even among severe cases, substantial differences were observed among the patients. In one severely-infected elderly patient, an effective antibody response seemed to have failed, which may have caused prolonged viral clearance. Naïve T cell depletion, low T cell receptor repertoire diversity, and aberrant hyperactivation of most immune cell subsets were observed during the acute phase in this patient. Through this study, we provided a better understanding of the diversity of immune landscapes and responses. The information obtained from this study can help medical professionals develop personalized optimal clinical treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yukie Kashima
- Laboratory of Functional Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan
| | - Taketoshi Mizutani
- Laboratory of Functional Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan
| | | | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Yoshimura
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Hiroaki Sasaki
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Hiroshi Horiuchi
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Nobuyuki Miyata
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Kazuhito Miyazaki
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Natsuo Tachikawa
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Kanagawa, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sumio Sugano
- Institute of Kashiwa-No-Ha Omics Gate, Kashiwa, Chiba, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan.,Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.,Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan.,Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.,Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yutaka Suzuki
- Laboratory of Functional Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan.
| |
Collapse
|
52
|
Buondonno I, Sassi F, Cattaneo F, D’Amelio P. Association between Immunosenescence, Mitochondrial Dysfunction and Frailty Syndrome in Older Adults. Cells 2022; 12:cells12010044. [PMID: 36611837 PMCID: PMC9818926 DOI: 10.3390/cells12010044] [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: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Aging is associated with changes in the immune system, increased inflammation and mitochondrial dysfunction. The relationship between these phenomena and the clinical phenotype of frailty is unclear. Here, we evaluated the immune phenotypes, T cell functions and mitochondrial functions of immune cells in frail and robust older subjects. We enrolled 20 frail subjects age- and gender-matched with 20 robust controls, and T cell phenotype, response to immune stimulation, cytokine production and immune cell mitochondrial function were assessed. Our results showed that numbers of CD4+ and CD8+ T cells were decreased in frail subjects, without impairment to their ratios. Memory and naïve T cells were not significantly affected by frailty, whereas the expression of CD28 but not that of ICOS was decreased in T cells from frail subjects. T cells from robust subjects produced more IL-17 after CD28 stimulation. Levels of serum cytokines were similar in frail subjects and controls. Mitochondrial bioenergetics and ATP levels were significantly lower in immune cells from frail subjects. In conclusion, we suggest that changes in T cell profiles are associated with aging rather than with frailty syndrome; however, changes in T cell response to immune stimuli and reduced mitochondrial activity in immune cells may be considered hallmarks of frailty.
Collapse
Affiliation(s)
- Ilaria Buondonno
- Geriatric and Bone Disease Unit, Department of Medical Science, University of Torino, 10126 Torino, Italy
| | - Francesca Sassi
- Geriatric and Bone Disease Unit, Department of Medical Science, University of Torino, 10126 Torino, Italy
| | - Francesco Cattaneo
- Department of Public Health Sciences and Pediatrics, University of Torino, 10126 Torino, Italy
| | - Patrizia D’Amelio
- Geriatric and Bone Disease Unit, Department of Medical Science, University of Torino, 10126 Torino, Italy
- Department of Medicine, Service of Geriatric Medicine & Geriatric Rehabilitation, University of Lausanne Hospital (CHUV), 1011 Lausanne, Switzerland
- Correspondence: ; Tel.: +41-213143712
| |
Collapse
|
53
|
Guaraldi G, Milic J, Cesari M, Leibovici L, Mandreoli F, Missier P, Rozzini R, Cattelan AM, Motta F, Mussini C, Cossarizza A. The interplay of post-acute COVID-19 syndrome and aging: a biological, clinical and public health approach. Ageing Res Rev 2022; 81:101686. [PMID: 35820609 PMCID: PMC9270773 DOI: 10.1016/j.arr.2022.101686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/07/2022] [Indexed: 01/31/2023]
Abstract
The post-acute COVID-19 syndrome (PACS) is characterized by the persistence of fluctuating symptoms over three months from the onset of the possible or confirmed COVID-19 acute phase. Current data suggests that at least 10% of people with previously documented infection may develop PACS, and up to 50-80% of prevalence is reported among survivors after hospital discharge. This viewpoint will discuss various aspects of PACS, particularly in older adults, with a specific hypothesis to describe PACS as the expression of a modified aging trajectory induced by SARS CoV-2. This hypothesis will be argued from biological, clinical and public health view, addressing three main questions: (i) does SARS-CoV-2-induced alterations in aging trajectories play a role in PACS?; (ii) do people with PACS face immuno-metabolic derangements that lead to increased susceptibility to age-related diseases?; (iii) is it possible to restore the healthy aging trajectory followed by the individual before pre-COVID?. A particular focus will be given to the well-being of people with PACS that could be assessed by the intrinsic capacity model and support the definition of the healthy aging trajectory.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy,Department of Infectious Diseases, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy,Correspondence to: Department of Surgical, Medical, Dental and Morphological Sciences University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41124 Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Federica Mandreoli
- Department of Physical, Computer and Mathematical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Missier
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Renzo Rozzini
- Geriatric Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | | | - Federico Motta
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy,Department of Infectious Diseases, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
54
|
Bröde P, Claus M, Gajewski PD, Getzmann S, Golka K, Hengstler JG, Wascher E, Watzl C. Calibrating a Comprehensive Immune Age Metric to Analyze the Cross Sectional Age-Related Decline in Cardiorespiratory Fitness. BIOLOGY 2022; 11:1576. [PMID: 36358277 PMCID: PMC9687950 DOI: 10.3390/biology11111576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/30/2022]
Abstract
Cardiorespiratory fitness (CRF) is essential for sustained work ability in good health, but declines with aging, as does the functionality of the immune system, the latter process commonly referred to as immunosenescence. This study aimed to compare the capacity of immunosenescence biomarkers with chronological age for predicting low CRF in a cross-sectional sample recruited from the regional working population. CRF was determined by submaximal bicycle ergometer testing in a cross-sectional sample of 597 volunteers aged 20-70 years from the 'Dortmund Vital Study' (DVS, ClinicalTrials.gov Identifier: NCT05155397). Low CRF was scored if the ergometer test was not completed due to medical reasons or if the power output projected to a heart rate of 130 bpm divided by body mass was below sex-specific reference values of 1.25 W/kg for females and 1.5 W/kg for males, respectively. In addition to established biomarkers of immunosenescence, we calibrated a comprehensive metric of immune age to our data and compared its predictive capacity for low CRF to chronological age, while adjusting our analysis for the influence of sex, obesity, and the level of regular physical activity, by applying univariate and multiple logistic regression. While obesity, low physical activity, chronological and immune age were all associated with increased probability for low CRF in univariate analyses, multiple logistic regression revealed that obesity and physical activity together with immune age, but not chronological age, were statistically significant predictors of low CRF outcome. Sex was non-significant due to the applied sex-specific reference values. These results demonstrate that biological age assessed by our immunological metric can outperform chronological age as a predictor for CRF and indicate a potential role for immunosenescence in explaining the inter-individual variability of the age-related decline in cardiorespiratory fitness.
Collapse
Affiliation(s)
- Peter Bröde
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, D-44139 Dortmund, Germany
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Hovakimyan A, Zagorski K, Chailyan G, Antonyan T, Melikyan L, Petrushina I, Batt DG, King O, Ghazaryan M, Donthi A, Foose C, Petrovsky N, Cribbs DH, Agadjanyan MG, Ghochikyan A. Immunogenicity of MultiTEP platform technology-based Tau vaccine in non-human primates. NPJ Vaccines 2022; 7:117. [PMID: 36224191 PMCID: PMC9556597 DOI: 10.1038/s41541-022-00544-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Pathological forms of Tau protein are directly associated with neurodegeneration and correlate with Alzheimer's Disease (AD) symptoms, progression, and severity. Previously, using various mouse models of Tauopathies and AD, we have demonstrated the immunogenicity and efficacy of the MultiTEP-based adjuvanted vaccine targeting the phosphatase activating domain (PAD) of Tau, AV-1980R/A. Here, we analyzed its immunogenicity in non-human primates (NHP), the closest phylogenic relatives to humans with a similar immune system, to initiate the transition of this vaccine into clinical trials. We have demonstrated that AV-1980R/A is highly immunogenic in these NHPs, activating a broad but unique to each monkey repertoire of MultiTEP-specific T helper (Th) cells that, in turn, activate B cells specific to PAD. The resulting anti-PAD IgG antibodies recognize pathological Tau tangles and Tau-positive neuritis in AD case brain sections with no staining in control non-AD cases. These published data and efficacy results support the AV-1980R/A vaccine progression to first-in-human clinical trials.
Collapse
Affiliation(s)
- Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Levon Melikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Dash G Batt
- Charles C. Gates manufacturing Facility, University of Colorado/Anschutz Medical Campus, Aurora, CO, USA
| | - Olga King
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Manush Ghazaryan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Aashrit Donthi
- Charles C. Gates manufacturing Facility, University of Colorado/Anschutz Medical Campus, Aurora, CO, USA
| | - Caitlynn Foose
- Charles C. Gates manufacturing Facility, University of Colorado/Anschutz Medical Campus, Aurora, CO, USA
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders Medical Center, Bedford Park, Adelaide, SA, 5042, Australia
- Department of Diabetes and Endocrinology, Faculty of Medicine, Flinders University, Adelaide, SA, 5042, Australia
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| |
Collapse
|
56
|
Marrella V, Facoetti A, Cassani B. Cellular Senescence in Immunity against Infections. Int J Mol Sci 2022; 23:11845. [PMID: 36233146 PMCID: PMC9570409 DOI: 10.3390/ijms231911845] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is characterized by irreversible cell cycle arrest in response to different triggers and an inflammatory secretome. Although originally described in fibroblasts and cell types of solid organs, cellular senescence affects most tissues with advancing age, including the lymphoid tissue, causing chronic inflammation and dysregulation of both innate and adaptive immune functions. Besides its normal occurrence, persistent microbial challenge or pathogenic microorganisms might also accelerate the activation of cellular aging, inducing the premature senescence of immune cells. Therapeutic strategies counteracting the detrimental effects of cellular senescence are being developed. Their application to target immune cells might have the potential to improve immune dysfunctions during aging and reduce the age-dependent susceptibility to infections. In this review, we discuss how immune senescence influences the host's ability to resolve more common infections in the elderly and detail the different markers proposed to identify such senescent cells; the mechanisms by which infectious agents increase the extent of immune senescence are also reviewed. Finally, available senescence therapeutics are discussed in the context of their effects on immunity and against infections.
Collapse
Affiliation(s)
- Veronica Marrella
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, 20138 Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Amanda Facoetti
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Barbara Cassani
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, Università Degli Studi di Milano, 20089 Milan, Italy
| |
Collapse
|
57
|
Scordo JM, Piergallini TJ, Olmo-Fontánez AM, Thomas A, Raué HP, Slifka M, Turner J. Recall responses in the lung environment are impacted by age in a pilot study of Mycobacterium bovis-BCG vaccinated rhesus macaques. Exp Gerontol 2022; 167:111904. [PMID: 35918043 PMCID: PMC9907331 DOI: 10.1016/j.exger.2022.111904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023]
Abstract
Age-related changes in the immune system increase susceptibility to infectious diseases. Vaccines are an important tool to prevent infection or boost immunological memory; however, vaccines are less effective in aged individuals. In order to protect our aging population from the threat of infectious diseases, we must gain a better understanding of age-related alterations in the immune response at the site of infection. The lung is one site of frequent infection in older individuals. In this study, we expanded on our previous work to study vaccine-induced immune responses in the local lung environment in a pilot study of aged rhesus macaques. To do this, we developed an in vivo model to probe recall responses to tuberculin challenge in the lungs 8 weeks and 16 weeks post-Mycobacterium bovis BCG vaccination by performing targeted bronchoalveolar lavages. In parallel, we determined peripheral blood responses in vaccinated animals to compare systemic and local tissue responses to tuberculin challenge. We found that following lung tuberculin challenge 8 weeks post-vaccination, aged animals had reduced T cell responses, particularly within the CD8+ T cell compartment. Aged animals had decreased CD8+ effector and memory T cell recall responses and less activated CD8+ T cells. This diminished lung CD8+ T cell response in aged animals was maintained over time. Despite changes in the CD8+ T cell compartment, lung CD4+ T cell responses were similar between age groups. In the peripheral blood, we observed age-related changes in immune cell populations and plasma levels of immune mediators that were present prior to vaccination. Lastly, we found that peripheral blood mononuclear cells from aged BCG-vaccinated animals were functional in their response to antigen stimulation, behaving in a similar manner to those from their adult counterparts. These systemic observations were similar to those found in our previous study of BCG-vaccinated baboons, supporting the notion that tissue immune responses, and not systemic responses, to vaccination and challenge are impaired with age. These findings expand on our previous work to show that in addition to the skin, age-related changes in the lung environment impact recall immune responses to vaccination and challenge. The impact of age on local tissue responses to infectious challenge should be accounted for in the development of therapeutics or medical interventions aimed at boosting immune recall responses of aged individuals.
Collapse
Affiliation(s)
- Julia M. Scordo
- Texas Biomedical Institute, San Antonio, TX,The University of Texas Health Science Center of San Antonio, San Antonio, TX
| | | | - Angélica M. Olmo-Fontánez
- Texas Biomedical Institute, San Antonio, TX,The University of Texas Health Science Center of San Antonio, San Antonio, TX
| | | | | | - Mark Slifka
- Oregon Health and Science University, Portland, OR
| | - Joanne Turner
- Texas Biomedical Institute, San Antonio, TX, United States of America.
| |
Collapse
|
58
|
Liisborg C. Age-related macular degeneration and myeloproliferative neoplasms - A common pathway. Acta Ophthalmol 2022; 100 Suppl 271:3-35. [PMID: 36200281 PMCID: PMC9828081 DOI: 10.1111/aos.15247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/22/2021] [Indexed: 01/12/2023]
Abstract
DANSK RESUMÉ (DANISH SUMMARY): Aldersrelateret makuladegeneration (AMD) er den hyppigste årsag til uopretteligt synstab og blindhed i højindkomstlande. Det er en progredierende nethindesygdom som gradvist fører til ødelaeggelse af de celler som er ansvarlige for vores centralsyn. De tidlige stadier er ofte asymptomatiske, imens senstadie AMD, som opdeles i to former, neovaskulaer AMD (nAMD) og geografisk atrofi (GA), begge udviser gradvist synstab, dog generelt med forskellig hastighed. Tidlig AMD er karakteriseret ved tilstedevaerelsen af druser og pigmentforandringer i nethinden mens nAMD og GA udviser henholdsvis karnydannelse i og atrofi af nethinden. AEtiologien er multifaktoriel og udover alder omfatter patogenesen miljø- og genetiske risikofaktorer. Forskning har specielt fokuseret på lokale forandringer i øjet hvor man har fundet at inflammation spiller en betydelig rolle for udviklingen af sygdommen, men flere studier tyder også på at systemiske forandringer og specielt systemisk inflammation spiller en vaesentlig rolle i patogenesen. De Philadelphia-negative myeloproliferative neoplasier (MPNs) er en gruppe af haematologiske kraeftsygdomme med en erhvervet genetisk defekt i den tidlige pluripotente stamcelle som medfører en overproduktion af en eller flere af blodets modne celler. Sygdommene er fundet at udvikle sig i et biologisk kontinuum fra tidligt cancerstadie, essentiel trombocytose (ET) over polycytaemi vera (PV) og endelig til det sene myelofibrose stadie (PMF). Symptomer hos disse patienter skyldes isaer den aendrede sammensaetning af blodet, hyperviskositet, kompromitteret mikrocirkulation og nedsat vaevsgennemblødning. Den øgede morbiditet og mortalitet beror i høj grad på tromboembolier, blødninger og leukemisk transformation. En raekke mutationer som driver MPN sygdommene er identificeret, bl.a. JAK2V617F-mutationen som medfører en deregulering JAK/STAT signalvejen, der bl.a. har betydning for cellers vaekst og overlevelse. Et tidligere stort registerstudie har vist at patienter med MPNs har en øget risiko for neovaskulaer AMD og et pilotstudie har vist øget forekomst af intermediaer AMD. Dette ønsker vi at undersøge naermere i et større studie i dette Ph.d.- projekt. Flere studier har også vist at kronisk inflammation spiller en vigtig rolle for både initiering og udvikling af den maligne celleklon hos MPNs og herfra er en "Human Inflammationsmodel" blevet udviklet. Siden er MPN sygdommene blevet anvendt som "model sygdomme" for en tilsvarende inflammationsmodel for udvikling af Alzheimers sygdom. I dette Ph.d.-projekt vil vi tilsvarende forsøge at undersøge systemisk inflammation i forhold til forekomst af druser. Det vil vi gøre ved at sammenligne systemiske immunologiske markører som tidligere er undersøgt hos patienter med AMD og sammenligne med MPN. Specielt er vi interesseret i systemiske immunologiske forskelle på patienter med MPN og druser (MPNd) og MPN med normale nethinder (MPNn). Denne afhandling består af to overordnede studier. I Studie I, undersøgte vi forekomsten af retinale forandringer associeret med AMD hos 200 patienter med MPN (artikel I). Studie II, omhandlede immunologiske ligheder ved AMD og MPN, og var opdelt i yderligere tre delstudier hvor vi undersøgte hhv. systemiske markører for inflammation, aldring og angiogenese (artikel II, III og IV). Vi undersøgte markørerne i fire typer af patienter: nAMD, intermediaer AMD (iAMD), MPNd og MPNn. Undersøgelsen af forskelle mellem MPNd og MPNn, vil gøre det muligt at identificere forandringer i immunsystemet som kunne vaere relevante for AMD-patogenesen. Vi vil endvidere sammenholde resultaterne for patienter med MPN med patienter som har iAMD og nAMD. I studie I (Artikel I) fandt vi at patienter med MPN har en signifikant højere praevalens af store druser og AMD tidligere i livet sammenlignet med estimater fra tre store befolkningsundersøgelser. Vi fandt også at forekomst af druser var associeret med højere neutrofil-lymfocyt ratio, hvilket indikerer et højere niveau af kronisk inflammation i patienterne med druser sammenlignet med dem uden druser. I studie II (Artikel II, III og IV) fandt vi flere immunologiske forskelle mellem patienter med MPNd og MPNn. Da vi undersøgte markører for inflammation, fandt vi en højere grad af systemisk inflammation i MPNd end MPNn. Dette blev vist ved en højere inflammationsscore (udregnet på baggrund af niveauer af pro-inflammatoriske markører), en højere neutrofil-lymfocyt ratio, samt indikationer på et dereguleret komplementsystem. Ved undersøgelse af aldringsmarkører fandt vi tegn på accelereret immunaldring hos MPNd i forhold til MPNn, hvilket kommer til udtryk ved en større procentdel af "effector memory T celler". Endelig fandt vi en vaesentlig lavere ekspression af CXCR3 på T celler og monocytter hos patienter med nAMD sammenlignet med iAMD, MPNd og MPNn. Dette er i overensstemmelse med tidligere studier hvor CXCR3 ekspression er fundet lavere end hos raske kontroller. Derudover fandt vi en faldende CXCR3 ekspression på monocytter over det biologiske MPN-kontinuum. Disse studier indikerer en involvering af CXCR3 i både nAMD og PMF, begge sygdomsstadier som er karakteriseret ved angiogenese og fibrose. Ud fra resultaterne af denne afhandling kan vi konkludere at forekomsten af druser og AMD hos MPN er øget i forhold til baggrundsbefolkningen. Endvidere viser vores resultater at systemisk inflammation muligvis spiller en vaesentlig større rolle i udviklingen af AMD end tidligere antaget. Vi foreslår derfor en AMD-model (Figur 18) hvor inflammation kan initiere og accelerere den normale aldersafhaengige akkumulation af affaldsstoffer i nethinden, som senere udvikler sig til druser, medførende øget lokal inflammation og med tiden tidlig og intermediaer AMD. Dette resulterer i den øgede risiko for udvikling til de invaliderende senstadier af AMD. ENGLISH SUMMARY: Age-related macular degeneration (AMD) is the most common cause of irreversible vision loss and blindness in high-income countries. It is a progressive retinal disease leading to damage of the cells responsible for central vision. The early stages of the disease are often asymptomatic, while late-stage AMD, which is divided into two entities, neovascular AMD and geographic atrophy (GA), both show vision loss, though generally with different progression rates. Drusen and pigmentary abnormalities in the retina characterise early AMD, while nAMD and GA show angiogenesis in and atrophy of the retina, respectively. The aetiology is multifactorial and, in addition to ageing, which is the most significant risk factor for developing AMD, environmental- and genetic risk factors are implicated in the pathogenesis. Research has focused on local changes in the eye where inflammation has been found to play an essential role, but studies also point to systemic alterations and especially systemic inflammation to be involved in the pathogenesis. The Philadelphia-negative myeloproliferative neoplasms (MPN) are a group of haematological cancers with an acquired genetic defect of the pluripotent haematopoietic stem cell, characterised by excess haematopoiesis of the myeloid cell lineage. The diseases have been found to evolve in a biological continuum from early cancer state, essential thrombocythemia, over polycythaemia vera (PV), to the advanced myelofibrosis stage (PMF). The symptoms in these patients are often a result of the changes in the blood composition, hyperviscosity, microvascular disturbances, and reduced tissue perfusion. The major causes of morbidity and mortality are thromboembolic- and haemorrhagic events, and leukemic transformation. A group of mutations that drive the MPNs has been identified, e.g., the JAK2V617F mutation, which results in deregulation of the JAK/STAT signal transduction pathway important, for instance, in cell differentiation and survival. A previous large register study has shown that patients with MPNs have an increased risk of neovascular AMD, and a pilot study has shown an increased prevalence of intermediate AMD. We wish to study this further in a larger scale study. Several studies have also shown that systemic inflammation plays an essential role in both the initiation and progression of the malignant cell clone in MPNs. From this knowledge, a "Human inflammation model" has been developed. Since then, the MPNs has been used as model diseases for a similar inflammation model for the development of Alzheimer's disease. In this PhD project, we would like to investigate systemic inflammation in relation to drusen presence. We will do this by comparing systemic immunological markers previously investigated in patients with AMD and compare with MPN. We are primarily interested in systemic immunological differences between patients with MPN and drusen (MPNd) and MPN with normal retinas (MPNn). This thesis consists of two main studies. Study I investigated the prevalence of retinal changes associated with AMD and the prevalence of different AMD stages in 200 patients with MPN (paper I). Study II examined immunological similarities between AMD and MPNs. This study was divided into three substudies exploring systemic markers of inflammation, ageing and angiogenesis, respectively. This was done in four types of patients: nAMD, intermediate AMD (iAMD), MPNd and MPNn. Investigating, differences between MPNd and MPNn, will make it possible to identify changes in the immune system, relevant for AMD pathogenesis. Additionally, we will compare patients with MPNs with patients with iAMD and nAMD. In study I (Paper I), we found that patients with MPNs have a significantly higher prevalence of large drusen and consequently AMD from an earlier age compared to the estimates from three large population-based studies. We also found that drusen prevalence was associated with a higher neutrophil-to-lymphocyte ratio indicating a higher level of chronic low-grade inflammation in patients with drusen compared to those without drusen. In study II (papers II, III and IV), we found immunological differences between patients with MPNd and MPNn. When we investigated markers of inflammation, we found a higher level of systemic inflammation in MPNd than MPNn. This was indicated by a higher inflammation score (based on levels of pro-inflammatory markers), a higher neutrophil-to-lymphocyte ratio, and indications of a deregulated complement system. When examining markers of ageing, we found signs of accelerated immune ageing in MPNd compared to MPNn, shown by more senescent effector memory T cells. Finally, when exploring a marker of angiogenesis, we found a lower CXCR3 expression on monocytes and T cells in nAMD compared to iAMD, MPNd and MPNn, in line with previous studies of nAMD compared to healthy controls. Further, we found decreasing CXCR3 expression over the MPN biological continuum. These studies indicate CXCR3 involvement in both nAMD and PMF, two disease stages characterised by angiogenesis and fibrosis. From the results of this PhD project, we can conclude that the prevalence of drusen and AMD is increased in patients with MPN compared to the general population. Further, our results show that systemic inflammation may play a far more essential role in AMD pathogenesis than previously anticipated. We, therefore, propose an AMD model (Figure 18) where inflammation can initiate and accelerate the normal age-dependent accumulation of debris in the retina, which later evolve into drusen, resulting in increased local inflammation, and over time early- and intermediate AMD. This results in the increased risk of developing the late debilitating stages of AMD.
Collapse
|
59
|
Hernández-Silva D, Cantón-Sandoval J, Martínez-Navarro FJ, Pérez-Sánchez H, de Oliveira S, Mulero V, Alcaraz-Pérez F, Cayuela ML. Senescence-Independent Anti-Inflammatory Activity of the Senolytic Drugs Dasatinib, Navitoclax, and Venetoclax in Zebrafish Models of Chronic Inflammation. Int J Mol Sci 2022; 23:ijms231810468. [PMID: 36142384 PMCID: PMC9499634 DOI: 10.3390/ijms231810468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Telomere shortening is the main molecular mechanism of aging, but not the only one. The adaptive immune system also ages, and older organisms tend to develop a chronic pro-inflammatory status with low-grade inflammation characterized by chronic activation of the innate immune system, called inflammaging. One of the main stimuli that fuels inflammaging is a high nutrient intake, triggering a metabolic inflammation process called metainflammation. In this study, we report the anti-inflammatory activity of several senolytic drugs in the context of chronic inflammation, by using two different zebrafish models: (i) a chronic skin inflammation model with a hypomorphic mutation in spint1a, the gene encoding the serine protease inhibitor, kunitz-type, 1a (also known as hai1a) and (ii) a non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH) model with inflammation induced by a high-fat diet. Our results show that, although these models do not manifest premature aging, the senolytic drugs dasatinib, navitoclax, and venetoclax have an anti-inflammatory effect that results in the amelioration of chronic inflammation.
Collapse
Affiliation(s)
- David Hernández-Silva
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Joaquín Cantón-Sandoval
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Francisco Juan Martínez-Navarro
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Victoriano Mulero
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Francisca Alcaraz-Pérez
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
- Correspondence: (F.A.-P.); (M.L.C.); Tel.: +34-968395328 (M.L.C.)
| | - María Luisa Cayuela
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
- Correspondence: (F.A.-P.); (M.L.C.); Tel.: +34-968395328 (M.L.C.)
| |
Collapse
|
60
|
Cunha LL, Valsecchi VADS, Ward LS. Investigating population-level immunosenescence: From bench to bedside. Front Immunol 2022; 13:949928. [PMID: 36059504 PMCID: PMC9428264 DOI: 10.3389/fimmu.2022.949928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response is remodeled with aging in a process called immunosenescence. Some immunologists conceive immunosenescence as an adaptation of immunity to the aged immune-environment rather than a merely collapsed reactivity of immune cells against microbes and tumor cells. Others believe on an uninterrupted activation of the innate immune system with aging, leading to a low grade, sterile and chronic proinflammatory state called inflammaging. For instance, it is possible that chronic infection by cytomegalovirus leads to persistent production of viral load. This phenomenon offers periodic stimuli to the immune system that ultimately contribute to the remodeling of the immune response. If investigating immunosenescence at the cellular level is already a difficult task, considering the population level is much more complex. However, by studying immunosenescence at the population level, we can extract valuable results with viable applications. While studies with animal models allow scientists to deepen their understanding of the mechanisms of immunosenescence, studying large populations can bring practical innovations to medicine and the health system. Many researchers and funders have dedicated themselves to producing methods for the evaluation of immunosenescence on a large scale, aiming to elucidate new mechanisms by which diseases are established in the elderly. The description of how the immune response is remodeled with aging emerges as a new tool to identify the subset of subjects in which unhealthy aging is a matter of time, to help better individualize clinical management and select patients who may benefit. of early interventions. This review focuses on functional assays as valuable methods for measuring the remodeling of the immune response with aging and discuss their clinical impact. We also recall fundamental concepts for understanding the aging process of the immune response. In addition, we highlight future prospects for immunosenescence research.
Collapse
Affiliation(s)
- Lucas Leite Cunha
- Laboratory of Molecular and Translational Endocrinology, Division of Endocrinology, Federal University of São Paulo, São Paulo, Brazil
- Discipline of Internal Medicine and Laboratory Medicine, Federal University of São Paulo, São Paulo, Brazil
- *Correspondence: Lucas Leite Cunha,
| | - Victor Alexandre dos Santos Valsecchi
- Laboratory of Molecular and Translational Endocrinology, Division of Endocrinology, Federal University of São Paulo, São Paulo, Brazil
- Discipline of Internal Medicine and Laboratory Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Laura Sterian Ward
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| |
Collapse
|
61
|
Bracci PM, Rice T, Hansen HM, Francis SS, Lee S, McCoy LS, Shrestha PP, Warrier G, Clarke JL, Molinaro AM, Taylor JW, Wiencke JK, Wrensch MR. Pre-surgery immune profiles of adult glioma patients. J Neurooncol 2022; 159:103-115. [PMID: 35716311 PMCID: PMC9325836 DOI: 10.1007/s11060-022-04047-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/24/2022] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Although immunosuppression is a known characteristic of glioma, no previous large studies have reported peripheral blood immune cell profiles prior to patient surgery and chemoradiation. This report describes blood immune cell characteristics and associated variables prior to surgery among typical glioma patients seen at a large University practice. METHODS We analyzed pre-surgery blood samples from 139 glioma patients diagnosed with a new or recurrent grade II/III glioma (LrGG, n = 64) or new glioblastoma (GBM, n = 75) and 454 control participants without glioma. Relative cell fractions of CD4, CD8, B-cells, Natural Killer cells, monocytes, and neutrophils, were estimated via a validated deconvolution algorithm from blood DNA methylation measures from Illumina EPIC arrays. RESULTS Dexamethasone use at time of blood draw varied by glioma type being highest among patients with IDH wild-type (wt) GBM (75%) and lowest for those with oligodendroglioma (14%). Compared to controls, glioma patients showed statistically significant lower cell fractions for all immune cell subsets except for neutrophils which were higher (all p-values < 0.001), in part because of the higher prevalence of dexamethasone use at time of blood draw for IDHwt GBM. Patients who were taking dexamethasone were more likely to have a low CD4 count (< 200, < 500), increased neutrophils, low absolute lymphocyte counts, higher total cell count and higher NLR. CONCLUSION We show that pre-surgery blood immune profiles vary by glioma subtype, age, and more critically, by use of dexamethasone. Our results highlight the importance of considering dexamethasone exposures in all studies of immune profiles and of obtaining immune measures prior to use of dexamethasone, if possible.
Collapse
Affiliation(s)
- Paige M Bracci
- Department of Epidemiology and Biostatistics, UCSF, 1450 3rd Street, San Francisco, CA, 94158, USA.
| | - Terri Rice
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | | | - Sean Lee
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Lucie S McCoy
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Pavan P Shrestha
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurology, UCSF, San Francisco, CA, USA
| | - Annette M Molinaro
- Department of Epidemiology and Biostatistics, UCSF, 1450 3rd Street, San Francisco, CA, 94158, USA
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurology, UCSF, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | | |
Collapse
|
62
|
Kwok T, Medovich SC, Silva-Junior IA, Brown EM, Haug JC, Barrios MR, Morris KA, Lancaster JN. Age-Associated Changes to Lymph Node Fibroblastic Reticular Cells. FRONTIERS IN AGING 2022; 3:838943. [PMID: 35821826 PMCID: PMC9261404 DOI: 10.3389/fragi.2022.838943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
The decreased proportion of antigen-inexperienced, naïve T cells is a hallmark of aging in both humans and mice, and contributes to reduced immune responses, particularly against novel and re-emerging pathogens. Naïve T cells depend on survival signals received during their circulation among the lymph nodes by direct contacts with stroma, in particular fibroblastic reticular cells. Macroscopic changes to the architecture of the lymph nodes have been described, but it is unclear how lymph node stroma are altered with age, and whether these changes contribute to reduced naïve T cell maintenance. Here, using 2-photon microscopy, we determined that the aged lymph node displayed increased fibrosis and correspondingly, that naïve T-cell motility was impaired in the aged lymph node, especially in proximity to fibrotic deposition. Functionally, adoptively transferred young naïve T-cells exhibited reduced homeostatic turnover in aged hosts, supporting the role of T cell-extrinsic mechanisms that regulate their survival. Further, we determined that early development of resident fibroblastic reticular cells was impaired, which may correlate to the declining levels of naïve T-cell homeostatic factors observed in aged lymph nodes. Thus, our study addresses the controversy as to whether aging impacts the composition lymph node stroma and supports a model in which impaired differentiation of lymph node fibroblasts and increased fibrosis inhibits the interactions necessary for naïve T cell homeostasis.
Collapse
Affiliation(s)
- Tina Kwok
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | | | - Elise M Brown
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Joel C Haug
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | - Karina A Morris
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | |
Collapse
|
63
|
Bazioti V, La Rose AM, Maassen S, Bianchi F, de Boer R, Halmos B, Dabral D, Guilbaud E, Flohr-Svendsen A, Groenen AG, Marmolejo-Garza A, Koster MH, Kloosterhuis NJ, Havinga R, Pranger AT, Langelaar-Makkinje M, de Bruin A, van de Sluis B, Kohan AB, Yvan-Charvet L, van den Bogaart G, Westerterp M. T cell cholesterol efflux suppresses apoptosis and senescence and increases atherosclerosis in middle aged mice. Nat Commun 2022; 13:3799. [PMID: 35778407 PMCID: PMC9249754 DOI: 10.1038/s41467-022-31135-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease driven by hypercholesterolemia. During aging, T cells accumulate cholesterol, potentially affecting inflammation. However, the effect of cholesterol efflux pathways mediated by ATP-binding cassette A1 and G1 (ABCA1/ABCG1) on T cell-dependent age-related inflammation and atherosclerosis remains poorly understood. In this study, we generate mice with T cell-specific Abca1/Abcg1-deficiency on the low-density-lipoprotein-receptor deficient (Ldlr-/-) background. T cell Abca1/Abcg1-deficiency decreases blood, lymph node, and splenic T cells, and increases T cell activation and apoptosis. T cell Abca1/Abcg1-deficiency induces a premature T cell aging phenotype in middle-aged (12-13 months) Ldlr-/- mice, reflected by upregulation of senescence markers. Despite T cell senescence and enhanced T cell activation, T cell Abca1/Abcg1-deficiency decreases atherosclerosis and aortic inflammation in middle-aged Ldlr-/- mice, accompanied by decreased T cells in atherosclerotic plaques. We attribute these effects to T cell apoptosis downstream of T cell activation, compromising T cell functionality. Collectively, we show that T cell cholesterol efflux pathways suppress T cell apoptosis and senescence, and induce atherosclerosis in middle-aged Ldlr-/- mice.
Collapse
Affiliation(s)
- Venetia Bazioti
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5252.00000 0004 1936 973XInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Anouk M. La Rose
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Sjors Maassen
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Frans Bianchi
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Rinse de Boer
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Benedek Halmos
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Deepti Dabral
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Emma Guilbaud
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Arthur Flohr-Svendsen
- grid.4494.d0000 0000 9558 4598European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Anouk G. Groenen
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alejandro Marmolejo-Garza
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Mirjam H. Koster
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Rick Havinga
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alle T. Pranger
- grid.4494.d0000 0000 9558 4598Laboratory of Medicine, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alain de Bruin
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5477.10000000120346234Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Bart van de Sluis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alison B. Kohan
- grid.21925.3d0000 0004 1936 9000Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Laurent Yvan-Charvet
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Geert van den Bogaart
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Marit Westerterp
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| |
Collapse
|
64
|
Del Cuore A, Pacinella G, Riolo R, Tuttolomondo A. The Role of Immunosenescence in Cerebral Small Vessel Disease: A Review. Int J Mol Sci 2022; 23:ijms23137136. [PMID: 35806140 PMCID: PMC9266569 DOI: 10.3390/ijms23137136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the most important causes of vascular dementia. Immunosenescence and inflammatory response, with the involvement of the cerebrovascular system, constitute the basis of this disease. Immunosenescence identifies a condition of deterioration of the immune organs and consequent dysregulation of the immune response caused by cellular senescence, which exposes older adults to a greater vulnerability. A low-grade chronic inflammation status also accompanies it without overt infections, an “inflammaging” condition. The correlation between immunosenescence and inflammaging is fundamental in understanding the pathogenesis of age-related CSVD (ArCSVD). The production of inflammatory mediators caused by inflammaging promotes cellular senescence and the decrease of the adaptive immune response. Vice versa, the depletion of the adaptive immune mechanisms favours the stimulation of the innate immune system and the production of inflammatory mediators leading to inflammaging. Furthermore, endothelial dysfunction, chronic inflammation promoted by senescent innate immune cells, oxidative stress and impairment of microglia functions constitute, therefore, the framework within which small vessel disease develops: it is a concatenation of molecular events that promotes the decline of the central nervous system and cognitive functions slowly and progressively. Because the causative molecular mechanisms have not yet been fully elucidated, the road of scientific research is stretched in this direction, seeking to discover other aberrant processes and ensure therapeutic tools able to enhance the life expectancy of people affected by ArCSVD. Although the concept of CSVD is broader, this manuscript focuses on describing the neurobiological basis and immune system alterations behind cerebral aging. Furthermore, the purpose of our work is to detect patients with CSVD at an early stage, through the evaluation of precocious MRI changes and serum markers of inflammation, to treat untimely risk factors that influence the burden and the worsening of the cerebral disease.
Collapse
Affiliation(s)
- Alessandro Del Cuore
- Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialised Medicine (PROMISE) G. D’Alessandro, University of Palermo, 90133 Palermo, Italy; (G.P.); (R.R.); (A.T.)
- Internal Medicine and Stroke Care Ward, Policlinico “P. Giaccone”, 90127 Palermo, Italy
- Correspondence: ; Tel.: +39-091-655-2197
| | - Gaetano Pacinella
- Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialised Medicine (PROMISE) G. D’Alessandro, University of Palermo, 90133 Palermo, Italy; (G.P.); (R.R.); (A.T.)
- Internal Medicine and Stroke Care Ward, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Renata Riolo
- Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialised Medicine (PROMISE) G. D’Alessandro, University of Palermo, 90133 Palermo, Italy; (G.P.); (R.R.); (A.T.)
- Internal Medicine and Stroke Care Ward, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialised Medicine (PROMISE) G. D’Alessandro, University of Palermo, 90133 Palermo, Italy; (G.P.); (R.R.); (A.T.)
- Internal Medicine and Stroke Care Ward, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
65
|
Sun X, Nguyen T, Achour A, Ko A, Cifello J, Ling C, Sharma J, Hiroi T, Zhang Y, Chia CW, Wood Iii W, Wu WW, Zukley L, Phue JN, Becker KG, Shen RF, Ferrucci L, Weng NP. Longitudinal analysis reveals age-related changes in the T cell receptor repertoire of human T cell subsets. J Clin Invest 2022; 132:158122. [PMID: 35708913 PMCID: PMC9433102 DOI: 10.1172/jci158122] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
A diverse T cell receptor (TCR) repertoire is essential for protection against a variety of pathogens, and TCR repertoire size is believed to decline with age. However, the precise size of human TCR repertoires, in both total and subsets of T cells, as well as their changes with age, are not fully characterized. We conducted a longitudinal analysis of the human blood TCRα and TCRβ repertoire of CD4+ and CD8+ T cell subsets using a unique molecular identifier–based (UMI-based) RNA-seq method. Thorough analysis of 1.9 × 108 T cells yielded the lower estimate of TCR repertoire richness in an adult at 3.8 × 108. Alterations of the TCR repertoire with age were observed in all 4 subsets of T cells. The greatest reduction was observed in naive CD8+ T cells, while the greatest clonal expansion was in memory CD8+ T cells, and the highest increased retention of TCR sequences was in memory CD8+ T cells. Our results demonstrated that age-related TCR repertoire attrition is subset specific and more profound for CD8+ than CD4+ T cells, suggesting that aging has a more profound effect on cytotoxic as opposed to helper T cell functions. This may explain the increased susceptibility of older adults to novel infections.
Collapse
Affiliation(s)
- Xiaoping Sun
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Thomas Nguyen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Achouak Achour
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Annette Ko
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jeffrey Cifello
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jay Sharma
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Toyoko Hiroi
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Yongqing Zhang
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chee W Chia
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, United States of America
| | - William Wood Iii
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Wells W Wu
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Linda Zukley
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Je-Nie Phue
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Kevin G Becker
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Nan-Ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| |
Collapse
|
66
|
Hirai T, Yoshioka Y. Considerations of CD8+ T Cells for Optimized Vaccine Strategies Against Respiratory Viruses. Front Immunol 2022; 13:918611. [PMID: 35774782 PMCID: PMC9237416 DOI: 10.3389/fimmu.2022.918611] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The primary goal of vaccines that protect against respiratory viruses appears to be the induction of neutralizing antibodies for a long period. Although this goal need not be changed, recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have drawn strong attention to another arm of acquired immunity, CD8+ T cells, which are also called killer T cells. Recent evidence accumulated during the coronavirus disease 2019 (COVID-19) pandemic has revealed that even variants of SARS-CoV-2 that escaped from neutralizing-antibodies that were induced by either infection or vaccination could not escape from CD8+ T cell-mediated immunity. In addition, although traditional vaccine platforms, such as inactivated virus and subunit vaccines, are less efficient in inducing CD8+ T cells, newly introduced platforms for SARS-CoV-2, namely, mRNA and adenoviral vector vaccines, can induce strong CD8+ T cell-mediated immunity in addition to inducing neutralizing antibodies. However, CD8+ T cells function locally and need to be at the site of infection to control it. To fully utilize the protective performance of CD8+ T cells, it would be insufficient to induce only memory cells circulating in blood, using injectable vaccines; mucosal immunization could be required to set up CD8+ T cells for the optimal protection. CD8+ T cells might also contribute to the pathology of the infection, change their function with age and respond differently to booster vaccines in comparison with antibodies. Herein, we overview cutting-edge ideas on CD8+ T cell-mediated immunity that can enable the rational design of vaccines for respiratory viruses.
Collapse
Affiliation(s)
- Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- *Correspondence: Toshiro Hirai,
| | - Yasuo Yoshioka
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Japan
| |
Collapse
|
67
|
Lagger C, de Magalhães JP. Single-cell gene regulation across aging tissues. NATURE AGING 2022; 2:468-470. [PMID: 37118455 DOI: 10.1038/s43587-022-00238-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
68
|
Zagorski K, Chailyan G, Hovakimyan A, Antonyan T, Kiani Shabestari S, Petrushina I, Davtyan H, Cribbs DH, Blurton-Jones M, Masliah E, Agadjanyan MG, Ghochikyan A. Immunogenicity of MultiTEP-Platform-Based Recombinant Protein Vaccine, PV-1950R, Targeting Three B-Cell Antigenic Determinants of Pathological α-Synuclein. Int J Mol Sci 2022; 23:6080. [PMID: 35682759 PMCID: PMC9181659 DOI: 10.3390/ijms23116080] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are characterized by the aberrant accumulation of intracytoplasmic misfolded and aggregated α-synuclein (α-Syn), resulting in neurodegeneration associated with inflammation. The propagation of α-Syn aggregates from cell to cell is implicated in the spreading of pathological α-Syn in the brain and disease progression. We and others demonstrated that antibodies generated after active and passive vaccinations could inhibit the propagation of pathological α-Syn in the extracellular space and prevent/inhibit disease/s in the relevant animal models. We recently tested the immunogenicity and efficacy of four DNA vaccines on the basis of the universal MultiTEP platform technology in the DLB/PD mouse model. The antibodies generated by these vaccines efficiently reduced/inhibited the accumulation of pathological α-Syn in the different brain regions and improved the motor deficit of immunized female mice. The most immunogenic and preclinically effective vaccine, PV-1950D, targeting three B-cell epitopes of pathological α-Syn simultaneously, has been selected for future IND-enabling studies. However, to ensure therapeutically potent concentrations of α-Syn antibodies in the periphery of the vaccinated elderly, we developed a recombinant protein-based MultiTEP vaccine, PV-1950R/A, and tested its immunogenicity in young and aged D-line mice. Antibody responses induced by immunizations with the PV-1950R/A vaccine and its homologous DNA counterpart, PV-1950D, in a mouse model of PD/DLB have been compared.
Collapse
Affiliation(s)
- Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Sepideh Kiani Shabestari
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD 20814, USA;
| | - Michael G. Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| |
Collapse
|
69
|
He Y, Peng K, Li R, Zhang Z, Pan L, Zhang T, Lin A, Hong R, Nie Z, Guan Q, Jin L. Changes of T lymphocyte subpopulations and their roles in predicting the risk of Parkinson's disease. J Neurol 2022; 269:5368-5381. [PMID: 35608657 PMCID: PMC9467943 DOI: 10.1007/s00415-022-11190-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/29/2022]
Abstract
T lymphocytes are involved in the pathogenesis of Parkinson's disease (PD), while the heterogeneity of T-cell subpopulations remains elusive. In this study, we analyzed up to 22 subpopulations of T lymphocytes in 115 PD patients and 60 matched healthy controls (HC) using flow cytometry. We found that PD patients exhibited decreased naïve CD8+ T cells (CD3+ CD8+ CD45RA+ CD45RO-) and increased late-differentiated CD4+ T cells (CD3+ CD4+ CD28- CD27-), compared to HC, which were not affected by anti-parkinsonism medication administration. The proportion of naïve CD8+ T cells in PD patients was positively correlated with their severity of autonomic dysfunction and psychiatric complications, but negatively associated with the severity of rapid eye movement and sleep behavior disorder. The proportion of late-differentiated CD4+ T cells was negatively correlated with the onset age of the disease. We further developed individualized PD risk prediction models with high reliability and accuracy on the base of the T lymphocyte subpopulations. These data suggest that peripheral cellular immunity is disturbed in PD patients, and changes in CD8+ T cells and late-differentiated CD4+ T cells are representative and significant. Therefore, we recommend naïve CD8 + and late-differentiated CD4+ T cells as candidates for multicentric clinical study and pathomechanism study of PD.
Collapse
Affiliation(s)
- Yijing He
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Kangwen Peng
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ruoyu Li
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Zhuoyu Zhang
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Lizhen Pan
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Tianyu Zhang
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ao Lin
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ronghua Hong
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Zhiyu Nie
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Qiang Guan
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China.
| | - Lingjing Jin
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China. .,Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China. .,Shanghai Clinical Research Center for Aging and Medicine, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
70
|
Hu M, Alashkar Alhamwe B, Santner-Nanan B, Miethe S, Harb H, Renz H, Potaczek DP, Nanan RK. Short-Chain Fatty Acids Augment Differentiation and Function of Human Induced Regulatory T Cells. Int J Mol Sci 2022; 23:ijms23105740. [PMID: 35628549 PMCID: PMC9143307 DOI: 10.3390/ijms23105740] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Regulatory T cells (Tregs) control immune system activity and inhibit inflammation. While, in mice, short-chain fatty acids (SCFAs) are known to be essential regulators of naturally occurring and in vitro induced Tregs (iTregs), data on their contribution to the development of human iTregs are sparse, with no reports of the successful SCFAs-augmented in vitro generation of fully functional human iTregs. Likewise, markers undoubtedly defining human iTregs are missing. Here, we aimed to generate fully functional human iTregs in vitro using protocols involving SCFAs and to characterize the underlying mechanism. Our target was to identify the potential phenotypic markers best characterizing human iTregs. Naïve non-Treg CD4+ cells were isolated from the peripheral blood of 13 healthy adults and cord blood of 12 healthy term newborns. Cells were subjected to differentiation toward iTregs using a transforming growth factor β (TGF-β)-based protocol, with or without SCFAs (acetate, butyrate, or propionate). Thereafter, they were subjected to flow cytometric phenotyping or a suppression assay. During differentiation, cells were collected for chromatin-immunoprecipitation (ChIP)-based analysis of histone acetylation. The enrichment of the TGF-β-based protocol with butyrate or propionate potentiated the in vitro differentiation of human naïve CD4+ non-Tregs towards iTregs and augmented the suppressive capacity of the latter. These seemed to be at least partly underlain by the effects of SCFAs on the histone acetylation levels in differentiating cells. GITR, ICOS, CD39, PD-1, and PD-L1 were proven to be potential markers of human iTregs. Our results might boost the further development of Treg-based therapies against autoimmune, allergic and other chronic inflammatory disorders.
Collapse
Affiliation(s)
- Mingjing Hu
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Discipline of Obstetrics, Gynaecology and Neonatology, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia
- Nepean Hospital, Derby Street, Kingswood, NSW 2747, Australia
| | - Bilal Alashkar Alhamwe
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute for Tumor Immunology, Clinic for Hematology, Immunology, and Oncology, Philipps University Marburg, 35043 Marburg, Germany
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Brigitte Santner-Nanan
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
| | - Sarah Miethe
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany
| | - Hani Harb
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Institute of Medical Microbiology and Virology, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany
| | - Harald Renz
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
| | - Daniel P. Potaczek
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany
| | - Ralph K. Nanan
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Correspondence: ; Tel.: +61-2-4734-2612; Fax: +61-2-4734-1144
| |
Collapse
|
71
|
Bell MR, Kutzler MA. An old problem with new solutions: Strategies to improve vaccine efficacy in the elderly. Adv Drug Deliv Rev 2022; 183:114175. [PMID: 35202770 DOI: 10.1016/j.addr.2022.114175] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 11/01/2022]
Abstract
Vaccination is the most effective measure to protect against infections. However, with increasing age, there is a progressive decline in the ability of the immune system to both protect against infection and develop protective immunity from vaccination. This age-related decline of the immune system is due to age-related changes in both the innate and adaptive immune systems. With an aging world population and increased risk of pandemics, there is a need to continue to develop strategies to increase vaccine responses in the elderly. Here, the major age-related changes that occur in both the innate and adaptive immune responses that impair the response to vaccination in the elderly will be highlighted. Existing and future strategies to improve vaccine efficacy in the elderly will then be discussed, including adjuvants, delivery methods, and formulation. These strategies provide mechanisms to improve the efficacy of existing vaccines and develop novel vaccines for the elderly.
Collapse
|
72
|
Shafqat A, Shafqat S, Salameh SA, Kashir J, Alkattan K, Yaqinuddin A. Mechanistic Insights Into the Immune Pathophysiology of COVID-19; An In-Depth Review. Front Immunol 2022; 13:835104. [PMID: 35401519 PMCID: PMC8989408 DOI: 10.3389/fimmu.2022.835104] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), which causes coronavirus-19 (COVID-19), has caused significant morbidity and mortality globally. In addition to the respiratory manifestations seen in severe cases, multi-organ pathologies also occur, making management a much-debated issue. In addition, the emergence of new variants can potentially render vaccines with a relatively limited utility. Many investigators have attempted to elucidate the precise pathophysiological mechanisms causing COVID-19 respiratory and systemic disease. Spillover of lung-derived cytokines causing a cytokine storm is considered the cause of systemic disease. However, recent studies have provided contradictory evidence, whereby the extent of cytokine storm is insufficient to cause severe illness. These issues are highly relevant, as management approaches considering COVID-19 a classic form of acute respiratory distress syndrome with a cytokine storm could translate to unfounded clinical decisions, detrimental to patient trajectory. Additionally, the precise immune cell signatures that characterize disease of varying severity remain contentious. We provide an up-to-date review on the immune dysregulation caused by COVID-19 and highlight pertinent discussions in the scientific community. The response from the scientific community has been unprecedented regarding the development of highly effective vaccines and cutting-edge research on novel therapies. We hope that this review furthers the conversations held by scientists and informs the aims of future research projects, which will potentially further our understanding of COVID-19 and its immune pathogenesis.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Center of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
73
|
Niemiro GM, Coletta AM, Agha NH, Mylabathula PL, Baker FL, Brewster AM, Bevers TB, Fuentes-Mattei E, Basen-Engquist K, Katsanis E, Gilchrist SC, Simpson RJ. Salutary effects of moderate but not high intensity aerobic exercise training on the frequency of peripheral T-cells associated with immunosenescence in older women at high risk of breast cancer: a randomized controlled trial. Immun Ageing 2022; 19:17. [PMID: 35321743 PMCID: PMC8941789 DOI: 10.1186/s12979-022-00266-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/31/2022] [Indexed: 12/20/2022]
Abstract
Background Immunosenescence is described as age-associated changes within the immune system that are responsible for decreased immunity and increased cancer risk. Physically active individuals have fewer ‘senescent’ and more naïve T-cells compared to their sedentary counterparts, but it is not known if exercise training can rejuvenate ‘older looking’ T-cell profiles. We determined the effects of 12-weeks supervised exercise training on the frequency of T-cell subtypes in peripheral blood and their relationships with circulating levels of the muscle-derived cytokines (i.e. ‘myokines’) IL-6, IL-7, IL-15 and osteonectin in older women at high risk of breast cancer. The intervention involved 3 sessions/week of either high intensity interval exercise (HIIT) or moderate intensity continuous exercise (MICT) and were compared to an untrained control (UC) group. Results HIIT decreased total granulocytes, CD4+ T-cells, CD4+ naïve T-cells, CD4+ recent thymic emigrants (RTE) and the CD4:CD8 ratio after training, whereas MICT increased total lymphocytes and CD8 effector memory (EM) T-cells. The change in total T-cells, CD4+ naïve T-cells, CD4+ central memory (CM) T-cells and CD4+ RTE was elevated after MICT compared to HIIT. Changes in \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$ \dot{\mathrm{V}}{\mathrm{O}}_{2\max } $$\end{document}V˙O2max after training, regardless of exercise prescription, was inversely related to the change in highly differentiated CD8+ EMRA T-cells and positively related to changes in β2-adrenergic receptor (β2-AR) expression on CM CD4+ and CM CD8+ T-cells. Plasma myokine levels did not change significantly among the groups after training, but individual changes in IL-7 were positively related to changes in the number of β2-AR expressing CD4 naïve T cells in both exercise groups but not controls. Further, CD4 T-cells and CD4 naive T-cells were negatively related to changes in IL-6 and osteonectin after HIIT but not MICT, whereas CD8 EMRA T-cells were inversely related to changes in IL-15 after MICT but not HIIT. Conclusions Aerobic exercise training alters the frequency of peripheral T-cells associated with immunosenescence in middle aged/older women at high risk of breast cancer, with HIIT (pro-senescent) and MICT (anti-senescent) evoking divergent effects. Identifying the underlying mechanisms and establishing whether exercise-induced changes in peripheral T-cell numbers can alter the risk of developing breast cancer warrants investigation. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00266-z.
Collapse
Affiliation(s)
- Grace M Niemiro
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA.,The University of Arizona Cancer Center, Tucson, Arizona, USA
| | - Adriana M Coletta
- Department of Health and Kinesiology, The University of Utah, Salt Lake City, Utah, USA.,Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Nadia H Agha
- Department of Health and Human Performance, University of Houston, Houston, Texas, USA
| | - Preteesh Leo Mylabathula
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA.,Department of Health and Human Performance, University of Houston, Houston, Texas, USA.,School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, Arizona, USA
| | - Forrest L Baker
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA.,Department of Health and Human Performance, University of Houston, Houston, Texas, USA.,School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, Arizona, USA
| | - Abenaa M Brewster
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Therese B Bevers
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Enrique Fuentes-Mattei
- Department of Radiation Oncology Clinical Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Karen Basen-Engquist
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA.,The University of Arizona Cancer Center, Tucson, Arizona, USA.,Department of Immunobiology, The University of Arizona, Tucson, Arizona, USA
| | - Susan C Gilchrist
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard J Simpson
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA. .,The University of Arizona Cancer Center, Tucson, Arizona, USA. .,Department of Health and Human Performance, University of Houston, Houston, Texas, USA. .,School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, Arizona, USA. .,Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA. .,Department of Immunobiology, The University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|
74
|
Matthe DM, Thoma OM, Sperka T, Neurath MF, Waldner MJ. Telomerase deficiency reflects age-associated changes in CD4+ T cells. Immun Ageing 2022; 19:16. [PMID: 35321714 PMCID: PMC8941756 DOI: 10.1186/s12979-022-00273-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/02/2022] [Indexed: 01/01/2023]
Abstract
Background Amongst other systemic changes, aging leads to an immune dysfunction. On the molecular level, a hallmark of aging is telomere shortening. The functional relevance of telomerase, an enzyme capable of elongating telomeres in T cells upon antigen stimulation, is not fully understood. Studying the impact of telomere shortening on CD4+ T cells and especially Th1 effector function can provide a better understanding on immune dysfunctions in elderly. Results We investigated T cell numbers and differentiation in telomerase-deficient (mTerc−/−) mice under steady-state conditions and the functional role of telomerase in CD4+ T cells using in vitro stimulation and Th1 polarization protocols by comparing T cells from mTerc−/− and control mice. We report reduced relative CD4+ T cell numbers in blood and secondary lymphoid organs and a relative decline in the naïve T cell population in thymus, blood and spleen of mTerc−/− mice compared to control mice. Importantly, after in vitro polarization, mTerc−/− G3 CD4+ T cells showed higher numbers of IFNγ-producing cells and reduced expression of CD28. Notably, telomerase-deficient T cells were more susceptible to inhibition of Th1 polarization by IL-6 in vitro. These results demonstrate that telomerase deficiency recapitulates several changes of CD4+ T cells seen in aged humans regarding the naïve T cell population, expression of CD28 and cytokine production. Conclusion Our data suggest that telomere shortening could play a key role in the aging of T cell immunity, with clinical implications for immune diseases and tumor development and that mTerc−/− mice are a suitable model to study aging-related defects of adaptive immunity. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00273-0.
Collapse
Affiliation(s)
- Diana M Matthe
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Oana-Maria Thoma
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany.,Erlangen Graduate School of Advanced Optical Technologies (SAOT), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Sperka
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany. .,Erlangen Graduate School of Advanced Optical Technologies (SAOT), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
75
|
Molinaro AM, Wiencke JK, Warrier G, Koestler DC, Chunduru P, Lee JY, Hansen HM, Lee S, Anguiano J, Rice T, Bracci PM, McCoy L, Salas LA, Christensen BC, Wrensch M, Kelsey KT, Taylor JW, Clarke JL. Interactions of Age and Blood Immune Factors and Noninvasive Prediction of Glioma Survival. J Natl Cancer Inst 2022; 114:446-457. [PMID: 34597382 PMCID: PMC8902347 DOI: 10.1093/jnci/djab195] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Tumor-based classification of human glioma portends patient prognosis, but considerable unexplained survival variability remains. Host factors (eg, age) also strongly influence survival times, partly reflecting a compromised immune system. How blood epigenetic measures of immune characteristics and age augment molecular classifications in glioma survival has not been investigated. We assess the prognostic impact of immune cell fractions and epigenetic age in archived blood across glioma molecular subtypes for the first time. METHODS We evaluated immune cell fractions and epigenetic age in archived blood from the University of California San Francisco Adult Glioma Study, which included a training set of 197 patients with IDH-wild type, 1p19q intact, TERT wild type (IDH/1p19q/TERT-WT) glioma, an evaluation set of 350 patients with other subtypes of glioma, and 454 patients without glioma. RESULTS IDH/1p19q/TERT-WT patients had lower lymphocyte fractions (CD4+ T, CD8+ T, natural killer, and B cells) and higher neutrophil fractions than people without glioma. Recursive partitioning analysis delineated 4 statistically significantly different survival groups for patients with IDH/1p19q/TERT-WT based on an interaction between chronological age and 2 blood immune factors, CD4+ T cells, and neutrophils. Median overall survival ranged from 0.76 years (95% confidence interval = 0.55-0.99) for the worst survival group (n = 28) to 9.72 years (95% confidence interval = 6.18 to not available) for the best (n = 33). The recursive partitioning analysis also statistically significantly delineated 4 risk groups in patients with other glioma subtypes. CONCLUSIONS The delineation of different survival groups in the training and evaluation sets based on an interaction between chronological age and blood immune characteristics suggests that common host immune factors among different glioma types may affect survival. The ability of DNA methylation-based markers of immune status to capture diverse, clinically relevant information may facilitate noninvasive, personalized patient evaluation in the neuro-oncology clinic.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ji Yoon Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sean Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joaquin Anguiano
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Departments of Molecular and Systems Biology and Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
76
|
Kurban LAS, AlDhaheri S, Elkkari A, Khashkhusha R, AlEissaee S, AlZaabi A, Ismail M, Bakoush O. Predicting Severe Disease and Critical Illness on Initial Diagnosis of COVID-19: Simple Triage Tools. Front Med (Lausanne) 2022; 9:817549. [PMID: 35223916 PMCID: PMC8866724 DOI: 10.3389/fmed.2022.817549] [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: 11/18/2021] [Accepted: 01/17/2022] [Indexed: 01/08/2023] Open
Abstract
Rationale This study was conducted to develop, validate, and compare prediction models for severe disease and critical illness among symptomatic patients with confirmed COVID-19. Methods For development cohort, 433 symptomatic patients diagnosed with COVID-19 between April 15th 2020 and June 30th, 2020 presented to Tawam Public Hospital, Abu Dhabi, United Arab Emirates were included in this study. Our cohort included both severe and non-severe patients as all cases were admitted for purpose of isolation as per hospital policy. We examined 19 potential predictors of severe disease and critical illness that were recorded at the time of initial assessment. Univariate and multivariate logistic regression analyses were used to construct predictive models. Discrimination was assessed by the area under the receiver operating characteristic curve (AUC). Calibration and goodness of fit of the models were assessed. A cohort of 213 patients assessed at another public hospital in the country during the same period was used to validate the models. Results One hundred and eighty-six patients were classified as severe while the remaining 247 were categorized as non-severe. For prediction of progression to severe disease, the three independent predictive factors were age, serum lactate dehydrogenase (LDH) and serum albumin (ALA model). For progression to critical illness, the four independent predictive factors were age, serum LDH, kidney function (eGFR), and serum albumin (ALKA model). The AUC for the ALA and ALKA models were 0.88 (95% CI, 0.86–0.89) and 0.85 (95% CI, 0.83–0.86), respectively. Calibration of the two models showed good fit and the validation cohort showed excellent discrimination, with an AUC of 0.91 (95% CI, 0.83–0.99) for the ALA model and 0.89 (95% CI, 0.80–0.99) for the ALKA model. A free web-based risk calculator was developed. Conclusions The ALA and ALKA predictive models were developed and validated based on simple, readily available clinical and laboratory tests assessed at presentation. These models may help frontline clinicians to triage patients for admission or discharge, as well as for early identification of patients at risk of developing critical illness.
Collapse
Affiliation(s)
| | - Sharina AlDhaheri
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Abdulbaset Elkkari
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ramzi Khashkhusha
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Shaikha AlEissaee
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Amna AlZaabi
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Mohamed Ismail
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Omran Bakoush
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
77
|
Fernandes JR, Pinto TNC, Arruda LB, da Silva CCBM, de Carvalho CRF, Pinto RMC, da Silva Duarte AJ, Benard G. Age-associated phenotypic imbalance in TCD4 and TCD8 cell subsets: comparison between healthy aged, smokers, COPD patients and young adults. IMMUNITY & AGEING 2022; 19:9. [PMID: 35164774 PMCID: PMC8842531 DOI: 10.1186/s12979-022-00267-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
Abstract
Abstract
Background
COPD is associated with an abnormal lung immune response that leads to tissue damage and remodeling of the lung, but also to systemic effects that compromise immune responses. Cigarette smoking also impacts on innate and adaptative immune responses, exerting dual, pro- and anti-inflammatory effects. Previously, we showed that COPD patients presented accelerated telomere shortening and decreased telomerase activity, while, paradoxically, cigarette-smokers exhibited preserved telomerase activity and slower rate of telomere shortening.
Results
Here, we evaluated the naive, CM, EM and TEMRA subsets of TCD4 and TCD8 cells according to the expression of CCR7/CD45RA. We compared age-matched COPD patients, cigarette-smokers without clinical-laboratory evidence of pulmonary compromise, and healthy individuals. They were additionally compared with a group of young adults. For each subset we analysed the expression of markers associated with late differentiation, senescence and exhaustion (CD27/CD28/CD57/KLRG1/PD1). We show that COPD patients presented a drastically reduced naive cells pool, and, paradoxically, increased fractions of naive cells expressing late differentiation, senescence or exhaustion markers, likely impacting on their immunocompetence. Pronounced phenotypic alterations were also evidenced in their three memory T-cell subsets compared with the other aged and young groups, suggesting an also dysfunctional memory pool. Surprisingly, our smokers showed a profile closer to the Healthy aged than COPD patients. They exhibited the usual age-associated shift of naive to EM TCD4 and TCD8 cells, but not to CM or TEMRA T-cells. Nonetheless, their naive T-cells phenotypes were in general similar to those of the Youngs and Healthy aged, suggesting a rather phenotypically preserved subset, while the memory T-cells exhibited increased proportions of cells with the late-differentiation or senescence/exhaustion markers as in the Healthy aged.
Conclusion
Our study extends previous findings by showing that COPD patients have cells expressing a full range of late differentiated, senescent or exhausted phenotypes encompassing all TCD4 and TCD8 subsets, consistent with a premature immunosenescence phenotype. Surprisingly, the smokers group’s results suggest that moderate to heavy chronic cigarette smoking did not accelerate the pace of immunosenescence as compared with the Healthy aged.
Collapse
|
78
|
Nicoli F, Cabral-Piccin MP, Papagno L, Gallerani E, Fusaro M, Folcher V, Dubois M, Clave E, Vallet H, Frere JJ, Gostick E, Llewellyn-Lacey S, Price DA, Toubert A, Dupré L, Boddaert J, Caputo A, Gavioli R, Appay V. Altered Basal Lipid Metabolism Underlies the Functional Impairment of Naive CD8 + T Cells in Elderly Humans. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:562-570. [PMID: 35031578 PMCID: PMC7615155 DOI: 10.4049/jimmunol.2100194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/24/2021] [Indexed: 12/26/2022]
Abstract
Aging is associated with functional deficits in the naive T cell compartment, which compromise the generation of de novo immune responses against previously unencountered Ags. The mechanisms that underlie this phenomenon have nonetheless remained unclear. We found that naive CD8+ T cells in elderly humans were prone to apoptosis and proliferated suboptimally in response to stimulation via the TCR. These abnormalities were associated with dysregulated lipid metabolism under homeostatic conditions and enhanced levels of basal activation. Importantly, reversal of the bioenergetic anomalies with lipid-altering drugs, such as rosiglitazone, almost completely restored the Ag responsiveness of naive CD8+ T cells. Interventions that favor lipid catabolism may therefore find utility as adjunctive therapies in the elderly to promote vaccine-induced immunity against targetable cancers and emerging pathogens, such as seasonal influenza viruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Francesco Nicoli
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France;
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Mariela P Cabral-Piccin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
| | - Laura Papagno
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
| | - Eleonora Gallerani
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Mathieu Fusaro
- Toulouse Institute for Infectious and Inflammatory Diseases, Université Toulouse III, INSERM UMR1291/CNRS UMR5051, Toulouse, France
| | - Victor Folcher
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
| | - Marion Dubois
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
| | - Emmanuel Clave
- Institut de Recherche Saint Louis, EMiLy, Université de Paris, INSERM U1160, Paris, France
| | - Hélène Vallet
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
- Service de Gériatrie, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Justin J Frere
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine Tucson, Tucson, AZ
| | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Antoine Toubert
- Institut de Recherche Saint Louis, EMiLy, Université de Paris, INSERM U1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Loïc Dupré
- Toulouse Institute for Infectious and Inflammatory Diseases, Université Toulouse III, INSERM UMR1291/CNRS UMR5051, Toulouse, France
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Jacques Boddaert
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France
- Service de Gériatrie, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Antonella Caputo
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Riccardo Gavioli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Victor Appay
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, INSERM U1135, Paris, France;
- International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan; and
- Université de Bordeaux, CNRS UMR5164, INSERM ERL1303, ImmunoConcEpT, Bordeaux, France
| |
Collapse
|
79
|
Hou Y, Zhou Y, Jehi L, Luo Y, Gack MU, Chan T, Yu H, Eng C, Pieper AA, Cheng F. Aging-related cell type-specific pathophysiologic immune responses that exacerbate disease severity in aged COVID-19 patients. Aging Cell 2022; 21:e13544. [PMID: 35023286 PMCID: PMC8844132 DOI: 10.1111/acel.13544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID‐19) is especially severe in aged patients, defined as 65 years or older, for reasons that are currently unknown. To investigate the underlying basis for this vulnerability, we performed multimodal data analyses on immunity, inflammation, and COVID‐19 incidence and severity as a function of age. Our analysis leveraged age‐specific COVID‐19 mortality and laboratory testing from a large COVID‐19 registry, along with epidemiological data of ~3.4 million individuals, large‐scale deep immune cell profiling data, and single‐cell RNA‐sequencing data from aged COVID‐19 patients across diverse populations. We found that decreased lymphocyte count and elevated inflammatory markers (C‐reactive protein, D‐dimer, and neutrophil–lymphocyte ratio) are significantly associated with age‐specific COVID‐19 severities. We identified the reduced abundance of naïve CD8 T cells with decreased expression of antiviral defense genes (i.e., IFITM3 and TRIM22) in aged severe COVID‐19 patients. Older individuals with severe COVID‐19 displayed type I and II interferon deficiencies, which is correlated with SARS‐CoV‐2 viral load. Elevated expression of SARS‐CoV‐2 entry factors and reduced expression of antiviral defense genes (LY6E and IFNAR1) in the secretory cells are associated with critical COVID‐19 in aged individuals. Mechanistically, we identified strong TGF‐beta‐mediated immune–epithelial cell interactions (i.e., secretory‐non‐resident macrophages) in aged individuals with critical COVID‐19. Taken together, our findings point to immuno‐inflammatory factors that could be targeted therapeutically to reduce morbidity and mortality in aged COVID‐19 patients.
Collapse
Affiliation(s)
- Yuan Hou
- Genomic Medicine Institute Lerner Research Institute Cleveland Clinic Cleveland Ohio USA
| | - Yadi Zhou
- Genomic Medicine Institute Lerner Research Institute Cleveland Clinic Cleveland Ohio USA
| | - Lara Jehi
- Quantitative Health Sciences, Lerner Research Institute Cleveland Clinic Cleveland Ohio USA
- Neurological Institute Cleveland Clinic Cleveland Ohio USA
| | - Yuan Luo
- Division of Health and Biomedical Informatics Department of Preventive Medicine Clinical and Translational Sciences Institute and Center for Health Information Partnerships Northwestern University Evanston Illinois USA
| | - Michaela U. Gack
- Florida Research and Innovation Center Cleveland Clinic Port Saint Lucie Florida USA
| | - Timothy A. Chan
- Center for Immunotherapy and Precision Immuno‐Oncology Cleveland Clinic Cleveland Ohio USA
| | - Haiyuan Yu
- Weill Institute for Cell and Molecular Biology Cornell University Ithaca New York USA
- Department of Computational Biology Cornell University Ithaca New York USA
| | - Charis Eng
- Genomic Medicine Institute Lerner Research Institute Cleveland Clinic Cleveland Ohio USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland Ohio USA
- Department of Genetics and Genome Sciences Case Western Reserve University School of Medicine Cleveland Ohio USA
- Case Comprehensive Cancer Center Case Western Reserve University School of Medicine Cleveland Ohio USA
| | - Andrew A. Pieper
- Harrington Discovery Institute University Hospitals Cleveland Medical Center Cleveland Ohio USA
- Department of Psychiatry Case Western Reserve University Cleveland Ohio USA
- Geriatric Psychiatry GRECC Louis Stokes Cleveland VA Medical Center Cleveland Ohio USA
- Institute for Transformative Molecular Medicine School of Medicine Case Western Reserve University Cleveland Ohio USA
- Weill Cornell Autism Research Program Weill Cornell Medicine of Cornell University New York New York USA
| | - Feixiong Cheng
- Genomic Medicine Institute Lerner Research Institute Cleveland Clinic Cleveland Ohio USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center Case Western Reserve University School of Medicine Cleveland Ohio USA
| |
Collapse
|
80
|
Framme JL, Lundqvist C, Lundell AC, van Schouwenburg PA, Lemarquis AL, Thörn K, Lindgren S, Gudmundsdottir J, Lundberg V, Degerman S, Zetterström RH, Borte S, Hammarström L, Telemo E, Hultdin M, van der Burg M, Fasth A, Oskarsdóttir S, Ekwall O. Long-Term Follow-Up of Newborns with 22q11 Deletion Syndrome and Low TRECs. J Clin Immunol 2022; 42:618-633. [PMID: 35080750 PMCID: PMC9016018 DOI: 10.1007/s10875-021-01201-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/12/2021] [Indexed: 01/03/2023]
Abstract
Background Population-based neonatal screening using T-cell receptor excision circles (TRECs) identifies infants with profound T lymphopenia, as seen in cases of severe combined immunodeficiency, and in a subgroup of infants with 22q11 deletion syndrome (22q11DS). Purpose To investigate the long-term prognostic value of low levels of TRECs in newborns with 22q11DS. Methods Subjects with 22q11DS and low TRECs at birth (22q11Low, N=10), matched subjects with 22q11DS and normal TRECs (22q11Normal, N=10), and matched healthy controls (HC, N=10) were identified. At follow-up (median age 16 years), clinical and immunological characterizations, covering lymphocyte subsets, immunoglobulins, TRECs, T-cell receptor repertoires, and relative telomere length (RTL) measurements were performed. Results At follow-up, the 22q11Low group had lower numbers of naïve T-helper cells, naïve T-regulatory cells, naïve cytotoxic T cells, and persistently lower TRECs compared to healthy controls. Receptor repertoires showed skewed V-gene usage for naïve T-helper cells, whereas for naïve cytotoxic T cells, shorter RTL and a trend towards higher clonality were found. Multivariate discriminant analysis revealed a clear distinction between the three groups and a skewing towards Th17 differentiation of T-helper cells, particularly in the 22q11Low individuals. Perturbations of B-cell subsets were found in both the 22q11Low and 22q11Normal group compared to the HC group, with larger proportions of naïve B cells and lower levels of memory B cells, including switched memory B cells. Conclusions This long-term follow-up study shows that 22q11Low individuals have persistent immunologic aberrations and increased risk for immune dysregulation, indicating the necessity of lifelong monitoring. Clinical Implications This study elucidates the natural history of childhood immune function in newborns with 22q11DS and low TRECs, which may facilitate the development of programs for long-term monitoring and therapeutic choices. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-021-01201-5.
Collapse
Affiliation(s)
- Jenny Lingman Framme
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Pediatrics, Halland Hospital Halmstad, Halmstad, Region Halland, Sweden.
| | - Christina Lundqvist
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna-Carin Lundell
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pauline A van Schouwenburg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Andri L Lemarquis
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Susanne Lindgren
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Judith Gudmundsdottir
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Children's Medical Center, National University Hospital of Iceland, Reykjavík, Iceland
| | - Vanja Lundberg
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sofie Degerman
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Rolf H Zetterström
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Stephan Borte
- ImmunoDeficiencyCenter Leipzig (IDCL), Municipal Hospital St. Georg Leipzig, Leipzig, Germany
| | - Lennart Hammarström
- Department of Biosciences and Nutrition, Neo, Karolinska Institute, Stockholm, Sweden
| | - Esbjörn Telemo
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Magnus Hultdin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Anders Fasth
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sólveig Oskarsdóttir
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Olov Ekwall
- Department of Pediatrics, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
81
|
Teissier T, Boulanger E, Cox LS. Interconnections between Inflammageing and Immunosenescence during Ageing. Cells 2022; 11:359. [PMID: 35159168 PMCID: PMC8834134 DOI: 10.3390/cells11030359] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023] Open
Abstract
Acute inflammation is a physiological response to injury or infection, with a cascade of steps that ultimately lead to the recruitment of immune cells to clear invading pathogens and heal wounds. However, chronic inflammation arising from the continued presence of the initial trigger, or the dysfunction of signalling and/or effector pathways, is harmful to health. While successful ageing in older adults, including centenarians, is associated with low levels of inflammation, elevated inflammation increases the risk of poor health and death. Hence inflammation has been described as one of seven pillars of ageing. Age-associated sterile, chronic, and low-grade inflammation is commonly termed inflammageing-it is not simply a consequence of increasing chronological age, but is also a marker of biological ageing, multimorbidity, and mortality risk. While inflammageing was initially thought to be caused by "continuous antigenic load and stress", reports from the last two decades describe a much more complex phenomenon also involving cellular senescence and the ageing of the immune system. In this review, we explore some of the main sources and consequences of inflammageing in the context of immunosenescence and highlight potential interventions. In particular, we assess the contribution of cellular senescence to age-associated inflammation, identify patterns of pro- and anti-inflammatory markers characteristic of inflammageing, describe alterations in the ageing immune system that lead to elevated inflammation, and finally assess the ways that diet, exercise, and pharmacological interventions can reduce inflammageing and thus, improve later life health.
Collapse
Affiliation(s)
- Thibault Teissier
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| | - Eric Boulanger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France;
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| |
Collapse
|
82
|
Kim C, Hovakimyan A, Zagorski K, Antonyan T, Petrushina I, Davtyan H, Chailyan G, Hasselmann J, Iba M, Adame A, Rockenstein E, Szabo M, Blurton-Jones M, Cribbs DH, Ghochikyan A, Masliah E, Agadjanyan MG. Efficacy and immunogenicity of MultiTEP-based DNA vaccines targeting human α-synuclein: prelude for IND enabling studies. NPJ Vaccines 2022; 7:1. [PMID: 35013319 PMCID: PMC8748802 DOI: 10.1038/s41541-021-00424-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulation of misfolded proteins such as amyloid-β (Aβ), tau, and α-synuclein (α-Syn) in the brain leads to synaptic dysfunction, neuronal damage, and the onset of relevant neurodegenerative disorder/s. Dementia with Lewy bodies (DLB) and Parkinson's disease (PD) are characterized by the aberrant accumulation of α-Syn intracytoplasmic Lewy body inclusions and dystrophic Lewy neurites resulting in neurodegeneration associated with inflammation. Cell to cell propagation of α-Syn aggregates is implicated in the progression of PD/DLB, and high concentrations of anti-α-Syn antibodies could inhibit/reduce the spreading of this pathological molecule in the brain. To ensure sufficient therapeutic concentrations of anti-α-Syn antibodies in the periphery and CNS, we developed four α-Syn DNA vaccines based on the universal MultiTEP platform technology designed especially for the elderly with immunosenescence. Here, we are reporting on the efficacy and immunogenicity of these vaccines targeting three B-cell epitopes of hα-Syn aa85-99 (PV-1947D), aa109-126 (PV-1948D), aa126-140 (PV-1949D) separately or simultaneously (PV-1950D) in a mouse model of synucleinopathies mimicking PD/DLB. All vaccines induced high titers of antibodies specific to hα-Syn that significantly reduced PD/DLB-like pathology in hα-Syn D line mice. The most significant reduction of the total and protein kinase resistant hα-Syn, as well as neurodegeneration, were observed in various brain regions of mice vaccinated with PV-1949D and PV-1950D in a sex-dependent manner. Based on these preclinical data, we selected the PV-1950D vaccine for future IND enabling preclinical studies and clinical development.
Collapse
Affiliation(s)
- Changyoun Kim
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Jonathan Hasselmann
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Michiyo Iba
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Marcell Szabo
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA.
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| |
Collapse
|
83
|
Hofer S, Hofstätter N, Punz B, Hasenkopf I, Johnson L, Himly M. Immunotoxicity of nanomaterials in health and disease: Current challenges and emerging approaches for identifying immune modifiers in susceptible populations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1804. [PMID: 36416020 PMCID: PMC9787548 DOI: 10.1002/wnan.1804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 11/24/2022]
Abstract
Nanosafety assessment has experienced an intense era of research during the past decades driven by a vivid interest of regulators, industry, and society. Toxicological assays based on in vitro cellular models have undergone an evolution from experimentation using nanoparticulate systems on singular epithelial cell models to employing advanced complex models more realistically mimicking the respective body barriers for analyzing their capacity to alter the immune state of exposed individuals. During this phase, a number of lessons were learned. We have thus arrived at a state where the next chapters have to be opened, pursuing the following objectives: (1) to elucidate underlying mechanisms, (2) to address effects on vulnerable groups, (3) to test material mixtures, and (4) to use realistic doses on (5) sophisticated models. Moreover, data reproducibility has become a significant demand. In this context, we studied the emerging concept of adverse outcome pathways (AOPs) from the perspective of immune activation and modulation resulting in pro-inflammatory versus tolerogenic responses. When considering the interaction of nanomaterials with biological systems, protein corona formation represents the relevant molecular initiating event (e.g., by potential alterations of nanomaterial-adsorbed proteins). Using this as an example, we illustrate how integrated experimental-computational workflows combining in vitro assays with in silico models aid in data enrichment and upon comprehensive ontology-annotated (meta)data upload to online repositories assure FAIRness (Findability, Accessibility, Interoperability, Reusability). Such digital twinning may, in future, assist in early-stage decision-making during therapeutic development, and hence, promote safe-by-design innovation in nanomedicine. Moreover, it may, in combination with in silico-based exposure-relevant dose-finding, serve for risk monitoring in particularly loaded areas, for example, workplaces, taking into account pre-existing health conditions. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Sabine Hofer
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Norbert Hofstätter
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Benjamin Punz
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Ingrid Hasenkopf
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Litty Johnson
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Martin Himly
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| |
Collapse
|
84
|
Cai G, Guan Z, Jin Y, Su Z, Chen X, Liu Q, Wang C, Yin X, Zhang L, Ye G, Luo W. Circulating T-Cell Repertoires Correlate With the Tumor Response in Patients With Breast Cancer Receiving Neoadjuvant Chemotherapy. JCO Precis Oncol 2022; 6:e2100120. [PMID: 35025620 PMCID: PMC8769146 DOI: 10.1200/po.21.00120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) has been widely used in patients with breast cancer to minish tumor burden and increase resection rate of cancer. T-cell repertoire has been believed to be able to monitor antitumor immune responses. This study aimed to explore the dynamic change of T-cell repertoire and its clinical value in evaluating the tumor response in patients with breast cancer receiving NAC. MATERIALS AND METHODS Ninety-four patients who underwent NAC before surgery were recruited, and peripheral blood samples were collected at multiple time points during NAC. High-throughput T-cell receptor (TCR)-β sequencing was used to characterize the T-cell repertoire of every sample and analyzed the changes in circulating T-cell repertoire during NAC. RESULTS We found that the diversity of TCR repertoires was associated with age and clinical stage of the patients with breast cancer. The distribution of Vβ and Jβ genes in TCR repertoires was skewed in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer. Vβ20.1 and Vβ30 expression levels before NAC correlate with tumor response after all cycles of NAC in HER2- and HER2+ patients, respectively. Some CDR3 motifs that correlated with clinical response in either HER2+ or HER2- patients were identified. Besides, TCR repertoire evolved during NAC and the diversity of TCR repertoire decreased more after two cycles of NAC in patients with good tumor response after all cycles of NAC (P = .0061). CONCLUSION Our results demonstrated that TCR repertoire correlated with the characteristics of the tumor, such as the expression status of HER2. Moreover, some characteristics of TCR repertoires that correlated with clinical response were identified and they might provide useful information to tailor therapeutic regimens at the early cycle of NAC.
Collapse
Affiliation(s)
- Gengxi Cai
- The First People's Hospital of Foshan, Foshan, China
| | - Zhanwen Guan
- The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- The First People's Hospital of Foshan, Foshan, China
| | - Zuhui Su
- The First People's Hospital of Foshan, Foshan, China
| | | | - Qing Liu
- The First People's Hospital of Foshan, Foshan, China
| | | | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, China
| | - Lifang Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Guolin Ye
- The First People's Hospital of Foshan, Foshan, China
| | - Wei Luo
- The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
85
|
Witkowski JM. Immune system aging and the aging-related diseases in the COVIID-19 era. Immunol Lett 2022; 243:19-27. [PMID: 35108570 PMCID: PMC8801734 DOI: 10.1016/j.imlet.2022.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 12/13/2022]
Abstract
The interest in the process of aging, and specifically in how aging affects the working of our immune system, has recently enormously grown among both specialists (immunologists and gerontologists) and representatives of other disciplines of health sciences. An obvious reason for this interest is the current pandemics of COVID-19, known to affect the elderly more than younger people. In this paper current knowledge about mechanisms and complex facets of human immune system aging is presented, stemming from the knowledge about the working of various parts of the immune system, and leading to understanding of immunological mechanisms of chronic, inflammatory, aging-related diseases and of COVID-19.
Collapse
Affiliation(s)
- Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland.
| |
Collapse
|
86
|
Liisborg C, Skov V, Kjær L, Hasselbalch HC, Sørensen TL. Retinal drusen in patients with chronic myeloproliferative blood cancers are associated with an increased proportion of senescent T cells and signs of an aging immune system. Aging (Albany NY) 2021; 13:25763-25777. [PMID: 34954692 PMCID: PMC8751607 DOI: 10.18632/aging.203803] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
The cause of age-related macular degeneration (AMD) is unknown, but evidence indicates that both innate and adaptive immunity play a role in the pathogenesis. Our recent work has investigated AMD in patients with myeloproliferative neoplasms (MPNs) since they have increased drusen and AMD prevalence. We have previously found increased levels of chronic low-grade inflammation (CLI) in MPN patients with drusen (MPNd) compared to MPN patients with normal retinas (MPNn). CLI and AMD are both associated with aging, and we, therefore, wanted to study immunosenescence markers in MPNd, MPNn, and AMD. The purpose was to identify differences between MPNd and MPNn, which might reveal novel information relevant to drusen pathophysiology and thereby the AMD pathogenesis. Our results suggest that MPNd have a T cell differentiation profile resembling AMD and more effector memory T cells than MPNn. The senescence-associated-secretory-phenotype (SASP) is associated with effector T cells. SASP is thought to play a role in driving CLI seen with advancing age. Senescent cells with SASP may damage healthy tissue, including the eye tissues affected in AMD. The finding of increased effector cells in MPNd could implicate a role for adaptive immunity and senescent T cells together with increased CLI in drusen pathophysiology.
Collapse
Affiliation(s)
- Charlotte Liisborg
- Department of Ophthalmology, Zealand University Hospital, Roskilde 4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Hans Carl Hasselbalch
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde 4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
87
|
Weinberger B. Vaccines and Vaccination against SARS-CoV-2: Considerations for the Older Population. Vaccines (Basel) 2021; 9:1435. [PMID: 34960181 PMCID: PMC8704374 DOI: 10.3390/vaccines9121435] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Age is among the most prominent risk factors for developing severe COVID-19 disease, and therefore older adults are a major target group for vaccination against SARS-CoV-2. This review focusses on age-associated aspects of COVID-19 vaccines and vaccination strategies, and summarizes data on immunogenicity, efficacy and effectiveness of the four COVID-19 vaccines, which are licensed in the US and/or Europe; namely, the two mRNA vaccines by BioNTech/Pfizer (BNT162b2) and Moderna (mRNA-1273), and the adenovector vaccines developed by AstraZeneca/University Oxford (ChAdOx1-nCoV-19, AZD1222) and Janssen/Johnson&Johnson (Ad26.COV2-S), respectively. After very high protection rates in the first months after vaccination even in the older population, effectiveness of the vaccines, particularly against asymptomatic infection and mild disease, declined at later time points and with the emergence of virus variants. Many high-income countries have recently started administration of additional doses to older adults and other high-risk groups, whereas other parts of the world are still struggling to acquire and distribute vaccines for primary vaccination. Other vaccines are available in other countries and clinical development for more vaccine candidates is ongoing, but a complete overview of COVID-19 vaccine development is beyond the scope of this article.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
88
|
Gámez-García A, Vazquez BN. Nuclear Sirtuins and the Aging of the Immune System. Genes (Basel) 2021; 12:1856. [PMID: 34946805 PMCID: PMC8701065 DOI: 10.3390/genes12121856] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
The immune system undergoes major changes with age that result in altered immune populations, persistent inflammation, and a reduced ability to mount effective immune responses against pathogens and cancer cells. Aging-associated changes in the immune system are connected to other age-related diseases, suggesting that immune system rejuvenation may provide a feasible route to improving overall health in the elderly. The Sir2 family of proteins, also called sirtuins, have been broadly implicated in genome homeostasis, cellular metabolism, and aging. Sirtuins are key responders to cellular and environmental stress and, in the case of the nuclear sirtuins, they do so by directing responses to chromatin that include gene expression regulation, retrotransposon repression, enhanced DNA damage repair, and faithful chromosome segregation. In the immune system, sirtuins instruct cellular differentiation from hematopoietic precursors and promote leukocyte polarization and activation. In hematopoietic stem cells, sirtuins safeguard quiescence and stemness to prevent cellular exhaustion. Regulation of cytokine production, which, in many cases, requires NF-κB regulation, is the best-characterized mechanism by which sirtuins control innate immune reactivity. In adaptive immunity, sirtuins promote T cell subset differentiation by controlling master regulators, thereby ensuring an optimal balance of helper (Th) T cell-dependent responses. Sirtuins are very important for immune regulation, but the means by which they regulate immunosenescence are not well understood. This review provides an integrative overview of the changes associated with immune system aging and its potential relationship with the roles of nuclear sirtuins in immune cells and overall organismal aging. Given the anti-aging properties of sirtuins, understanding how they contribute to immune responses is of vital importance and may help us develop novel strategies to improve immune performance in the aging organism.
Collapse
Affiliation(s)
- Andrés Gámez-García
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Spain;
| | - Berta N. Vazquez
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Spain;
- Unitat de Citologia i d’Histologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Valles, 08193 Barcelona, Spain
| |
Collapse
|
89
|
Santoro A, Bientinesi E, Monti D. Immunosenescence and inflammaging in the aging process: age-related diseases or longevity? Ageing Res Rev 2021; 71:101422. [PMID: 34391943 DOI: 10.1016/j.arr.2021.101422] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
During aging the immune system (IS) undergoes remarkable changes that collectively are known as immunosenescence. It is a multifactorial and dynamic phenomenon that affects both natural and acquired immunity and plays a critical role in most chronic diseases in older people. For a long time, immunosenescence has been considered detrimental because it may lead to a low-grade, sterile chronic inflammation we proposed to call "inflammaging" and a progressive reduction in the ability to trigger effective antibody and cellular responses against infections and vaccinations. Recently, many scientists revised this negative meaning because it can be considered an essential adaptation/remodeling resulting from the lifelong immunological biography of single individuals from an evolutionary perspective. Inflammaging can be considered an adaptive process because it can trigger an anti-inflammatory response to counteract the age-related pro-inflammatory environment. Centenarians represent a valuable model to study the beneficial changes occurring in the IS with age. These extraordinary individuals reached the extreme limits of human life by slowing down the aging process and, in most cases, delaying, avoiding or surviving the major age-associated diseases. They indeed show a complex and heterogeneous phenotype determined by an improved ability to adapt and remodel in response to harmful stimuli. This review aims to point out the intimate relationship between immunosenescence and inflammaging and how these processes impact unsuccessful aging rather than longevity. We also describe the gut microbiota age-related changes as one of the significant triggers of inflammaging and the sex/gender differences in the immune system of the elderly, contributing to the sex/gender disparity in terms of epidemiology, pathophysiology, symptoms and severity of age-related diseases. Finally, we discuss how these phenomena could influence the susceptibility to COVID-19 infection.
Collapse
|
90
|
Eschborn M, Pawlitzki M, Wirth T, Nelke C, Pfeuffer S, Schulte-Mecklenbeck A, Lohmann L, Rolfes L, Pape K, Eveslage M, Bittner S, Gross CC, Ruck T, Wiendl H, Meuth SG, Klotz L. Evaluation of Age-Dependent Immune Signatures in Patients With Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1094. [PMID: 34667129 DOI: 10.1212/nxi.0000000000001094] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES In MS, an age-related decline in disease activity and a decreased efficacy of disease-modifying treatment have been linked to immunosenescence, a state of cellular dysfunction associated with chronic inflammation. METHODS To evaluate age-related immunologic alterations in MS, we compared immune signatures in peripheral blood (PB) and CSF by flow cytometry in patients with relapsing-remitting (RR) (PB n = 38; CSF n = 51) and primary progressive (PP) MS (PB n = 40; CSF n = 36) and respective controls (PB n = 40; CSF n = 85). RESULTS Analysis revealed significant age-related changes in blood immune cell composition, especially in the CD8 T-cell compartment of healthy donors (HDs) and patients with MS. However, HDs displayed a strong age-dependent decline in the expression of the immunoregulatory molecules KLRG1, LAG3, and CTLA-4 on memory CD8 T cells, whereas this age-dependent reduction was completely abrogated in patients with MS. An age-dependent increase in the expression of the costimulatory molecule CD226 on memory CD8 T cells was absent in patients with MS. CD226 expression correlated with disability in younger (≤50 years) patients with MS. CSF analysis revealed a significant age-dependent decline in various immune cell populations in PPMS but not RRMS, suggesting a differential effect of aging on the intrathecal compartment in PPMS. DISCUSSION Our data illustrate that aging in MS is associated with a dysbalance between costimulatory and immunoregulatory signals provided by CD8 T cells favoring a proinflammatory phenotype and, more importantly, a pattern of premature immune aging in the CD8 T-cell compartment of young patients with MS with potential implications for disease severity.
Collapse
Affiliation(s)
- Melanie Eschborn
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Marc Pawlitzki
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Timo Wirth
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Christopher Nelke
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Steffen Pfeuffer
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Andreas Schulte-Mecklenbeck
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Lisa Lohmann
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Leoni Rolfes
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Katrin Pape
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Maria Eveslage
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Stefan Bittner
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Catharina C Gross
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Tobias Ruck
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Heinz Wiendl
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Sven G Meuth
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Luisa Klotz
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
91
|
Induri SNR, Kansara P, Thomas SC, Xu F, Saxena D, Li X. The Gut Microbiome, Metformin, and Aging. Annu Rev Pharmacol Toxicol 2021; 62:85-108. [PMID: 34449247 DOI: 10.1146/annurev-pharmtox-051920-093829] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metformin has been extensively used for the treatment of type 2 diabetes, and it may also promote healthy aging. Despite its widespread use and versatility, metformin's mechanisms of action remain elusive. The gut typically harbors thousands of bacterial species, and as the concentration of metformin is much higher in the gut as compared to plasma, it is plausible that microbiome-drug-host interactions may influence the functions of metformin. Detrimental perturbations in the aging gut microbiome lead to the activation of the innate immune response concomitant with chronic low-grade inflammation. With the effectiveness of metformin in diabetes and antiaging varying among individuals, there is reason to believe that the gut microbiome plays a role in the efficacy of metformin. Metformin has been implicated in the promotion and maintenance of a healthy gut microbiome and reduces many age-related degenerative pathologies. Mechanistic understanding of metformin in the promotion of a healthy gut microbiome and aging will require a systems-level approach. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sri Nitya Reddy Induri
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Payalben Kansara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; .,Department of Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| |
Collapse
|
92
|
Li T, Wang X, Zhuang X, Wang H, Li A, Huang L, Zhang X, Xue Y, Wei F, Ma C. Baseline characteristics and changes of biomarkers in disease course predict prognosis of patients with COVID-19. Intern Emerg Med 2021; 16:1165-1172. [PMID: 33565034 PMCID: PMC7872821 DOI: 10.1007/s11739-020-02560-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/31/2020] [Indexed: 01/08/2023]
Abstract
The outbreak of coronavirus disease (COVID-19) has brought great challenges to the world. The objectives of this study were to describe the baseline characteristics and changes of biomarkers of these COVID-19 patients and identify predictive value of the above markers for patient death. Using patient death as the observational endpoints, clinical data of inpatients in a special ward for COVID-19 in Wuhan, China were retrospectively collected. Univariate and multivariate Cox regression analyses were used to evaluate prognostic value of baseline characteristics and laboratory data changes. This study included clinical data of 75 patients. Age, c-reactive protein (CRP) and interleukin-6 levels were independent predictors of patient death. Survivors were characterized as having declining neutrophil counts, D-dimer, N-terminal pronatriuretic peptide, troponin I (TnI) and c-reactive protein levels, while counts of lymphocyte gradually came back. Non-survivors were characterized with increasing white blood cell counts (WBC) and neutrophil counts. Changes of WBC, TnI and interleukin-6 were also independently associated with patient death. Older age, baseline CRP and IL-6 levels may be used as meaningful predictors to identify patients with poor prognosis. Changes of biomarkers should be closely monitored in the management of patients with COVID-19, while constantly increasing levels of WBC, TnI and interleukin-6 in the disease course also predict patient death.
Collapse
Affiliation(s)
- Tao Li
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Infectious Disease and Hepatology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Wang
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Road, Jinan, China
| | - Xianghua Zhuang
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Wang
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ai Li
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Laigang Huang
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Rehabilitation Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xingqian Zhang
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Road, Jinan, China
| | - Yan Xue
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Infectious Disease and Hepatology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fengtao Wei
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Road, Jinan, China.
| | - Cheng'en Ma
- Medical Team To Hubei Province, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Intensive Care Unit, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Road, Jinan, China.
| |
Collapse
|
93
|
Badedi M, Darraj H, Alnami AQ, Makrami A, Mahfouz MS, Alhazmi K, Mahmoud N, Mosa H. Epidemiological and Clinical Characteristics of Deceased COVID-19 Patients. Int J Gen Med 2021; 14:3809-3819. [PMID: 34335047 PMCID: PMC8317935 DOI: 10.2147/ijgm.s320713] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Fatalities due to coronavirus disease 2019 (COVID-19) continue to increase, and information on the epidemiological and clinical characteristics of deceased patients who were hospitalized with COVID-19 is limited in the Arab region. The current study aimed to address this gap. Methods Three hundred and four Saudi patients in Jazan Region, Saudi Arabia, who died after being hospitalized with COVID-19 between July 1, 2020, and December 31, 2020, were analyzed in this retrospective cohort study. Results A greater proportion of male patients (59%), compared to female patients (41%), died due to COVID-19. Just over half (55%) of the deaths due to COVID-19 affected patients aged ≥65 years. More than two-thirds of the deceased COVID-19 patients had diabetes (70%) and hypertension (69%); other comorbidities were obesity (30%), heart disease (30%), and chronic kidney disease (14%). Dyspnea (91%), cough (80%), and fever (70%) were the most frequently reported clinical symptoms. Eighty-five per cent of COVID-19 deaths occurred in patients admitted to the intensive care unit (ICU), and 90% of the patients required mechanical ventilation. Typically, lymphopenia, and neutrophilia were observed on admission and 24 hours prior to death. Creatinine and serum ferritin levels and erythrocyte sedimentation rate and D-dimer plasma levels increased significantly following infection with COVID-19. Lung infiltrates and pulmonary opacity (83%) were the most common findings on chest X-ray. Respiratory failure (70%) and acute respiratory distress syndrome (52%) were the leading complications to death. Logistic and Cox regression revealed that a higher age, smoking, high creatinine and aspartate transaminase levels, and respiratory failure were significantly associated with the risk of mortality during the early stay in hospitals. Conclusion The proportion of comorbidities was high in deceased patients who were hospitalized with COVID-19 in Jazan region, Saudi Arabia. A higher age, smoking, and respiratory failure were significant predictors of mortality during the early stay in hospitals.
Collapse
Affiliation(s)
- Mohammed Badedi
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| | - Hussain Darraj
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| | - Awaji Qasem Alnami
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| | - Ali Makrami
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| | | | | | - Nahid Mahmoud
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| | - Halimh Mosa
- General Directorate of Health Affairs, Jazan, Jazan Region, Saudi Arabia
| |
Collapse
|
94
|
Picón C, Tejeda-Velarde A, Fernández-Velasco JI, Comabella M, Álvarez-Lafuente R, Quintana E, Sainz de la Maza S, Monreal E, Villarrubia N, Álvarez-Cermeño JC, Domínguez-Mozo MI, Ramió-Torrentà L, Rodríguez-Martín E, Roldán E, Aladro Y, Medina S, Espiño M, Masjuan J, Matute-Blanch C, Muñoz-San Martín M, Espejo C, Guaza C, Muriel A, Costa-Frossard L, Villar LM. Identification of the Immunological Changes Appearing in the CSF During the Early Immunosenescence Process Occurring in Multiple Sclerosis. Front Immunol 2021; 12:685139. [PMID: 34322119 PMCID: PMC8311928 DOI: 10.3389/fimmu.2021.685139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/24/2021] [Indexed: 01/21/2023] Open
Abstract
Patients with multiple sclerosis (MS) suffer with age an early immunosenescence process, which influence the treatment response and increase the risk of infections. We explored whether lipid-specific oligoclonal IgM bands (LS-OCMB) associated with highly inflammatory MS modify the immunological profile induced by age in MS. This cross-sectional study included 263 MS patients who were classified according to the presence (M+, n=72) and absence (M-, n=191) of LS-OCMB. CSF cellular subsets and molecules implicated in immunosenescence were explored. In M- patients, aging induced remarkable decreases in absolute CSF counts of CD4+ and CD8+ T lymphocytes, including Th1 and Th17 cells, and of B cells, including those secreting TNF-alpha. It also increased serum anti-CMV IgG antibody titers (indicative of immunosenescence) and CSF CHI3L1 levels (related to astrocyte activation). In contrast, M+ patients showed an age-associated increase of TIM-3 (a biomarker of T cell exhaustion) and increased values of CHI3L1, independently of age. Finally, in both groups, age induced an increase in CSF levels of PD-L1 (an inductor of T cell tolerance) and activin A (part of the senescence-associated secretome and related to inflammaging). These changes were independent of the disease duration. Finally, this resulted in augmented disability. In summary, all MS patients experience with age a modest induction of T-cell tolerance and an activation of the innate immunity, resulting in increased disability. Additionally, M- patients show clear decreases in CSF lymphocyte numbers, which could increase the risk of infections. Thus, age and immunological status are important for tailoring effective therapies in MS.
Collapse
Affiliation(s)
- Carmen Picón
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
- Department of Brain Science, Imperial College London, London, United Kingdom
| | - Amalia Tejeda-Velarde
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - José Ignacio Fernández-Velasco
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d’ Esclerosi Múltiple de Catalunya (Cemcat), Vall d’ Hebron Institut de Recerca, Hospital Universitari Vall d’ Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto Álvarez-Lafuente
- Department of Neurology, Hospital Clínico San Carlos, Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), REEM, Madrid, Spain
| | - Ester Quintana
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Girona, Medical Sciences Department, Universitat de Girona, REEM, Girona, Spain
| | | | - Enric Monreal
- Department of Neurology, Hospital Universitario Ramón y Cajal, IRYCIS, REEM, Madrid, Spain
| | - Noelia Villarrubia
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | | | - María Inmaculada Domínguez-Mozo
- Department of Neurology, Hospital Clínico San Carlos, Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), REEM, Madrid, Spain
| | - Lluís Ramió-Torrentà
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Girona, Medical Sciences Department, Universitat de Girona, REEM, Girona, Spain
| | - Eulalia Rodríguez-Martín
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Ernesto Roldán
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Yolanda Aladro
- Department of Neurology, Hospital Universitario de Getafe, REEM, Madrid, Spain
| | - Silvia Medina
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Mercedes Espiño
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Jaime Masjuan
- Department of Neurology, Hospital Universitario Ramón y Cajal, IRYCIS, REEM, Madrid, Spain
| | - Clara Matute-Blanch
- Servei de Neurologia-Neuroimmunologia, Centre d’ Esclerosi Múltiple de Catalunya (Cemcat), Vall d’ Hebron Institut de Recerca, Hospital Universitari Vall d’ Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Muñoz-San Martín
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Girona, Medical Sciences Department, Universitat de Girona, REEM, Girona, Spain
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d’ Esclerosi Múltiple de Catalunya (Cemcat), Vall d’ Hebron Institut de Recerca, Hospital Universitari Vall d’ Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carmen Guaza
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain
| | - Alfonso Muriel
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal, IRYCIS, CIBERESP, Nursing Department, Universidad de Alcalá, Madrid, Spain
| | | | - Luisa María Villar
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacón Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| |
Collapse
|
95
|
Chen Y, Zhi H, Zhang K, Zhu G, Liu L, Yan X, Cai Z, Zhao C, Hu Z. Combined predictive performance of age and neutrophilic percentage on admission for severe novel coronavirus disease 2019. Int J Clin Pract 2021; 75:e14257. [PMID: 33884718 PMCID: PMC8250231 DOI: 10.1111/ijcp.14257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Novel coronavirus disease 2019 (COVID-19) poses a huge threat to the global public health. This study aimed to identify predictive indicators of severe COVID-19. METHODS We retrospectively collected clinical data on hospital admission of all patients with severe COVID-19 and a control cohort (1:1) of gender- and hospital-matched patients with mild disease from 13 designated hospitals in the Hebei Province between 22 January and 15 April 2020. RESULTS A total of 104 patients (52 with severe COVID-19 and 52 with mild disease) were included. Only age, fever, duration from symptom onset to confirmation, respiratory rate, percutaneous oxygen saturation (SpO2 ) and neutrophilic percentage were independent predictors of severe COVID-19. Age and neutrophilic percentage performed best in predicting severe COVID-19, followed by SpO2 . 'Age + neutrophilic percentage' (the sum of age and neutrophilic percentage) (area under the curve [AUC] 0.900, 95% confidence interval [CI] 0.825-0.950, P < .001) and 'age and neutrophilic percentage' (the prediction probability of age and neutrophilic percentage for severe type obtained by logistic regression analysis) (AUC 0.899, 95% CI 0.824-0.949, P < .001) had excellent predictive performance for severe type. The optimal cut-off for 'age + neutrophilic percentage' was >119.1 (sensitivity, 86.5%; specificity, 84.6%; Youden index, 0.712). CONCLUSION The combination of age and neutrophil percentage could effectively predict severe COVID-19. The sum of age and neutrophil percentage was recommended for clinical application because of its excellent predictive value and practicability. TRAIL REGISTRATION China Clinical Trial Registry, number ChiCTR2000030226. Registered 26 February 2020-Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=49855.
Collapse
Affiliation(s)
- Yuhong Chen
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| | - Haijun Zhi
- Emergency DepartmentCangzhou Central HospitalCangzhouChina
| | - Kun Zhang
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| | - Guijun Zhu
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| | - Lixia Liu
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| | - Xixin Yan
- Department of RespirationHebei Medical University Second Affiliated HospitalShijiazhuangChina
| | - Zhigang Cai
- Department of RespirationHebei Medical University Second Affiliated HospitalShijiazhuangChina
| | - Congcong Zhao
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| | - Zhenjie Hu
- Department of Intensive Care UnitHebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor HospitalShijiazhuangChina
| |
Collapse
|
96
|
Löhr P, Schiele S, Arndt TT, Grützner S, Claus R, Römmele C, Müller G, Schmid C, Dennehy KM, Rank A. Impact of age and gender on lymphocyte subset counts in patients with COVID-19. Cytometry A 2021; 103:127-135. [PMID: 34125495 PMCID: PMC8426831 DOI: 10.1002/cyto.a.24470] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
In symptomatic patients with acute Coronavirus disease 2019 (COVID-19), lymphocytopenia is one of the most prominent laboratory findings. However, to date age and gender have not been considered in assessment of COVID-19-related cell count alterations. In this study, the impact of COVID-19 as well as age and gender on a large variety of lymphocyte subsets was analyzed in 33 COVID-19 patients and compared with cell counts in 50 healthy humans. We confirm that cell counts of total lymphocytes, B, NK, cytotoxic and helper T cells are reduced in patients with severe COVID-19, and this tendency was observed in patients with moderate COVID-19. Decreased cell counts were also found in all subsets of these cell types, except for CD4+ and CD8+ effector memory RA+ (EMRA) and terminal effector CD8+ cells. In multivariate analysis however, we show that in addition to COVID-19, there is an age-dependent reduction of total, central memory (CM), and early CD8+ cell subsets, as well as naïve, CM, and regulatory CD4+ cell subsets. Remarkably, reduced naïve CD8+ cell counts could be attributed to age alone, and not to COVID-19. By contrast, decreases in other subsets could be largely attributed to COVID-19, and only partly to age. In addition to COVID-19, male gender was a major factor influencing lower counts of CD3+ and CD4+ lymphocyte numbers. Our study confirms that cell counts of lymphocytes and their subsets are reduced in patients with COVID-19, but that age and gender must be considered when interpreting the altered cell counts.
Collapse
Affiliation(s)
- Phillip Löhr
- Department of Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Stefan Schiele
- Institute of MathematicsFaculty of Mathematics and Natural Sciences, University of AugsburgAugsburgGermany
| | - Tim Tobias Arndt
- Institute of MathematicsFaculty of Mathematics and Natural Sciences, University of AugsburgAugsburgGermany
| | - Stefanie Grützner
- Institute for Transfusion Medicine and HaemostasisMedical Faculty, University of AugsburgAugsburgGermany
| | - Rainer Claus
- Department of Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Christoph Römmele
- Department of Gastroenterology and Infectious Diseases, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Gernot Müller
- Institute of MathematicsFaculty of Mathematics and Natural Sciences, University of AugsburgAugsburgGermany
| | - Christoph Schmid
- Department of Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Kevin M. Dennehy
- Institute for Laboratory Medicine and MicrobiologyMedical Faculty, University of AugsburgAugsburgGermany
| | - Andreas Rank
- Department of Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| |
Collapse
|
97
|
Tu WJ, McCuaig RD, Melino M, Rawle DJ, Le TT, Yan K, Suhrbier A, Johnston RL, Koufariotis LT, Waddell N, Cross EM, Tsimbalyuk S, Bain A, Ahern E, Collinson N, Phipps S, Forwood JK, Seddiki N, Rao S. Targeting novel LSD1-dependent ACE2 demethylation domains inhibits SARS-CoV-2 replication. Cell Discov 2021; 7:37. [PMID: 34031383 PMCID: PMC8143069 DOI: 10.1038/s41421-021-00279-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Treatment options for COVID-19 remain limited, especially during the early or asymptomatic phase. Here, we report a novel SARS-CoV-2 viral replication mechanism mediated by interactions between ACE2 and the epigenetic eraser enzyme LSD1, and its interplay with the nuclear shuttling importin pathway. Recent studies have shown a critical role for the importin pathway in SARS-CoV-2 infection, and many RNA viruses hijack this axis to re-direct host cell transcription. LSD1 colocalized with ACE2 at the cell surface to maintain demethylated SARS-CoV-2 spike receptor-binding domain lysine 31 to promote virus-ACE2 interactions. Two newly developed peptide inhibitors competitively inhibited virus-ACE2 interactions, and demethylase access to significantly inhibit viral replication. Similar to some other predominantly plasma membrane proteins, ACE2 had a novel nuclear function: its cytoplasmic domain harbors a nuclear shuttling domain, which when demethylated by LSD1 promoted importin-α-dependent nuclear ACE2 entry following infection to regulate active transcription. A novel, cell permeable ACE2 peptide inhibitor prevented ACE2 nuclear entry, significantly inhibiting viral replication in SARS-CoV-2-infected cell lines, outperforming other LSD1 inhibitors. These data raise the prospect of post-exposure prophylaxis for SARS-CoV-2, either through repurposed LSD1 inhibitors or new, nuclear-specific ACE2 inhibitors.
Collapse
Affiliation(s)
- Wen Juan Tu
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Robert D McCuaig
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Michelle Melino
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Daniel J Rawle
- The Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Thuy T Le
- The Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kexin Yan
- The Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Suhrbier
- The Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rebecca L Johnston
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lambros T Koufariotis
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicola Waddell
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Emily M Cross
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Sofiya Tsimbalyuk
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Amanda Bain
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Elizabeth Ahern
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
- School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Natasha Collinson
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jade K Forwood
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Nabila Seddiki
- U955, Equipe 16, Créteil, France
- Université Paris-Est Créteil, Faculté de médecine, Créteil, France
- Vaccine Research Institute (VRI), Créteil, France
| | - Sudha Rao
- Gene Regulation and Translational Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
98
|
AlSafar H, Grant WB, Hijazi R, Uddin M, Alkaabi N, Tay G, Mahboub B, Al Anouti F. COVID-19 Disease Severity and Death in Relation to Vitamin D Status among SARS-CoV-2-Positive UAE Residents. Nutrients 2021; 13:1714. [PMID: 34069412 PMCID: PMC8159141 DOI: 10.3390/nu13051714] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
Insufficient blood levels of the neurohormone vitamin D are associated with increased risk of COVID-19 severity and mortality. Despite the global rollout of vaccinations and promising preliminary results, the focus remains on additional preventive measures to manage COVID-19. Results conflict on vitamin D's plausible role in preventing and treating COVID-19. We examined the relation between vitamin D status and COVID-19 severity and mortality among the multiethnic population of the United Arab Emirates. Our observational study used data for 522 participants who tested positive for SARS-CoV-2 at one of the main hospitals in Abu Dhabi and Dubai. Only 464 of those patients were included for data analysis. Demographic and clinical data were retrospectively analyzed. Serum samples immediately drawn at the first hospital visit were used to measure serum 25-hydroxyvitamin D [25(OH)D] concentrations through automated electrochemiluminescence. Levels < 12 ng/mL were significantly associated with higher risk of severe COVID-19 infection and of death. Age was the only other independent risk factor, whereas comorbidities and smoking did not contribute to the outcomes upon adjustment. Sex of patients was not an important predictor for severity or death. Our study is the first conducted in the UAE to measure 25(OH)D levels in SARS-CoV-2-positive patients and confirm the association of levels < 12 ng/mL with COVID-19 severity and mortality.
Collapse
Affiliation(s)
- Habiba AlSafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (H.A.); (G.T.)
- Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
- Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - William B. Grant
- Sunlight, Nutrition and Health Research Center, P.O. Box 641603, San Francisco, CA 94164-1603, USA;
| | - Rafiq Hijazi
- Department of Mathematics and Statistics, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates;
| | - Maimunah Uddin
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates; (M.U.); (N.A.)
| | - Nawal Alkaabi
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates; (M.U.); (N.A.)
| | - Guan Tay
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (H.A.); (G.T.)
- Division of Psychiatry, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Bassam Mahboub
- Dubai Health Authority, Rashid Hospital, Dubai, United Arab Emirates;
| | - Fatme Al Anouti
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates
| |
Collapse
|
99
|
Mittelbrunn M, Kroemer G. Hallmarks of T cell aging. Nat Immunol 2021; 22:687-698. [PMID: 33986548 DOI: 10.1038/s41590-021-00927-z] [Citation(s) in RCA: 238] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
The aged adaptive immune system is characterized by progressive dysfunction as well as increased autoimmunity. This decline is responsible for elevated susceptibility to infection and cancer, as well as decreased vaccination efficacy. Recent evidence indicates that CD4+ T cell-intrinsic alteratins contribute to chronic inflammation and are sufficient to accelerate an organism-wide aging phenotype, supporting the idea that T cell aging plays a major role in body-wide deterioration. In this Review, we propose ten molecular hallmarks to represent common denominators of T cell aging. These hallmarks are grouped into four primary hallmarks (thymic involution, mitochondrial dysfunction, genetic and epigenetic alterations, and loss of proteostasis) and four secondary hallmarks (reduction of the TCR repertoire, naive-memory imbalance, T cell senescence, and lack of effector plasticity), and together they explain the manifestation of the two integrative hallmarks (immunodeficiency and inflammaging). A major challenge now is weighing the relative impact of these hallmarks on T cell aging and understanding their interconnections, with the final goal of defining molecular targets for interventions in the aging process.
Collapse
Affiliation(s)
- Maria Mittelbrunn
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain. .,Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France. .,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China. .,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
100
|
Biswas A, Mandal RS, Chakraborty S, Maiti G. Tapping the immunological imprints to design chimeric SARS-CoV-2 vaccine for elderly population. Int Rev Immunol 2021; 41:448-463. [PMID: 33978550 PMCID: PMC8127164 DOI: 10.1080/08830185.2021.1925267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023]
Abstract
The impact of SARS-CoV-2 and COVID-19 disease susceptibility varies depending on the age and health status of an individual. Currently, there are more than 140 COVID-19 vaccines under development. However, the challenge will be to induce an effective immune response in the elderly population. Analysis of B cell epitopes indicates the minor role of the stalk domain of spike protein in viral neutralization due to low surface accessibility. Nevertheless, the accumulation of mutations in the receptor-binding domain (RBD) might reduce the vaccine efficacy in all age groups. We also propose the concept of chimeric vaccines based on the co-expression of SARS-CoV-2 spike and influenza hemagglutinin (HA) and matrix protein 1 (M1) proteins to generate chimeric virus-like particles (VLP). This review discusses the possible approaches by which influenza-specific memory repertoire developed during the lifetime of the elderly populations can converge to mount an effective immune response against the SARS-CoV-2 spike protein with the possibilities of designing single vaccines for COVID-19 and influenza. HighlightsImmunosenescence aggravates COVID-19 symptoms in elderly individuals.Low immunogenicity of SARS-CoV-2 vaccines in elderly population.Tapping the memory T and B cell repertoire in elderly can enhance vaccine efficiency.Chimeric vaccines can mount effective immune response against COVID-19 in elderly.Chimeric vaccines co-express SARS-CoV-2 spike and influenza HA and M1 proteins.
Collapse
Affiliation(s)
- Asim Biswas
- Department of Ophthalmology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rahul Subhra Mandal
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suparna Chakraborty
- Division of Clinical Medicine, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | |
Collapse
|