1
|
Campos FA, Wikberg EC, Orkin JD, Park Y, Snyder-Mackler N, Cheves Hernandez S, Lopez Navarro R, Fedigan LM, Gurven M, Higham JP, Jack KM, Melin AD. Wild capuchin monkeys as a model system for investigating the social and ecological determinants of ageing. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230482. [PMID: 39463253 PMCID: PMC11513648 DOI: 10.1098/rstb.2023.0482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/29/2024] [Accepted: 05/10/2024] [Indexed: 10/29/2024] Open
Abstract
Studying biological ageing in animal models can circumvent some of the confounds exhibited by studies of human ageing. Ageing research in non-human primates has provided invaluable insights into human lifespan and healthspan. Yet data on patterns of ageing from wild primates remain relatively scarce, centred around a few populations of catarrhine species. Here, we introduce the white-faced capuchin, a long-lived platyrrhine primate, as a promising new model system for ageing research. Like humans, capuchins are highly social, omnivorous generalists, whose healthspan and lifespan relative to body size exceed that of other non-human primate model species. We review recent insights from capuchin ageing biology and outline our expanding, integrative research programme that combines metrics of the social and physical environments with physical, physiological and molecular hallmarks of ageing across the natural life courses of multiple longitudinally tracked individuals. By increasing the taxonomic breadth of well-studied primate ageing models, we generate new insights, increase the comparative value of existing datasets to geroscience and work towards the collective goal of developing accurate, non-invasive and reliable biomarkers with high potential for standardization across field sites and species, enhancing the translatability of primate studies.This article is part of the discussion meeting issue 'Understanding age and society using natural populations'.
Collapse
Affiliation(s)
- Fernando A. Campos
- Department of Anthropology, University of Texas at San Antonio, San Antonio, TX78249, USA
| | - Eva C. Wikberg
- Department of Anthropology, University of Texas at San Antonio, San Antonio, TX78249, USA
| | - Joseph D. Orkin
- Département d’anthropologie, Université de Montréal, Montréal, QuébecH3T 1N8, Canada
- Département de sciences biologiques, Université de Montréal, Montréal, QuébecH2V 0B3, Canada
| | - Yeonjoo Park
- Department of Management Science and Statistics, University of Texas at San Antonio, San Antonio, TX78249, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, School of Life Sciences, School of Human Evolution and Social Change, Arizona State University, Tempe, AZ85287, USA
| | | | | | - Linda M. Fedigan
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AlbertaT2N 1N4, Canada
| | - Michael Gurven
- Department of Anthropology, University of California, Santa Barbara, CA93106, USA
| | - James P. Higham
- Department of Anthropology, New York University, NY10003, USA
| | - Katharine M. Jack
- Department of Anthropology, Tulane University, New Orleans, LA70118, USA
| | - Amanda D. Melin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AlbertaT2N 1N4, Canada
- Department of Medical Genetics, University of Calgary, Calgary, AlbertaT2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AlbertaT2N 4N1, Canada
| |
Collapse
|
2
|
Durán-Galea A, Cristóbal-Verdejo JI, Macías-García B, Nicolás-Barceló P, Barrera-Chacón R, Ruiz-Tapia P, Zaragoza-Bayle MC, Duque-Carrasco FJ. Determination of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio and systemic immune-inflammation index in dogs with leptospirosis. Vet Res Commun 2024; 48:4105-4111. [PMID: 39254739 DOI: 10.1007/s11259-024-10469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/14/2024] [Indexed: 09/11/2024]
Abstract
Leptospirosis is a globally distributed zoonosis with multisystemic involvement in canine species, capable of causing a pulmonary hemorrhagic syndrome (LPHS) in the most severe cases. In humans, the neutrophil to lymphocyte ratio (NLR), platelets to lymphocytes (PLR) and systemic immune-inflammation index (SII) have been described as predictors of morbidity and mortality in various pathologies, but no such studies have been developed for canine leptospirosis. Hence, we aimed to assess the usefulness of NLR, PLR and SII in dogs affected with leptospirosis, focusing on those that died or survived after hospitalization, whether or not they developed LPHS. The leptospirosis group was composed by 36 dogs while the control group consisted of 32 healthy dogs. The NLR, associated with inflammation, demonstrated a threefold or greater increase in all leptospirosis groups compared to the control group (median 2.44 ± 1.66) (developing or not LPHS). Dogs that died (median 67.78 ± 158.67), developed LHPS (median 85.17 ± 143.77), or both developed LHPS and died (median 67.78 ± 155,14) had a lower PLR in comparison to the control group (median 101,82 ± 53,75) and the rest of groups, but no statistically significant differences were observed (p > 0.05). The SII was higher in leptospirosis-affected dogs that survived (median 1356,92 ± 2726,29) and statistically significant differences were observed in those who did not develop LPHS (median 1770,41 ± 2630,77; p < 0.05) compared to the control group (median 555,21 ± 313,26). Our data shows that NLR may be used as inflammation indicator, while more studies are needed for PLR and SII in canine leptospirosis.
Collapse
Affiliation(s)
- A Durán-Galea
- Universidad de Extremadura, Hospital Clínico Veterinario, Avenue University n/n, 10003, Cáceres, Spain
| | - J I Cristóbal-Verdejo
- Universidad de Extremadura, Hospital Clínico Veterinario, Avenue University n/n, 10003, Cáceres, Spain.
| | - B Macías-García
- Department of Animal Medicine, University of Extremadura, Avenue University n/n, 10003, Cáceres, Spain
| | - P Nicolás-Barceló
- Universidad de Extremadura, Hospital Clínico Veterinario, Avenue University n/n, 10003, Cáceres, Spain
| | - R Barrera-Chacón
- Department of Animal Medicine, University of Extremadura, Avenue University n/n, 10003, Cáceres, Spain
| | - P Ruiz-Tapia
- Department of Animal Medicine, University of Extremadura, Avenue University n/n, 10003, Cáceres, Spain
| | - M C Zaragoza-Bayle
- Department of Animal Medicine, University of Extremadura, Avenue University n/n, 10003, Cáceres, Spain
| | - F J Duque-Carrasco
- Department of Animal Medicine, University of Extremadura, Avenue University n/n, 10003, Cáceres, Spain
| |
Collapse
|
3
|
Serrano-Villar S, Moltó-Marhuenda J, Montero-Alonso M, Diaz-Torné C, López-Cavanillas M, Pérez de Isla L. Knowledge, attitudes and practices in HIV-related chronic inflammation and cardiovascular risk in Spain. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2024; 42:484-491. [PMID: 37945466 DOI: 10.1016/j.eimce.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/27/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND People with HIV (PWH) in suppressive antiretroviral treatment suffer from chronic inflammation-related comorbidities, mainly cardiovascular diseases. However, given the lack of specific evidence about inflammation in PWH, clinical guidelines do not provide recommendations for the management of this issue. To date, physician awareness of inflammation in PWH remains unclear. We analyzed the knowledge, attitudes, and practices (KAP) related to inflammation, particularly in the clinical management of PWH, of infectious disease specialists (IDS)/internists compared to other specialists treating inflammation directly (rheumatologists) or its cardiovascular consequences (cardiologists). METHODS A committee of IDS/internists treating PWH, cardiologists, and rheumatologists designed the KAP questionnaire. The survey was completed by 405 participants (135 physicians per specialty) stratified by Spanish geography, hospital size, and number of PWH under care (IDS/internists only). RESULTS IDS/internists treating PWH scored higher than cardiologists and rheumatologists on knowledge of inflammation (5.5±1.4 out of 8 points vs. 5.2±1.3 and 4.6±1.4 points, respectively; p<0.05). Nevertheless, rheumatologists showed the most proactive attitude toward inflammation (i.e., biomarkers monitoring, anti-inflammatory drug prescription and cardiologist referral), followed by cardiologists and IDS/internists (13±3 of a total of 16 points vs. 11±3 and 10±3.3 points, respectively; p<0.05), irrespective of hospital size and years of experience. Most IDS/internists (59%) include inflammation in their therapeutic recommendations. However, in IDS/internists treating PWH, we observed a negative correlation between years of experience and concern about the clinical consequences of inflammation. CONCLUSION Our findings show that, compared to other specialists, infectious disease specialists/internists have high knowledge about inflammation in HIV infection, but, in the absence of scientific evidence to base their decisions on inflammatory markers, the therapeutic implications are scarce. The results support the need for more evidence on the monitoring and treatment of inflammation in PWH.
Collapse
Affiliation(s)
- Sergio Serrano-Villar
- Department of Infectious Diseases, Ramon y Cajal Hospital and IRYCIS, Madrid, Spain; Centro de Investigación en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - José Moltó-Marhuenda
- Centro de Investigación en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain; Fight AIDS and Infectious Diseases Foundation, Badalona, Barcelona, Spain
| | | | - Cesar Diaz-Torné
- Rheumatology Department, Sant Pau i Santa Creu Hospital, Barcelona, Spain
| | | | - Leopoldo Pérez de Isla
- Cardiology Department, San Carlos Clinic Hospital, Universidad Complutense, Madrid, Spain
| |
Collapse
|
4
|
Liberale L, Torino C, Pizzini P, Mezzatesta S, D'Arrigo G, Gori M, Carbone F, Schiavetta E, Cugno V, Cabri M, Sgura C, Maioli E, Mbarga D, Rubini G, Tirandi A, Ramoni D, Mallamaci F, Tripepi G, Zoccali C, Montecucco F. Plasma levels of myeloperoxidase and resistin independently predict mortality in dialysis patients. Eur J Intern Med 2024; 129:87-92. [PMID: 39019736 DOI: 10.1016/j.ejim.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND In patients with kidney failure (KF) undergoing dialysis, neutrophils are dysfunctionally activated. Such chronic activation does not correspond to increased protection against infections and is thought to cause direct vascular damage accounting for the higher incidence of cardiovascular (CV) events. We hypothesized that circulating levels of neutrophil degranulation products (i.e. myeloperoxidase (MPO) and resistin) can predict overall and CV-specific mortality in dialysis patients. METHODS MPO and resistin levels were assessed in plasma samples from n = 1182 dialysis patients who were followed-up for median 2.9 years (IQR: 1.7-4.2). RESULTS Patients were 65 ± 14 (SD) years old and 36 % women. Median value of MPO and resistin were 78 ng/mL (IQR: 54 - 123) and 72 ng/mL (IQR: 46 - 110), respectively. MPO and resistin levels correlated with biomarkers of organ damage, nutritional status and inflammation. Both MPO and resistin levels predicted all-cause mortality even after adjustment for traditional risk factors and inflammation, nutritional and KF-related indexes (MPO, HRfor 1 ln unit increase: 1.26, 95 %CI 1.11 - 1.42, P < 0.001; Resistin, HRfor 1 ln unit increase: 1.25, 95 %CI 1.09 - 1.44, P = 0.001). Similarly, their predictive ability held true also for CV death (MPO, HRfor 1 ln unit increase: 1.19, 95 %CI 1.01 - 1.41, P = 0.04; Resistin, HRfor 1 ln unit increase: 1.29, 95 %CI 1.07 - 1.56, P = 0.007). CONCLUSION Plasma levels of MPO and resistin correlate with prospective overall and CV-specific mortality risk in KF patients undergoing dialysis and might be useful prognostic tools. Mediators of inflammation may be potential target to improve survival of those patients.
Collapse
Affiliation(s)
- Luca Liberale
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Claudia Torino
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Patrizia Pizzini
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Sabrina Mezzatesta
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Graziella D'Arrigo
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | | | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Elisa Schiavetta
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Valeria Cugno
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Mara Cabri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Cosimo Sgura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Elia Maioli
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Danielle Mbarga
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Gianluca Rubini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Francesca Mallamaci
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy; Nephrology, Hypertension and Renal Transplantation Unit, Grande Ospedale Metropolitano, Reggio Cal. Italy
| | - Giovanni Tripepi
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, USA; IPNET, c/o Nefrologia del Grande Ospedale Metropolitano, Reggio Cal. Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
5
|
Perepechaeva ML, Stefanova NA, Grishanova AY, Kolosova NG. The Expression of Genes CYP1A1, CYP1B1, and CYP2J3 in Distinct Regions of the Heart and Its Possible Contribution to the Development of Hypertension. Biomedicines 2024; 12:2374. [PMID: 39457686 PMCID: PMC11505345 DOI: 10.3390/biomedicines12102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND It is believed that alterations in the functioning of the cytochrome P450 (CYP), which participates in metabolic transformations of endogenous polyunsaturated fatty acids (PUFAs) (with the formation of cardioprotective or cardiotoxic products), affects the development of age-related cardiovascular diseases and reduces the effectiveness of some cardioselective drugs. For example, CYP2J2 activation or CYP1B1 inhibition protects against the cardiovascular toxicity of anticancer drugs. It is currently unclear whether CYPs capable of metabolizing arachidonic acid and ω-3 PUFAs to vasodilatory and vasoconstrictive derivatives are expressed in all heart regions. METHODS The work was performed on senescence-accelerated OXYS rats featuring elevated blood pressure, OXYSb rats (an OXYS substrain with normal blood pressure), and Wistar rats as a "healthy" control. The mRNA level was determined in the right and left ventricles, the right and left atria, and the aorta of 1-, 3-, and 12-month-old rats. RESULTS We showed that all heart regions express CYPs capable of metabolizing arachidonic acid and ω-3 PUFAs and revealed significant differences between heart regions both in the mRNA level of genes CYP1B1, CYP2J3, and CYP1A1 and in the time course of expression changes with age. CONCLUSIONS We noticed that expression levels of these CYPs in the heart regions and aorta differ between hypertensive OXYS rats, normotensive OXYSb rats, and healthy Wistar rats but could not detect any clear-cut patterns associated with the hypertensive status of OXYS rats.
Collapse
Affiliation(s)
- Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| | - Natalia A. Stefanova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia; (N.A.S.); (N.G.K.)
| | - Alevtina Y. Grishanova
- Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| | - Nataliya G. Kolosova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia; (N.A.S.); (N.G.K.)
| |
Collapse
|
6
|
Kucsera D, Ruppert M, Sayour NV, Tóth VE, Kovács T, Hegedűs ZI, Onódi Z, Fábián A, Kovács A, Radovits T, Merkely B, Pacher P, Ferdinandy P, Varga ZV. NASH triggers cardiometabolic HFpEF in aging mice. GeroScience 2024; 46:4517-4531. [PMID: 38630423 PMCID: PMC11336017 DOI: 10.1007/s11357-024-01153-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/20/2024] [Indexed: 08/22/2024] Open
Abstract
Both heart failure with preserved ejection fraction (HFpEF) and non-alcoholic fatty liver disease (NAFLD) develop due to metabolic dysregulation, has similar risk factors (e.g., insulin resistance, systemic inflammation) and are unresolved clinical challenges. Therefore, the potential link between the two disease is important to study. We aimed to evaluate whether NASH is an independent factor of cardiac dysfunction and to investigate the age dependent effects of NASH on cardiac function. C57Bl/6 J middle aged (10 months old) and aged mice (24 months old) were fed either control or choline deficient (CDAA) diet for 8 weeks. Before termination, echocardiography was performed. Upon termination, organ samples were isolated for histological and molecular analysis. CDAA diet led to the development of NASH in both age groups, without inducing weight gain, allowing to study the direct effect of NASH on cardiac function. Mice with NASH developed hepatomegaly, fibrosis, and inflammation. Aged animals had increased heart weight. Conventional echocardiography revealed normal systolic function in all cohorts, while increased left ventricular volumes in aged mice. Two-dimensional speckle tracking echocardiography showed subtle systolic and diastolic deterioration in aged mice with NASH. Histologic analyses of cardiac samples showed increased cross-sectional area, pronounced fibrosis and Col1a1 gene expression, and elevated intracardiac CD68+ macrophage count with increased Il1b expression. Conventional echocardiography failed to reveal subtle change in myocardial function; however, 2D speckle tracking echocardiography was able to identify diastolic deterioration. NASH had greater impact on aged animals resulting in cardiac hypertrophy, fibrosis, and inflammation.
Collapse
Affiliation(s)
- Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Viktória E Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsombor I Hegedűs
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Alexandra Fábián
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Kovács
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Surgical Research and Techniques, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute On Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
7
|
Liu H, Zhang Y, Zhong Z, Gong Y, Yu P, Yang Y, Zhang Y, Zhou T, Chen L. Immunomodulator AS101 restores colistin susceptibility of clinical colistin-resistant Escherichia coli and Klebsiella pneumoniae in vitro and in vivo. Int J Antimicrob Agents 2024; 64:107285. [PMID: 39111708 DOI: 10.1016/j.ijantimicag.2024.107285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES Colistin (COL) was once considered to be the last line of defence against multidrug-resistant bacteria belonging to the family Enterobacteriaceae. Due to the misuse of COL, COL-resistant (COL-R) Enterobacteriaceae have emerged. To address this clinical issue and combat COL resistance, novel approaches are urgently needed. METHODS In this study, the in vitro and in vivo antimicrobial and antibiofilm effects of the immunomodulator AS101 were investigated in combination with COL against COL-R Escherichia coli (E. coli) and Klebsiella pneumoniae (K. pneumoniae). RESULTS Checkerboard assay, time-kill assay, and scanning electron microscopy confirmed the in vitro antimicrobial phenotype, whereas, crystal violet staining and multidimensional confocal laser scanning microscopy with live/dead staining confirmed the antibiofilm capability of the combination therapy. Moreover, the Galleria mellonella infection model and the mouse infection model indicated the high in vivo efficacy of the combination therapy. Additionally, cytotoxicity experiments performed using human kidney-derived HK-2 cells and haemolysis assays performed using human erythrocytes collectively demonstrated safety at effective combination concentrations. Furthermore, quantification of the expression of inflammatory cytokines via enzyme-linked immunosorbent assay confirmed the anti-inflammatory advantage of combination therapy. At the mechanistic level, changes in outer and inner membrane permeability and accumulation of ROS levels, which might be potential mechanisms for synergistic antimicrobial effects. CONCLUSIONS This study found that AS101 can restore COL susceptibility in clinical COL-R E. coli and K. pneumoniae and also has synergistic antibiofilm and anti-inflammatory capabilities. This study provided a novel strategy to combat clinical infections caused by COL-R E. coli and K. pneumoniae.
Collapse
Affiliation(s)
- Haifeng Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Ying Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zeyong Zhong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanchun Gong
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Pingting Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yuhan Yang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yichi Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [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: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
9
|
Zhang J, Liu J, Ye M, Zhang M, Yao F, Cheng Y. Incidence and risk factors associated with atrioventricular block in the general population: the atherosclerosis risk in communities study and Cardiovascular Health Study. BMC Cardiovasc Disord 2024; 24:509. [PMID: 39327574 PMCID: PMC11428875 DOI: 10.1186/s12872-024-04163-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
OBJECTIVES To identify risk factors correlated with atrioventricular block (AVB) in the general population. METHODS Participants in the Atherosclerosis Risk in Communities study (ARIC) and the Cardiovascular Health study (CHS) were enrolled. The presence of AVB was confirmed at an electrocardiogram (ECG) reading center using Minnesota ECG Classification. Cox proportional hazards models were performed to investigate potential risk factors of AVB, after adjustment for age, sex, race and traditional cardiovascular risk factors. RESULTS During the 17 years of follow-up, a total of 731 high-degree AVB cases were identified. Age and sex-standardized rate of AVB was 2.79 and 2.35 per 1000 person-years in the white and the black population, respectively. With the increase of the geriatric population, the incidence of high-degree AVB will increase from 378,816 in 2020 to 535,076 in 2060, and most increment would occur among the elderly. Older age, male sex, the white race, overweight, comorbidities, declined forced vital capacity (FVC), elevated inflammation biomarkers, left bundle branch block and bifascicular block were independently associated with the incidence of high-degree AVB. CONCLUSION To conclude, older age, male sex, white population, overweight, combined diabetes or chronic kidney disease, impaired FVC, elevated inflammation biomarkers, left bundle branch block and bifascicular block were independent predictors for high-degree AVB. The next 40 years would witness a dramatic increase in the incidence of high-degree AVB.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Jia Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Min Ye
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fengjuan Yao
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China.
| | - Yunjiu Cheng
- Department of Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Liu Y, Qiu Z, Shen G, Sun Y, Mei J, Liu Z, Wang L, Li J. Associations between neutrophil-percentage-to-albumin ratio level and all-cause mortality and cardiovascular disease-cause mortality in general population: evidence from NHANES 1999-2010. Front Cardiovasc Med 2024; 11:1393513. [PMID: 39386385 PMCID: PMC11461234 DOI: 10.3389/fcvm.2024.1393513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Chronic inflammation is a recognized independent risk factor for cardiovascular disease (CVD), highlighting the need for reliable inflammatory indicator to predict CVDs. As an inflammatory indicator which has been proved to have predictive value for prognosis of CVDs, neutrophil percentage-to-albumin ratio (NPAR) has obtained increasing attention, but further research is needed to confirm the relationship with mortality in the general population. Method This prospective cohort study included 21,317 individuals who participated in the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2010, where baseline characteristics and NPAR level were extracted. Data for CVD and all-cause mortality were acquired by linking the cohort database with the National Death Index through December 31, 2019. We employed restricted cubic spline analyses to examine the nonlinear association. Weighted Kaplan-Meier curves with log-rank tests were conducted to access cumulative survival differences across different NPAR results. Multivariable Cox proportional hazards regression models were used to compute hazard ratios and 95% CIs. Receiver Operating Characteristic (ROC) curves were used to compare predictive value of NPAR with systemic immune inflammation index (SII) and neutrophils percent. Results In this cohort study, during 270,014 person-years of follow-up, 4,074 all-cause deaths and 1,116 CVD-cause deaths were documented. NPAR levels exhibited significant nonlinear associations with both CVD-cause (P = 0.018 for nonlinearity) and all-cause mortality (P < 0.001 for nonlinearity). Participants in the highest NPAR tertile had a significantly increased risk of all-cause mortality (HR: 1.46, 95% CI: 1.33-1.61) and CVD-cause mortality (HR: 1.54, 95% CI: 1.32-1.80) compared to those in the lowest tertile in the fully adjusted model, while no association was detected for individuals in the middle tertile. Further ROC analysis confirmed that NPAR had higher predictive value than neutrophil percent segment and SII. Conclusions Elevated NPAR level was significantly associated with an increased risk of all-cause and CVD-cause mortality in general population. The high predictive value of NPAR, combined with the easy-to-calculate property, suggests that its potential as a novel inflammatory indicator is worthy of further investigation.
Collapse
Affiliation(s)
- Yuting Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zifeng Qiu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Geng Shen
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - YangYang Sun
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jiarong Mei
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zhihao Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Leyi Wang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| |
Collapse
|
11
|
Uddin MM, Saadatagah S, Niroula A, Yu B, Hornsby WE, Ganesh S, Lannery K, Schuermans A, Honigberg MC, Bick AG, Libby P, Ebert BL, Ballantyne CM, Natarajan P. Long-term longitudinal analysis of 4,187 participants reveals insights into determinants of clonal hematopoiesis. Nat Commun 2024; 15:7858. [PMID: 39251642 PMCID: PMC11385577 DOI: 10.1038/s41467-024-52302-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 09/01/2024] [Indexed: 09/11/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is linked to diverse aging-related diseases, including hematologic malignancy and atherosclerotic cardiovascular disease (ASCVD). While CHIP is common among older adults, the underlying factors driving its development are largely unknown. To address this, we performed whole-exome sequencing on 8,374 blood DNA samples collected from 4,187 Atherosclerosis Risk in Communities Study (ARIC) participants over a median follow-up of 21 years. During this period, 735 participants developed incident CHIP. Splicing factor genes (SF3B1, SRSF2, U2AF1, and ZRSR2) and TET2 CHIP grow significantly faster than DNMT3A non-R882 clones. We find that age at baseline and sex significantly influence the incidence of CHIP, while ASCVD and other traditional ASCVD risk factors do not exhibit such associations. Additionally, baseline synonymous passenger mutations are strongly associated with CHIP status and are predictive of new CHIP clone acquisition and clonal growth over extended follow-up, providing valuable insights into clonal dynamics of aging hematopoietic stem and progenitor cells. This study also reveals associations between germline genetic variants and incident CHIP. Our comprehensive longitudinal assessment yields insights into cell-intrinsic and -extrinsic factors contributing to the development and progression of CHIP clones in older adults.
Collapse
Affiliation(s)
- Md Mesbah Uddin
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Seyedmohammad Saadatagah
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Abhishek Niroula
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Whitney E Hornsby
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Shriienidhie Ganesh
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Kim Lannery
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Art Schuermans
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Michael C Honigberg
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Libby
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Pradeep Natarajan
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Ravikrishnan A, Wijaya I, Png E, Chng KR, Ho EXP, Ng AHQ, Mohamed Naim AN, Gounot JS, Guan SP, Hanqing JL, Guan L, Li C, Koh JY, de Sessions PF, Koh WP, Feng L, Ng TP, Larbi A, Maier AB, Kennedy BK, Nagarajan N. Gut metagenomes of Asian octogenarians reveal metabolic potential expansion and distinct microbial species associated with aging phenotypes. Nat Commun 2024; 15:7751. [PMID: 39237540 PMCID: PMC11377447 DOI: 10.1038/s41467-024-52097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 08/23/2024] [Indexed: 09/07/2024] Open
Abstract
While rapid demographic changes in Asia are driving the incidence of chronic aging-related diseases, the limited availability of high-quality in vivo data hampers our ability to understand complex multi-factorial contributions, including gut microbial, to healthy aging. Leveraging a well-phenotyped cohort of community-living octogenarians in Singapore, we used deep shotgun-metagenomic sequencing for high-resolution taxonomic and functional characterization of their gut microbiomes (n = 234). Joint species-level analysis with other Asian cohorts identified distinct age-associated shifts characterized by reduction in microbial richness, and specific Alistipes and Bacteroides species enrichment (e.g., Alistipes shahii and Bacteroides xylanisolvens). Functional analysis confirmed these changes correspond to metabolic potential expansion in aging towards alternate pathways synthesizing and utilizing amino-acid precursors, vis-à-vis dominant microbial guilds producing butyrate in gut from pyruvate (e.g., Faecalibacterium prausnitzii, Roseburia inulinivorans). Extending these observations to key clinical markers helped identify >10 robust microbial associations to inflammation, cardiometabolic and liver health, including potential probiotic species (e.g., Parabacteroides goldsteinii) and pathobionts (e.g., Klebsiella pneumoniae), highlighting the microbiome's role as biomarkers and potential targets for promoting healthy aging.
Collapse
Affiliation(s)
- Aarthi Ravikrishnan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Indrik Wijaya
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Eileen Png
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Kern Rei Chng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Eliza Xin Pei Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Amanda Hui Qi Ng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Ahmad Nazri Mohamed Naim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Jean-Sebastien Gounot
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Shou Ping Guan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jasinda Lee Hanqing
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Lihuan Guan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Chenhao Li
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Jia Yu Koh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Paola Florez de Sessions
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Woon-Puay Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), 30 Medical Drive, Brenner Centre for Molecular Medicine, Singapore, 117609, Republic of Singapore
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Tze Pin Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Anis Larbi
- Singapore Immunology Network (SigN), Agency for Science Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore
| | - Andrea B Maier
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Brian K Kennedy
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Niranjan Nagarajan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
13
|
Liu C, Liu R, Tian N, Fa W, Liu K, Wang N, Zhu M, Liang X, Ma Y, Ren Y, Wang Y, Cong L, Tang S, Vetrano DL, Ngandu T, Kivipelto M, Hou T, Du Y, Qiu C. Cardiometabolic multimorbidity, peripheral biomarkers, and dementia in rural older adults: The MIND-China study. Alzheimers Dement 2024; 20:6133-6145. [PMID: 38982798 PMCID: PMC11497761 DOI: 10.1002/alz.14091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/20/2024] [Accepted: 06/01/2024] [Indexed: 07/11/2024]
Abstract
INTRODUCTION Evidence has emerged that cardiometabolic multimorbidity (CMM) is associated with dementia, but the underlying mechanisms are poorly understood. METHODS This population-based study included 5704 older adults. Of these, data were available in 1439 persons for plasma amyloid-β (Aβ), total tau, and neurofilament light chain (NfL) and in 1809 persons for serum cytokines. We defined CMM following two common definitions used in previous studies. Data were analyzed using general linear, logistic, and mediation models. RESULTS The presence of CMM was significantly associated with an increased likelihood of dementia, Alzheimer's disease (AD), and vascular dementia (VaD) (p < 0.05). CMM was significantly associated with increased plasma Aβ40, Aβ42, and NfL, whereas CMM that included visceral obesity was associated with increased serum cytokines. The mediation analysis suggested that plasma NfL significantly mediated the association of CMM with AD. DISCUSSION CMM is associated with dementia, AD, and VaD in older adults. The neurodegenerative pathway is involved in the association of CMM with AD. HIGHLIGHTS The presence of CMM was associated with increased likelihoods of dementia, AD, and VaD in older adults. CMM was associated with increased AD-related plasma biomarkers and serum inflammatory cytokines. Neurodegenerative pathway was partly involved in the association of CMM with AD.
Collapse
Affiliation(s)
- Cuicui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Rui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Na Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Wenxin Fa
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
| | - Keke Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Nan Wang
- Department of NeurologyShandong Provincial Hospital, Shandong UniversityJinanShandongP.R. China
| | - Min Zhu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Xiaoyan Liang
- Department of NeurologyShandong Provincial Hospital, Shandong UniversityJinanShandongP.R. China
| | - Yixun Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
| | - Yifei Ren
- Department of NeurologyShandong Provincial Hospital, Shandong UniversityJinanShandongP.R. China
| | - Yongxiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Department of NeurologyShandong Provincial Hospital, Shandong UniversityJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Aging Research Center and Center for Alzheimer Research, Department of NeurobiologyCare Sciences and Society, Karolinska Institutet‐Stockholm UniversitySolnaSweden
| | - Lin Cong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Shi Tang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Davide Liborio Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and SocietyKarolinska Institutet‐Stockholm UniversitySolnaSweden
- Stockholm Gerontology Research CenterStockholmSweden
| | - Tiia Ngandu
- Department of Public Health and WelfareFinnish Institute for Health and WelfareHelsinkiFinland
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of NeurobiologyCare Sciences and Society, Karolinska InstitutetStockholmSweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of NeurobiologyCare Sciences and Society, Karolinska InstitutetStockholmSweden
- Neuroepidemiology and Ageing Research Unit, School of Public HealthImperial College LondonLondonUK
- Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Tingting Hou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Department of NeurologyShandong Provincial Hospital, Shandong UniversityJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | - Chengxuan Qiu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of EducationDepartment of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Aging Research Center and Center for Alzheimer Research, Department of NeurobiologyCare Sciences and Society, Karolinska Institutet‐Stockholm UniversitySolnaSweden
| |
Collapse
|
14
|
Li D, Li X, Zhang X, Chen J, Wang Z, Yu Z, Wu M, Liu L. Geniposide for treating atherosclerotic cardiovascular disease: a systematic review on its biological characteristics, pharmacology, pharmacokinetics, and toxicology. Chin Med 2024; 19:111. [PMID: 39164773 PMCID: PMC11334348 DOI: 10.1186/s13020-024-00981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024] Open
Abstract
In recent years, the prevalence and fatality rates of atherosclerotic cardiovascular disease have not only shown a consistent rise that cannot be ignored, but have also become a pressing social health problem that requires urgent attention. While interventional surgery and drug therapy offer significant therapeutic results, they often come with common side effects. Geniposide, an active component extracted from the Chinese medicine Gardenia jasminoides Ellis, shows promise in the management of cardiac conditions. This review comprehensively outlines the underlying pharmacological mechanisms by which geniposide exerts its effects on atherosclerosis. Geniposide exhibits a range of beneficial effects including alleviating inflammation, inhibiting the development of macrophage foam cells, improving lipid metabolism, and preventing platelet aggregation and thrombosis. It also demonstrates mitochondrial preservation, anti-apoptotic effects, and modulation of autophagy. Moreover, geniposide shows potential in improving oxidative stress and endoplasmic reticulum stress by maintaining the body's antioxidant and oxidative balance. Additionally, this review comprehensively details the biological properties of geniposide, including methods of extraction and purification, as well as its pharmacokinetics and toxicological characteristics. It further discusses the clinical applications of related biopharmaceuticals, emphasizing the potential of geniposide in the prevention and treatment of atherosclerotic cardiovascular diseases. Furthermore, it highlights the limitations of current research, aiming to provide insights for future studies.
Collapse
Affiliation(s)
- Dexiu Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaoya Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaonan Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Jiye Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zeping Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zongliang Yu
- Beijing University of Chinese Medicine, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China.
| |
Collapse
|
15
|
Castro-Varela A, Martinez-Magallanes DM, Reyes-Chavez MF, Gonzalez-Rayas JM, Paredes-Vazquez JG, Vazquez-Garza E, Castillo-Perez M, Flores-Sayavedra YZ, Martinez A, Ramos Cazares RE, Guajardo J, Lopez-de la Garza H, Salinas-Casanova JA, Betancourt H, Molina-Rodriguez AM, Panneflek J, Fabiani MA, Jerjes-Sanchez C. Risk Factors, Clinical Presentation, Therapeutic Trends, and Outcomes in Arterial Thrombosis Complicating Unvaccinated COVID-19 Patients: A Systematic Review. Angiology 2024; 75:625-634. [PMID: 37005343 PMCID: PMC10083125 DOI: 10.1177/00033197231167055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Data on characteristics and outcomes of coronavirus (COVID)-19 patients complicated with arterial thrombosis (AT) are scarce. Therefore, we carried out a systematic review (PRISMA, PROSPERO statements; PubMed, Scopus, and Web of Science) to identify risk factors, clinical presentation, treatment, and outcomes. We included publications from December 2019 to October 2020. Groups: (a) ischemic stroke, (b) thrombotic storm, (c) peripheral vascular thrombosis, (d) myocardial infarction, and (e) left cardiac thrombus or in-transit thrombus (venous system thrombus floating or attaching to the right heart). We considered 131 studies. The most frequent cardiovascular risk factors were: hypertension, diabetes, and dyslipidemia. A high proportion presented with asymptomatic, mild, or moderate COVID-19 (n = 91, 41.4%). We identified a high percentage of isolated ischemic stroke and thrombotic storm. Groups with higher mortality rate: intracardiac thrombus (1/2, 50.0%), thrombotic storm (18/49, 36.7%), and ischemic stroke (48/131, 36.6%). A small number received thromboprophylaxis. Most patients received antithrombotic treatment. The most frequent bleeding complication was intracranial hemorrhage, primarily with isolated stroke. Overall mortality was 33.6% (74/220). Despite a wide range of COVID-19 severity, a high proportion had AT as a complication of non-severe disease. AT can affect different vascular territories; mortality is associated with stroke, intensive care unit stay, and severe COVID-19.
Collapse
Affiliation(s)
- Alejandra Castro-Varela
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Maria Fernanda Reyes-Chavez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | | | - Jose Gildardo Paredes-Vazquez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Eduardo Vazquez-Garza
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Mauricio Castillo-Perez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Arturo Martinez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Ray Erick Ramos Cazares
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Jaime Guajardo
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | - Hector Lopez-de la Garza
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Hector Betancourt
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
| | | | - Jathniel Panneflek
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Mario Alejandro Fabiani
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Carlos Jerjes-Sanchez
- Tecnologico de
Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey,
Nuevo Leon, Mexico
- Unidad Experimental de Terapias
Avanzadas del Hospital Zambrano Hellion, TecSalud, San Pedro Garza Garcia, Nuevo Leon, Mexico
- Instituto de Cardiologia y Medicina
Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud,
Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| |
Collapse
|
16
|
Pośpiech E, Bar A, Pisarek-Pacek A, Karaś A, Branicki W, Chlopicki S. Epigenetic clock in the aorta and age-related endothelial dysfunction in mice. GeroScience 2024; 46:3993-4002. [PMID: 38381284 PMCID: PMC11226569 DOI: 10.1007/s11357-024-01086-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/20/2024] [Indexed: 02/22/2024] Open
Abstract
While epigenetic age (EA) of mouse blood can be determined using DNA methylation analysis at three CpG sites in the Prima1, Hsf4 and Kcns1 genes it is not known whether this approach is useful for predicting vascular biological age. In this study we validated the 3-CpG estimator for age prediction in mouse blood, developed a new predictive model for EA in mouse aorta, and assessed whether epigenetic age acceleration (EAA) measured with blood and aorta samples correlates with age-dependent endothelial dysfunction. Endothelial function was characterized in vivo by MRI in 8-96-week-old C57BL/6 mice. Arterial stiffness was measured by USG-doppler. EA-related changes within 41 CpG sites in Prima1, Kcns1 and Hsf4 loci, were analyzed in the aorta and blood using bisulfite amplicon high-throughput sequencing. Progressive age-dependent endothelial dysfunction and changes in arterial stiffness were observed in 36-96-week-old C57BL/6 mice. Methylation levels of the investigated loci correlated with chronological age in blood and the aorta. The new model for EA estimation in aorta included three cytosines located in the Kcns1 and Hsf4, explained R2 = 87.8% of the variation in age, and predicted age with an mean absolute error of 9.6 weeks in the independent test set. EAA in the aorta was associated with endothelial dysfunction in the abdominal aorta and femoral artery what was consistent with the EAA direction estimated in blood samples. The rate of vascular biological ageing in mice, reflected by the age-dependent systemic endothelial dysfunction, could be estimated using DNA methylation measurements at three loci in aorta and blood samples.
Collapse
Affiliation(s)
- Ewelina Pośpiech
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Al. Powstancow Wielkopolskich 72, 70-204, Szczecin, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Aleksandra Pisarek-Pacek
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
| | - Agnieszka Karaś
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Wojciech Branicki
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland.
- Institute of Forensic Research, Westerplatte 9, 31-033, Kraków, Poland.
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland.
- Faculty of Medicine, Chair of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
17
|
Gil‐Salcedo A, Massart R, de Langavant LC, Bachoud‐Levi A. Modifiable factors associated with Huntington's disease progression in presymptomatic participants. Ann Clin Transl Neurol 2024; 11:1930-1941. [PMID: 38855890 PMCID: PMC11251488 DOI: 10.1002/acn3.52120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/11/2024] Open
Abstract
OBJECTIVE Huntington's disease (HD) is a neurodegenerative disorder characterized by progressive motor, cognitive, and psychiatric symptoms. Our aim here was to identify factors that can be modified to slow disease progression even before the first symptoms appear. METHODS We included 2636 presymptomatic individuals (comparison with family controls) drawn from the prospective observational cohort Enroll-HD, with more than 35 CAG repeats and at least two assessments of disease progression measured with the composite Huntington's disease rating Scale (cUHDRS). The association between sociodemographic factors, health behaviors, health history, and cUHDRS trajectory was assessed with a mixed-effects random forest using partial dependence plots and Shapley additive explanation method. RESULTS Participants were followed by an average of 3.4 (SD = 1.97) years. We confirmed the negative impact of age and a high number of CAG repeats. We found that a high level of education, a body mass index (BMI) <23 kg/m2 before the age of 40 and >23 kg/m2 thereafter, alcohol consumption of <15 units per week, current coffee consumption and no smoking were linked to slow disease progression, as did no previous exposure to antidepressants or anxiolytic, no psychiatric history or comorbidities, and being female. Other comorbidities or marital status showed no major association with HD evolution. INTERPRETATION Reducing modifiable risk factors for HD is one way to support the presymptomatic population. A high level of education, low-to-moderate alcohol consumption, no smoking, and BMI control are likely to slow disease progression in this population.
Collapse
Affiliation(s)
- Andres Gil‐Salcedo
- Département d'Études Cognitives, École Normale SupérieurePSL UniversityParis75005France
- Faculté de MédecineUniversité Paris‐Est CréteilCréteil94000France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Équipe NeuroPsychologie InterventionnelleCréteil94000France
- NeurATRIS, Mondor NodeCréteilFrance
| | - Renaud Massart
- Département d'Études Cognitives, École Normale SupérieurePSL UniversityParis75005France
- Faculté de MédecineUniversité Paris‐Est CréteilCréteil94000France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Équipe NeuroPsychologie InterventionnelleCréteil94000France
- NeurATRIS, Mondor NodeCréteilFrance
| | - Laurent Cleret de Langavant
- Département d'Études Cognitives, École Normale SupérieurePSL UniversityParis75005France
- Faculté de MédecineUniversité Paris‐Est CréteilCréteil94000France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Équipe NeuroPsychologie InterventionnelleCréteil94000France
- NeurATRIS, Mondor NodeCréteilFrance
- APHP, Hôpital Henri Mondor, service de neurologie, centre national de référence maladie de HuntingtonCréteil94000France
| | - Anne‐Catherine Bachoud‐Levi
- Département d'Études Cognitives, École Normale SupérieurePSL UniversityParis75005France
- Faculté de MédecineUniversité Paris‐Est CréteilCréteil94000France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Équipe NeuroPsychologie InterventionnelleCréteil94000France
- NeurATRIS, Mondor NodeCréteilFrance
- APHP, Hôpital Henri Mondor, service de neurologie, centre national de référence maladie de HuntingtonCréteil94000France
| |
Collapse
|
18
|
Müller L, Di Benedetto S. Inflammaging, immunosenescence, and cardiovascular aging: insights into long COVID implications. Front Cardiovasc Med 2024; 11:1384996. [PMID: 38988667 PMCID: PMC11233824 DOI: 10.3389/fcvm.2024.1384996] [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: 02/11/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Aging leads to physiological changes, including inflammaging-a chronic low-grade inflammatory state with significant implications for various physiological systems, particularly for cardiovascular health. Concurrently, immunosenescence-the age-related decline in immune function, exacerbates vulnerabilities to cardiovascular pathologies in older individuals. Examining the dynamic connections between immunosenescence, inflammation, and cardiovascular aging, this mini-review aims to disentangle some of these interactions for a better understanding of their complex interplay. In the context of cardiovascular aging, the chronic inflammatory state associated with inflammaging compromises vascular integrity and function, contributing to atherosclerosis, endothelial dysfunction, arterial stiffening, and hypertension. The aging immune system's decline amplifies oxidative stress, fostering an environment conducive to atherosclerotic plaque formation. Noteworthy inflammatory markers, such as the high-sensitivity C-reactive protein, interleukin-6, interleukin-1β, interleukin-18, and tumor necrosis factor-alpha emerge as key players in cardiovascular aging, triggering inflammatory signaling pathways and intensifying inflammaging and immunosenescence. In this review we aim to explore the molecular and cellular mechanisms underlying inflammaging and immunosenescence, shedding light on their nuanced contributions to cardiovascular diseases. Furthermore, we explore the reciprocal relationship between immunosenescence and inflammaging, revealing a self-reinforcing cycle that intensifies cardiovascular risks. This understanding opens avenues for potential therapeutic targets to break this cycle and mitigate cardiovascular dysfunction in aging individuals. Furthermore, we address the implications of Long COVID, introducing an additional layer of complexity to the relationship between aging, immunosenescence, inflammaging, and cardiovascular health. Our review aims to stimulate continued exploration and advance our understanding within the realm of aging and cardiovascular health.
Collapse
Affiliation(s)
- Ludmila Müller
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | | |
Collapse
|
19
|
Ramoni D, Tirandi A, Montecucco F, Liberale L. Sepsis in elderly patients: the role of neutrophils in pathophysiology and therapy. Intern Emerg Med 2024; 19:901-917. [PMID: 38294676 PMCID: PMC11186952 DOI: 10.1007/s11739-023-03515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Sepsis is among the most important causes of mortality, particularly within the elderly population. Sepsis prevalence is on the rise due to different factors, including increasing average population age and the concomitant rise in the prevalence of frailty and chronic morbidities. Recent investigations have unveiled a "trimodal" trajectory for sepsis-related mortality, with the ultimate zenith occurring from 60 to 90 days until several years after the original insult. This prolonged temporal course ostensibly emanates from the sustained perturbation of immune responses, persevering beyond the phase of clinical convalescence. This phenomenon is particularly associated with the aging immune system, characterized by a broad dysregulation commonly known as "inflammaging." Inflammaging associates with a chronic low-grade activation of the innate immune system preventing an appropriate response to infective agents. Notably, during the initial phases of sepsis, neutrophils-essential in combating pathogens-may exhibit compromised activity. Paradoxically, an overly zealous neutrophilic reaction has been observed to underlie multi-organ dysfunction during the later stages of sepsis. Given this scenario, discovering treatments that can enhance neutrophil activity during the early phases of sepsis while curbing their overactivity in the later phases could prove beneficial in fighting pathogens and reducing the detrimental effects caused by an overactive immune system. This narrative review delves into the potential key role of neutrophils in the pathological process of sepsis, focusing on how the aging process impacts their functions, and highlighting possible targets for developing immune-modulatory therapies. Additionally, the review includes tables that outline the principal potential targets for immunomodulating agents.
Collapse
Affiliation(s)
- Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy.
| |
Collapse
|
20
|
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Virdis A. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries. Hypertension 2024; 81:1218-1232. [PMID: 38511317 DOI: 10.1161/hypertensionaha.123.22483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - George Pavlidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Marcin Hellmann
- Cardiac Diagnostics (M.H.), Medical University of Gdansk, Poland
| | - Stefano Masi
- Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Mariarosaria Lopreiato
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Konstantinos Stavropoulos
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Greece (K. Stavropoulos)
| | - Pawel J Winklewski
- Departments of Human Physiology (P.J.W.), Medical University of Gdansk, Poland
| | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Sarah Costantino
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
| | - Michael Doumas
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - Ignatios Ikonomidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
- Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
- Division of Medicine, Spedali Civili di Brescia, Italy (D.R.)
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim (K. Stellos), Heidelberg University, Germany
- Department of Cardiology, University Hospital Mannheim (K. Stellos), Heidelberg University, Germany
- German Centre for Cardiovascular Research, Heidelberg/Mannheim Partner Site (K. Stellos)
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| |
Collapse
|
21
|
Chen PY, Shen M, Cai SQ, Tang ZW. Association Between Atopic Dermatitis and Aging: Clinical Observations and Underlying Mechanisms. J Inflamm Res 2024; 17:3433-3448. [PMID: 38828054 PMCID: PMC11144009 DOI: 10.2147/jir.s467099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
As one of the most prevalent chronic inflammatory skin diseases, atopic dermatitis (AD) increasingly affects the aging population. Amid the ongoing global aging trend, it's essential to recognize the intricate relationship between AD and aging. This paper reviews existing knowledge, summarizing clinical observations of associations between AD and aging-related diseases in various systems, including endocrine, cardiovascular, and neurological. Additionally, it discusses major theories explaining the correlation, encompassing skin-mucosal barriers, systemic inflammation and stress, genes, signal transduction, and environmental and behavioral factors. The association between AD and aging holds significant importance, both in population and basic perspectives. While further research is warranted, this paper aims to inspire deeper exploration of inflammation/allergy-aging dynamics and the timely management of elderly patients with AD.
Collapse
Affiliation(s)
- Peng-Yu Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease; Hunan Key Laboratory of Skin Cancer and Psoriasis (Xiangya Hospital), Changsha, 410008, People’s Republic of China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, 410078, People’s Republic of China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Zhen-Wei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| |
Collapse
|
22
|
Zhang H, Muhetarijiang M, Chen RJ, Hu X, Han J, Zheng L, Chen T. Mitochondrial Dysfunction: A Roadmap for Understanding and Tackling Cardiovascular Aging. Aging Dis 2024:AD.2024.0058. [PMID: 38739929 DOI: 10.14336/ad.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Cardiovascular aging is a progressive remodeling process constituting a variety of cellular and molecular alterations that are closely linked to mitochondrial dysfunction. Therefore, gaining a deeper understanding of the changes in mitochondrial function during cardiovascular aging is crucial for preventing cardiovascular diseases. Cardiac aging is accompanied by fibrosis, cardiomyocyte hypertrophy, metabolic changes, and infiltration of immune cells, collectively contributing to the overall remodeling of the heart. Similarly, during vascular aging, there is a profound remodeling of blood vessel structure. These remodeling present damage to endothelial cells, increased vascular stiffness, impaired formation of new blood vessels (angiogenesis), the development of arteriosclerosis, and chronic vascular inflammation. This review underscores the role of mitochondrial dysfunction in cardiac aging, exploring its impact on fibrosis and myocardial alterations, metabolic remodeling, immune response remodeling, as well as in vascular aging in the heart. Additionally, we emphasize the significance of mitochondria-targeted therapies in preventing cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mairedan Muhetarijiang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ryan J Chen
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Han
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
23
|
Lapikova-Bryhinska T, Ministrini S, Puspitasari YM, Kraler S, Mohamed SA, Costantino S, Paneni F, Khetsuriani M, Bengs S, Liberale L, Montecucco F, Krampla W, Riederer P, Hinterberger M, Fischer P, Lüscher TF, Grünblatt E, Akhmedov A, Camici GG. Long non-coding RNAs H19 and NKILA are associated with the risk of death and lacunar stroke in the elderly population. Eur J Intern Med 2024; 123:94-101. [PMID: 37981527 DOI: 10.1016/j.ejim.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Differential expression of long non-coding RNAs (lncRNAs) is a hallmark of cardiovascular aging, cerebrovascular diseases, and neurodegenerative disorders. This research article investigates the association between a panel of lncRNAs and the risk of death and ischemic stroke in a cohort of non-institutionalized elderly subjects. METHOD A total of 361 healthy individuals aged 75 years old, prospectively recruited in the Vienna Transdanube Aging (VITA) cohort, were included. Expression of lncRNAs at baseline was assessed using quantitative polymerase chain reaction PCR with pre-amplification reaction, using 18S for normalization. The primary endpoint was all-cause mortality; the secondary endpoint was the incidence of new ischemic brain lesions. Death was assessed over a 14-year follow-up, and ischemic brain lesions were evaluated by magnetic resonance imaging (MRI) over a 90-month follow-up. Ischemic brain lesions were divided into large brain infarcts (Ø≥ 1.5 cm) or lacunes (Ø< 1.5 cm) RESULTS: The primary endpoint occurred in 53.5 % of the study population. The incidence of the secondary endpoint was 16 %, with a 3.3 % being large brain infarcts, and a 12.7 % lacunes. After adjustment for potential confounders, the lncRNA H19 predicted the incidence of the primary endpoint (HR 1.194, 95 % C.I. 1.012-1.409, p = 0.036), whereas the lncRNA NKILA was associated with lacunar stroke (HR 0.571, 95 % C.I. 0.375-0.868, p = 0.006). CONCLUSION In a prospective cohort of non-institutionalized elderly subjects, high levels of lncRNA H19 are associated with a higher risk of death, while low levels of lncRNA NKILA predict an increased risk of lacunar stroke.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Kantonspital Baden, Baden, Switzerland
| | - Shafeeq Ahmed Mohamed
- Center for Translational and Experimental Cardiology, University Hospital of Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital of Zurich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital of Zurich, Zurich, Switzerland; University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Michael Khetsuriani
- Department of General and Molecular Pathophysiology, Bogomolets Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa 16132, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa 16132, Italy
| | | | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany; Department of Psychiatry, University of Southern Denmark Odense, Odense, Denmark
| | - Margareta Hinterberger
- Department of Psychiatry, Medical Research Society Vienna D.C., Danube Hospital Vienna, Vienna, Austria
| | - Peter Fischer
- Department of Psychiatry, Medical Research Society Vienna D.C., Danube Hospital Vienna, Vienna, Austria
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
24
|
Zhao D, Wang Y, Wong ND, Wang J. Impact of Aging on Cardiovascular Diseases: From Chronological Observation to Biological Insights: JACC Family Series. JACC. ASIA 2024; 4:345-358. [PMID: 38765662 PMCID: PMC11099824 DOI: 10.1016/j.jacasi.2024.02.002] [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: 10/02/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Cardiovascular disease (CVD) has increasing challenges for human health with an increasingly aging population worldwide, imposing a significant obstacle to the goal of healthy aging. Rapid advancements in our understanding of biological aging process have shed new light on some important insights to aging-related diseases. Although numerous reviews delved into the mechanisms through which biological aging affects CVD and age-related diseases, most of these reviews relied heavily on research related to cellular and molecular processes often observed from animal experiments. Few reviews have provided insights that connect hypotheses regarding the biological aging process with the observed patterns of chronological aging-related impacts on CVD in human populations. The purpose of this review is to highlight some of the major questions in studies of aging-related CVD and provide our perspectives in the context of real-world patterns of CVD with multidimensional information and potential biological insights.
Collapse
Affiliation(s)
- Dong Zhao
- Capital Medical University Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yibin Wang
- Duke-NUS Medical School and National Heart Center of Singapore, Singapore
| | - Nathan D. Wong
- Division of Cardiology, University of California, Irvine, California, USA
| | - Jian’an Wang
- Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Boulet J, Sridhar VS, Bouabdallaoui N, Tardif JC, White M. Inflammation in heart failure: pathophysiology and therapeutic strategies. Inflamm Res 2024; 73:709-723. [PMID: 38546848 PMCID: PMC11058911 DOI: 10.1007/s00011-023-01845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/09/2023] [Accepted: 12/19/2023] [Indexed: 04/30/2024] Open
Abstract
A role for inflammation in the development and progression of heart failure (HF) has been proposed for decades. Multiple studies have demonstrated the potential involvement of several groups of cytokines and chemokines in acute and chronic HF, though targeting these pathways in early therapeutic trials have produced mixed results. These studies served to highlight the complexity and nuances of how pro-inflammatory pathways contribute to the pathogenesis of HF. More recent investigations have highlighted how inflammation may play distinct roles based on HF syndrome phenotypes, findings that may guide the development of novel therapies. In this review, we propose a contemporary update on the role of inflammation mediated by the innate and adaptive immune systems with HF, highlighting differences that exist across the ejection fraction spectrum. This will specifically be looked at through the lens of established and novel biomarkers of inflammation. Subsequently, we review how improvements in inflammatory pathways may mediate clinical benefits of existing guideline-directed medical therapies for HF, as well as future therapies in the pipeline targeting HF and inflammation.
Collapse
Affiliation(s)
- Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Nadia Bouabdallaoui
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, QC, H1C 1C8, Montreal, Canada.
| |
Collapse
|
26
|
Zhang S, Qiu B, Lv B, Yang G, Tao Y, Hu Y, Li K, Yu X, Tang C, Du J, Jin H, Huang Y. Endogenous sulfur dioxide deficiency as a driver of cardiomyocyte senescence through abolishing sulphenylation of STAT3 at cysteine 259. Redox Biol 2024; 71:103124. [PMID: 38503216 PMCID: PMC10963856 DOI: 10.1016/j.redox.2024.103124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024] Open
Abstract
OBJECTIVE Cardiomyocyte senescence is an important contributor to cardiovascular diseases and can be induced by stressors including DNA damage, oxidative stress, mitochondrial dysfunction, epigenetic regulation, etc. However, the underlying mechanisms for the development of cardiomyocyte senescence remain largely unknown. Sulfur dioxide (SO2) is produced endogenously by aspartate aminotransferase 2 (AAT2) catalysis and plays an important regulatory role in the development of cardiovascular diseases. The present study aimed to explore the effect of endogenous SO2 on cardiomyocyte senescence and the underlying molecular mechanisms. APPROACH AND RESULTS We interestingly found a substantial reduction in the expression of AAT2 in the heart of aged mice in comparison to young mice. AAT2-knockdowned cardiomyocytes exhibited reduced SO2 content, elevated expression levels of Tp53, p21Cip/Waf, and p16INk4a, enhanced SA-β-Gal activity, and elevated level of γ-H2AX foci. Notably, supplementation with a SO2 donor ameliorated the spontaneous senescence phenotype and DNA damage caused by AAT2 deficiency in cardiomyocytes. Mechanistically, AAT2 deficiency suppressed the sulphenylation of signal transducer and activator of transcription 3 (STAT3) facilitated its nuclear translocation and DNA-binding capacity. Conversely, a mutation in the cysteine (Cys) 259 residue of STAT3 blocked SO2-induced STAT3 sulphenylation and subsequently prevented the inhibitory effect of SO2 on STAT3-DNA-binding capacity, DNA damage, and cardiomyocyte senescence. Additionally, cardiomyocyte (cm)-specific AAT2 knockout (AAT2cmKO) mice exhibited a deterioration in cardiac function, cardiomegaly, and cardiac aging, whereas supplementation with SO2 donors mitigated the cardiac aging and remodeling phenotypes in AAT2cmKO mice. CONCLUSION Downregulation of the endogenous SO2/AAT2 pathway is a crucial pathogenic mechanism underlying cardiomyocyte senescence. Endogenous SO2 modifies STAT3 by sulphenylating Cys259, leading to the inhibition of DNA damage and the protection against cardiomyocyte senescence.
Collapse
Affiliation(s)
- Shangyue Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Bingquan Qiu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Boyang Lv
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Guosheng Yang
- Laboratory Animal Facility, Peking University First Hospital, Beijing, 100034, China
| | - Yinghong Tao
- Laboratory Animal Facility, Peking University First Hospital, Beijing, 100034, China
| | - Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing, 100034, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Xiaoqi Yu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, 100191, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
27
|
Lin AE, Bapat AC, Xiao L, Niroula A, Ye J, Wong WJ, Agrawal M, Farady CJ, Boettcher A, Hergott CB, McConkey M, Flores-Bringas P, Shkolnik V, Bick AG, Milan D, Natarajan P, Libby P, Ellinor PT, Ebert BL. Clonal Hematopoiesis of Indeterminate Potential With Loss of Tet2 Enhances Risk for Atrial Fibrillation Through Nlrp3 Inflammasome Activation. Circulation 2024; 149:1419-1434. [PMID: 38357791 PMCID: PMC11058018 DOI: 10.1161/circulationaha.123.065597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1β (interleukin 1β) or IL-6 (interleukin 6). RESULTS Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1β or IL-6. CONCLUSIONS We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.
Collapse
Affiliation(s)
- Amy Erica Lin
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Aneesh C. Bapat
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
| | - Ling Xiao
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Abhishek Niroula
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Department of Laboratory Medicine, Lund University, Sweden (A.N.)
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Sweden (A.N.)
| | - Jiangchuan Ye
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Waihay J. Wong
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
| | - Mridul Agrawal
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Christopher J. Farady
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Christopher B. Hergott
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Marie McConkey
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Patricio Flores-Bringas
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Veronica Shkolnik
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (A.G.B.)
| | - David Milan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Leducq Foundation, Boston, MA (D.M.)
| | - Pradeep Natarajan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Howard Hughes Medical Institute, Boston, MA (B.L.E.)
| |
Collapse
|
28
|
Alonso Salinas GL, Cepas-Guillén P, León AM, Jiménez-Méndez C, Lozano-Vicario L, Martínez-Avial M, Díez-Villanueva P. The Impact of Geriatric Conditions in Elderly Patients with Coronary Heart Disease: A State-of-the-Art Review. J Clin Med 2024; 13:1891. [PMID: 38610656 PMCID: PMC11012545 DOI: 10.3390/jcm13071891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The growing geriatric population presenting with coronary artery disease poses a primary challenge for healthcare services. This is a highly heterogeneous population, often underrepresented in studies and clinical trials, with distinctive characteristics that render them particularly vulnerable to standard management/approaches. In this review, we aim to summarize the available evidence on the treatment of acute coronary syndrome in the elderly. Additionally, we contextualize frailty, comorbidity, sarcopenia, and cognitive impairment, common in these patients, within the realm of coronary artery disease, proposing strategies for each case that may assist in therapeutic approaches.
Collapse
Affiliation(s)
- Gonzalo Luis Alonso Salinas
- Cardiology Department, Hospital Universitario de Navarra (HUN-NOU), Calle de Irunlarrea, 3, 31008 Pamplona, Spain;
- Navarrabiomed (Miguel Servet Foundation), IdiSNA, 31008 Pamplona, Spain;
- Heath Sciences Department, Universidad Pública de Navarra (UPNA-NUP), 31006 Pamplona, Spain
| | - Pedro Cepas-Guillén
- Quebec Heart and Lung Institute, Laval University, 2725 Ch Ste-Foy, Quebec, QC G1V 4G5, Canada;
| | - Amaia Martínez León
- Cardiology Department, Hospital Universitario de Navarra (HUN-NOU), Calle de Irunlarrea, 3, 31008 Pamplona, Spain;
- Navarrabiomed (Miguel Servet Foundation), IdiSNA, 31008 Pamplona, Spain;
| | - César Jiménez-Méndez
- Cardiology Department, Hospital Universitario Puerta del Mar, Avda Ana de Viya 21, 11009 Cádiz, Spain;
| | - Lucia Lozano-Vicario
- Navarrabiomed (Miguel Servet Foundation), IdiSNA, 31008 Pamplona, Spain;
- Geriatric Medicine Department, Hospital Universitario de Navarra (HUN-NOU), Calle de Irunlarrea, 3, 31008 Pamplona, Spain
| | - María Martínez-Avial
- Cardiology Department, Hospital Universitario La Princesa, Calle Diego de León 62, 28006 Madrid, Spain; (M.M.-A.); (P.D.-V.)
| | - Pablo Díez-Villanueva
- Cardiology Department, Hospital Universitario La Princesa, Calle Diego de León 62, 28006 Madrid, Spain; (M.M.-A.); (P.D.-V.)
| |
Collapse
|
29
|
Schlake K, Teller J, Hinken L, Laser H, Lichtinghagen R, Schäfer A, Fegbeutel C, Weissenborn K, Jung C, Worthmann H, Gabriel MM. Butyrylcholinesterase activity in patients with postoperative delirium after cardiothoracic surgery or percutaneous valve replacement- an observational interdisciplinary cohort study. BMC Neurol 2024; 24:80. [PMID: 38424490 PMCID: PMC10905803 DOI: 10.1186/s12883-024-03580-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/23/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Postoperative delirium is a frequent and severe complication after cardiac surgery. Activity of butyrylcholinesterase (BChE) has been discussed controversially regarding a possible role in its development. This study aimed to investigate the relevance of BChE activity as a biomarker for postoperative delirium after cardiac surgery or percutaneous valve replacement. METHODS A total of 237 patients who received elective cardiothoracic surgery or percutaneous valve replacement at a tertiary care centre were admitted preoperatively. These patients were tested with the Montreal Cognitive Assessment investigating cognitive deficits, and assessed for postoperative delirium twice daily for three days via the 3D-CAM or the CAM-ICU, depending on their level of consciousness. BChE activity was measured at three defined time points before and after surgery. RESULTS Postoperative delirium occurred in 39.7% of patients (n = 94). Univariate analysis showed an association of pre- and postoperative BChE activity with its occurrence (p = 0.037, p = 0.001). There was no association of postoperative delirium and the decline in BChE activity (pre- to postoperative, p = 0.327). Multivariable analysis including either preoperative or postoperative BChE activity as well as age, MoCA, type 2 diabetes mellitus, coronary heart disease, type of surgery and intraoperative administration of red-cell concentrates was performed. Neither preoperative nor postoperative BChE activity was independently associated with the occurrence of postoperative delirium (p = 0.086, p = 0.484). Preoperative BChE activity was lower in older patients (B = -12.38 (95% CI: -21.94 to -2.83), p = 0.011), and in those with a history of stroke (B = -516.173 (95% CI: -893.927 to -138.420), p = 0.008) or alcohol abuse (B = -451.47 (95% CI: -868.38 to -34.55), p = 0.034). Lower postoperative BChE activity was independently associated with longer procedures (B = -461.90 (95% CI: -166.34 to -757.46), p = 0.002), use of cardiopulmonary bypass (B = -262.04 (95% CI: -485.68 to -38.39), p = 0.022), the number of administered red cell-concentrates (B = -40.99 (95% CI: -67.86 to -14.12), p = 0.003) and older age (B = -9.35 (95% CI: -16.04 to -2.66), p = 0.006). CONCLUSION BChE activity is not independently associated with the occurrence of postoperative delirium. Preoperative BChE values are related to patients' morbidity and vulnerability, while postoperative activities reflect the severity, length and complications of surgery.
Collapse
Affiliation(s)
- Konstantin Schlake
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Johannes Teller
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Lukas Hinken
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans Laser
- Department for Educational and Scientific IT Systems, Hannover Medical School, MHH Information Technology, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ralf Lichtinghagen
- Institute of Clinical Chemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Andreas Schäfer
- Cardiac Arrest Center, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christine Fegbeutel
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Carolin Jung
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Maria Magdalena Gabriel
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
30
|
Schrock JM. Accelerated aging in people living with HIV: The neuroimmune feedback model. Brain Behav Immun Health 2024; 36:100737. [PMID: 38356933 PMCID: PMC10864877 DOI: 10.1016/j.bbih.2024.100737] [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: 08/18/2023] [Revised: 01/02/2024] [Accepted: 02/04/2024] [Indexed: 02/16/2024] Open
Abstract
People living with HIV (PLWH) experience earlier onset of aging-related comorbidities compared to their counterparts without HIV. This paper lays out a theoretical model to explain why PLWH experience accelerated aging. Briefly, the model is structured as follows. PLWH experience disproportionately heavy burdens of psychosocial stress across the life course. This psychosocial stress increases risks for depressive symptoms and problematic substance use. Depressive symptoms and problematic substance use interfere with long-term adherence to antiretroviral therapy (ART). Lower ART adherence, in turn, exacerbates the elevated systemic inflammation stemming from HIV infection. This inflammation increases risks for aging-related comorbidities. Systemic inflammation also reduces connectivity in the brain's central executive network (CEN), a large-scale brain network that is critical for coping with stressful circumstances. This reduced capacity for coping with stress leads to further increases in depressive symptoms and problematic substance use. Together, these changes form a neuroimmune feedback loop that amplifies the impact of psychosocial stress on aging-related comorbidities. In this paper, I review the existing evidence relevant to this model and highlight directions for future research.
Collapse
Affiliation(s)
- Joshua M. Schrock
- Institute for Sexual and Gender Minority Health and Wellbeing, Northwestern University, 625 N. Michigan Avenue, Suite 1400, Chicago, IL, 60611, United states
| |
Collapse
|
31
|
Tirandi A, Schiavetta E, Maioli E, Montecucco F, Liberale L. Inflammation as a cause of acute myocardial infarction in patients with myeloproliferative neoplasm. World J Cardiol 2024; 16:58-63. [PMID: 38456066 PMCID: PMC10915890 DOI: 10.4330/wjc.v16.i2.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/01/2024] [Accepted: 01/18/2024] [Indexed: 02/21/2024] Open
Abstract
Myeloproliferative neoplasms (MPN) are a group of diseases characterized by the clonal proliferation of hematopoietic progenitor or stem cells. They are clinically classifiable into four main diseases: chronic myeloid leukemia, essential thrombocythemia, polycythemia vera, and primary myelofibrosis. These pathologies are closely related to cardio- and cerebrovascular diseases due to the increased risk of arterial thrombosis, the most common underlying cause of acute myocardial infarction. Recent evidence shows that the classical Virchow triad (hypercoagulability, blood stasis, endothelial injury) might offer an explanation for such association. Indeed, patients with MPN might have a higher number and more reactive circulating platelets and leukocytes, a tendency toward blood stasis because of a high number of circulating red blood cells, endothelial injury or overactivation as a consequence of sustained inflammation caused by the neoplastic clonal cell. These abnormal cancer cells, especially when associated with the JAK2V617F mutation, tend to proliferate and secrete several inflammatory cytokines. This sustains a pro-inflammatory state throughout the body. The direct consequence is the induction of a pro-thrombotic state that acts as a determinant in favoring both venous and arterial thrombus formation. Clinically, MPN patients need to be carefully evaluated to be treated not only with cytoreductive treatments but also with cardiovascular protective strategies.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elisa Schiavetta
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elia Maioli
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy
| |
Collapse
|
32
|
Camacho-Encina M, Booth LK, Redgrave RE, Folaranmi O, Spyridopoulos I, Richardson GD. Cellular Senescence, Mitochondrial Dysfunction, and Their Link to Cardiovascular Disease. Cells 2024; 13:353. [PMID: 38391966 PMCID: PMC10886919 DOI: 10.3390/cells13040353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
Cardiovascular diseases (CVDs), a group of disorders affecting the heart or blood vessels, are the primary cause of death worldwide, with an immense impact on patient quality of life and disability. According to the World Health Organization, CVD takes an estimated 17.9 million lives each year, where more than four out of five CVD deaths are due to heart attacks and strokes. In the decades to come, an increased prevalence of age-related CVD, such as atherosclerosis, coronary artery stenosis, myocardial infarction (MI), valvular heart disease, and heart failure (HF) will contribute to an even greater health and economic burden as the global average life expectancy increases and consequently the world's population continues to age. Considering this, it is important to focus our research efforts on understanding the fundamental mechanisms underlying CVD. In this review, we focus on cellular senescence and mitochondrial dysfunction, which have long been established to contribute to CVD. We also assess the recent advances in targeting mitochondrial dysfunction including energy starvation and oxidative stress, mitochondria dynamics imbalance, cell apoptosis, mitophagy, and senescence with a focus on therapies that influence both and therefore perhaps represent strategies with the most clinical potential, range, and utility.
Collapse
Affiliation(s)
- Maria Camacho-Encina
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Laura K. Booth
- Vascular Medicine and Biology Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (L.K.B.); (I.S.)
| | - Rachael E. Redgrave
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Omowumi Folaranmi
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Ioakim Spyridopoulos
- Vascular Medicine and Biology Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (L.K.B.); (I.S.)
| | - Gavin D. Richardson
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| |
Collapse
|
33
|
Ding W, Chen J, Zhao L, Wu S, Chen X, Chen H. Mitochondrial DNA leakage triggers inflammation in age-related cardiovascular diseases. Front Cell Dev Biol 2024; 12:1287447. [PMID: 38425502 PMCID: PMC10902119 DOI: 10.3389/fcell.2024.1287447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Mitochondrial dysfunction is one of the hallmarks of cardiovascular aging. The leakage of mitochondrial DNA (mtDNA) is increased in senescent cells, which are resistant to programmed cell death such as apoptosis. Due to its similarity to prokaryotic DNA, mtDNA could be recognized by cellular DNA sensors and trigger innate immune responses, resulting in chronic inflammatory conditions during aging. The mechanisms include cGAS-STING signaling, TLR-9 and inflammasomes activation. Mitochondrial quality controls such as mitophagy could prevent mitochondria from triggering harmful inflammatory responses, but when this homeostasis is out of balance, mtDNA-induced inflammation could become pathogenic and contribute to age-related cardiovascular diseases. Here, we summarize recent studies on mechanisms by which mtDNA promotes inflammation and aging-related cardiovascular diseases, and discuss the potential value of mtDNA in early screening and as therapeutic targets.
Collapse
Affiliation(s)
- Wanyue Ding
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Jingyu Chen
- Department of Chinese Medicine Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Zhao
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Shuang Wu
- Southern Medical University Affiliated Qiqihar Hospital, The First Hospital of Qiqihar, Qiqihaer, Heilongjiang, China
| | - Xiaomei Chen
- Integrated Traditional Chinese and Western Medicine Syndrome Laboratory, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hong Chen
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
34
|
Shackebaei D, Hesari M, Ramezani-Aliakbari S, Pashaei M, Yarmohammadi F, Ramezani-Aliakbari F. Cardioprotective effect of naringin against the ischemia/reperfusion injury of aged rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1209-1218. [PMID: 37650890 DOI: 10.1007/s00210-023-02692-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Aging is known as a main risk factor in the development of cardiovascular diseases. Naringin (NRG) is a flavonoid compound derived from citrus fruits. It possesses a wide spectrum of pharmacological properties, including antioxidant anti-inflammatory, and cardioprotective. This investigation aimed to assess the cardioprotective effect of NRG against the ischemia/reperfusion (I/R) injury in aged rats. In this study, D-galactose (D-GAL) at the dose of 150 mg/kg/day for 8 weeks was used to induce aging in rats. Rats were orally gavaged with NRG (40 or 100 mg/kg/day), in co-treatment with D-GAL, for 8 weeks. The Langendorff isolated heart was used to evaluate the effect of NRG on I/R injury in aged rats. NRG treatment diminished myocardial hypertrophy and maximum contracture level in aged animals. During the pre-ischemic phase, reduced heart rate was normalized by NRG. The effects of D-GAL on the left ventricular end diastolic pressure (LVDP), the rate pressure product (RPP), and the minimum and maximum rate of left ventricular pressure (±dp/dt) improved by NRG treatment in the perfusion period. NRG also enhanced post-ischemic recovery of cardiac functional parameters (± dp/dt, and RPP) in isolated hearts. An increase in serum levels of the lactate dehydrogenase (LDH), the creatine kinase-MB (CK-MB), and the tumor necrosis factor-alpha (TNF-α) were reversed by NRG in aged rats. It also normalized the D-GAL-decreased the superoxide dismutase (SOD) activity in the heart tissue. NRG treatment alleviated cardiac injury in aged hearts under conditions of I/R. NRG may improve aging-induced cardiac dysfunction through anti-oxidative and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Cardiovascular Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soudabeh Ramezani-Aliakbari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mosayeb Pashaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Ramezani-Aliakbari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
35
|
Hu M, Zhang X, Gao YP, Hu YX, Teng T, Wang SS, Tang QZ. Isthmin-1 Improves Aging-Related Cardiac Dysfunction in Mice through Enhancing Glycolysis and SIRT1 Deacetylase Activity. Aging Dis 2024:AD.2024.0113. [PMID: 38300636 DOI: 10.14336/ad.2024.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/13/2024] [Indexed: 02/02/2024] Open
Abstract
Aging-related cardiac dysfunction poses a major risk factor of mortality for elderly populations, however, efficient treatment for aging-related cardiac dysfunction is far from being known. Isthmin-1 (ISM1) is a novel adipokine that promotes glucose uptake and acts indispensable roles in restraining inflammatory and fibrosis. The present study aims to investigate the potential role and molecular mechanism of ISM1 in aging-related cardiac dysfunction. Aged and matched young mice were overexpressed or silenced with ISM1 to investigate the role of ISM1 in aging-related cardiac dysfunction. Moreover, H9C2 cells were stimulated with D-galactose (D-gal) to examine the role of ISM1 in vitro. Herein, we found that cardiac-specific overexpression of ISM1 significantly mitigated insulin resistance by promoting glucose uptake in aging mice. ISM1 overexpression alleviated while ISM1 silencing deteriorated cellular senescence, cardiac inflammation, and dysfunction in natural and accelerated cardiac aging. Mechanistically, ISM1 promoted glycolysis and activated Sirtuin-1 (SIRT1) through increasing glucose uptake. ISM1 increased glucose uptake via translocating GLUT4 to the surface, thereby enhancing glycolytic flux and hexosamine biosynthetic pathway (HBP) flux, ultimately leading to increased SIRT1 activity through O-GlcNAc modification. ISM1 may serve as a novel potential therapeutic target for preventing aging-related cardiac disease in elderly populations. ISM1 prevents aging-related cardiac dysfunction by promoting glycolysis and enhancing SIRT1 deacetylase activity, making it a promising therapeutic target for aging-related cardiac disease.
Collapse
Affiliation(s)
- Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xin Zhang
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi-Peng Gao
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Yu-Xin Hu
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Sha-Sha Wang
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
36
|
Maidana D, Arroyo-Álvarez A, Arenas-Loriente A, Barreres-Martín G, Muñoz-Alfonso C, Bompart Berroteran D, Esteve Claramunt F, Blanco del Burgo R, Cepas-Guillén P, Garcia-Blas S, Bonanad C. Inflammation as a New Therapeutic Target among Older Patients with Ischemic Heart Disease. J Clin Med 2024; 13:363. [PMID: 38256497 PMCID: PMC10816645 DOI: 10.3390/jcm13020363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular (CV) diseases remain a global health challenge, with ischemic heart disease (IHD) being the primary cause of both morbidity and mortality. Despite optimal pharmacological therapy, older patients with IHD exhibit an increased susceptibility to recurrent ischemic events, significantly impacting their prognosis. Inflammation is intricately linked with the aging process and plays a pivotal role in the evolution of atherosclerosis. Emerging anti-inflammatory therapies have shown promise in reducing ischemic events among high-risk populations. This review aims to explore the potential of targeted anti-inflammatory interventions in improving clinical outcomes and the quality of life for older patients with IHD.
Collapse
Affiliation(s)
- Daniela Maidana
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Andrea Arroyo-Álvarez
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Guillermo Barreres-Martín
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Carles Muñoz-Alfonso
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Daznia Bompart Berroteran
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Francisca Esteve Claramunt
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Regina Blanco del Burgo
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Sergio Garcia-Blas
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| | - Clara Bonanad
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| |
Collapse
|
37
|
Ye X, Wang Z, Lei W, Shen M, Tang J, Xu X, Yang Y, Zhang H. Pentraxin 3: A promising therapeutic target for cardiovascular diseases. Ageing Res Rev 2024; 93:102163. [PMID: 38092307 DOI: 10.1016/j.arr.2023.102163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023]
Abstract
Cardiovascular disease (CVD) is the primary global cause of death, and inflammation is a crucial factor in the development of CVDs. The acute phase inflammatory protein pentraxin 3 (PTX3) is a biomarker reflecting the immune response. Recent research indicates that PTX3 plays a vital role in CVDs and has been investigated as a possible biomarker for CVD in clinical trials. PTX3 is implicated in the progression of CVDs through mechanisms such as exacerbating vascular endothelial dysfunction, affecting angiogenesis, and regulating inflammation and oxidative stress. This review summarized the structure and function of PTX3, focusing on its multifaceted effects on CVDs, such as atherosclerosis, myocardial infarction, and hypertension. This may help in explaining the varying PTX3 functions and usage, as well as in utilizing target organs to manage diseases. Moreover, elucidating the opposite role of PTX3 in the cardiovascular system will demonstrate the therapeutic and predictive potential in human diseases.
Collapse
Affiliation(s)
- Xingyan Ye
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, China
| | - Wangrui Lei
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital, 80 Jianglin Road, Hainan, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| |
Collapse
|
38
|
Khan R, Di Gesù CM, Lee J, McCullough LD. The contribution of age-related changes in the gut-brain axis to neurological disorders. Gut Microbes 2024; 16:2302801. [PMID: 38237031 PMCID: PMC10798364 DOI: 10.1080/19490976.2024.2302801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Trillions of microbes live symbiotically in the host, specifically in mucosal tissues such as the gut. Recent advances in metagenomics and metabolomics have revealed that the gut microbiota plays a critical role in the regulation of host immunity and metabolism, communicating through bidirectional interactions in the microbiota-gut-brain axis (MGBA). The gut microbiota regulates both gut and systemic immunity and contributes to the neurodevelopment and behaviors of the host. With aging, the composition of the microbiota changes, and emerging studies have linked these shifts in microbial populations to age-related neurological diseases (NDs). Preclinical studies have demonstrated that gut microbiota-targeted therapies can improve behavioral outcomes in the host by modulating microbial, metabolomic, and immunological profiles. In this review, we discuss the pathways of brain-to-gut or gut-to-brain signaling and summarize the role of gut microbiota and microbial metabolites across the lifespan and in disease. We highlight recent studies investigating 1) microbial changes with aging; 2) how aging of the maternal microbiome can affect offspring health; and 3) the contribution of the microbiome to both chronic age-related diseases (e.g., Parkinson's disease, Alzheimer's disease and cerebral amyloidosis), and acute brain injury, including ischemic stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Romeesa Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Claudia M. Di Gesù
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
39
|
Wolf EJ, Miller MW, Hawn SE, Zhao X, Wallander SE, McCormick B, Govan C, Rasmusson A, Stone A, Schichman SA, Logue MW. Longitudinal study of traumatic-stress related cellular and cognitive aging. Brain Behav Immun 2024; 115:494-504. [PMID: 37967663 PMCID: PMC10843744 DOI: 10.1016/j.bbi.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/18/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
Traumatic stress is associated with both accelerated epigenetic age and increased risk for dementia. Accelerated epigenetic age might link symptoms of traumatic stress to dementia-associated biomarkers, such as amyloid-beta (Aβ) proteins, neurofilament light (NFL), and inflammatory molecules. We tested this hypothesis using longitudinal data obtained from 214 trauma-exposed military veterans (85 % male, mean age at baseline: 53 years, 75 % White) who were assessed twice over the course of an average of 5.6 years. Cross-lagged panel mediation models evaluated measures of lifetime posttraumatic stress disorder and internalizing and externalizing comorbidity (assessed at Time 1; T1) in association with T1 epigenetic age (per the GrimAge algorithm) and T1 plasma markers of neuropathology along with bidirectional temporal paths between T1 and T2 epigenetic age and the plasma markers. Results revealed that a measure of externalizing comorbidity was associated with accelerated epigenetic age (β = 0.30, p <.01), which in turn, was associated with subsequent increases in Aβ-40 (β = 0.20, p <.001), Aβ-42 (β = 0.18, p <.001), and interleukin-6 (β = 0.18, p <.01). T1 advanced epigenetic age and the T1 neuropathology biomarkers NFL and glial fibrillary acidic protein predicted worse performance on T2 neurocognitive tasks assessing working memory, executive/attentional control, and/or verbal memory (ps = 0.03 to 0.009). Results suggest that advanced GrimAge is predictive of subsequent increases in neuropathology and inflammatory biomarkers as well as worse cognitive function, highlighting the clinical significance of this biomarker with respect to cognitive aging and brain health over time. The finding that advanced GrimAge mediated the association between psychiatric comorbidity and future neuropathology is important for understanding potential pathways to neurodegeneration and early identification of those at greatest risk.
Collapse
Affiliation(s)
- Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA.
| | - Mark W Miller
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Sage E Hawn
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Old Dominion University, Department of Psychology, Norfolk, VA, USA
| | - Xiang Zhao
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Sara E Wallander
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Beth McCormick
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Christine Govan
- MAVERIC Central Biorepository, VA Boston Healthcare System, Boston, MA, USA
| | - Ann Rasmusson
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Steven A Schichman
- Pathology and Laboratory Medicine Service, Central Arkansas Veterans Healthcare System, USA; Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mark W Logue
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA; Boston University School of Medicine, Department of Medicine, Biomedical Genetics, Boston, MA, USA
| |
Collapse
|
40
|
Lin J, Sheng B, Zhang X. A mediation analysis of metabolic and inflammatory factors in the severe sleep apnea-coronary heart disease association. Postgrad Med J 2023; 100:28-35. [PMID: 37827536 DOI: 10.1093/postmj/qgad096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND It is unclear whether metabolic and inflammatory factors mediate the association between severe sleep apnea and coronary heart disease (CHD) in observational studies based on the large-scale population. This study aims to assess the association between severe sleep apnea and CHD and to explore the extent to which this association is mediated by metabolic factors and C-reactive protein (CRP). METHODS In UK Biobank, 213 442 CHD-free (mean age: 55.00) adults were followed up for 15 years to detect incident CHD. Metabolic factors included hyperglycemia, hypertension, dyslipidemia, hypertriglyceridemia, and hyperuricemia. A higher CRP concentration level was defined as a cutoff point of >3.0 mg/l. Data were analyzed using Cox proportional hazards models and the generalized structural equation model. RESULTS During follow-up, 9278 participants developed incident CHD (4.3%). The multi-adjusted hazard ratio and 95% confidence interval of CHD related to severe sleep apnea were 1.76 (1.44-2.15). In the mediation analysis, the strongest indirect association was for dyslipidemia, accounting for 20.8% of the association between severe sleep apnea and CHD (β = 0.22, 95% confidence interval = 0.16-0.28), followed by hypertriglyceridemia (12.3%). The proportion of mediation increased to 29.1% when CRP was added to the metabolic mediators. CONCLUSIONS Severe sleep apnea was associated with an increased risk of CHD. Lipid factors might play an essential role in the severe sleep apnea-CHD relationship. CRP increased the magnitude of mediation effects of overall metabolic factors. What is already known on this topic It is unknown whether the association between severe sleep apnea and CHD among the large population-based cohort study. Evidence on the mediating effect of metabolic and inflammatory factors in the severe sleep apnea-CHD association remains unclear. What this study adds Lipid factors were the biggest mediation driver in the severe sleep apnea-CHD path. C-reactive protein increased the magnitude of mediation effects of overall metabolic factors. How this study might affect research, practice or policy Investigating mediation analyses not only enhances comprehension of the pathophysiological connection between severe sleep apnea and CHD but also offers valuable insights into preventing CHD.
Collapse
Affiliation(s)
- Jing Lin
- Department of Public Health, Tianjin Medical University, Tianjin 300070, China
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Baihe Sheng
- Department of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Xinyu Zhang
- Department of Public Health, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
41
|
Montecucco F, Schindler TH. Future research with advanced imaging technology and biomarkers. Eur Heart J 2023; 44:5021-5023. [PMID: 37587554 DOI: 10.1093/eurheartj/ehad530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Affiliation(s)
- Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa 16132, Italy
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, 510 S. Kingshighway, Campus Box 8223, St. Louis, MO 63110, USA
| |
Collapse
|
42
|
Kalailingam P, Mohd‐Kahliab K, Ngan SC, Iyappan R, Melekh E, Lu T, Zien GW, Sharma B, Guo T, MacNeil AJ, MacPherson REK, Tsiani EL, O'Leary DD, Lim KL, Su IH, Gao Y, Richards AM, Kalaria RN, Chen CP, McCarthy NE, Sze SK. Immunotherapy targeting isoDGR-protein damage extends lifespan in a mouse model of protein deamidation. EMBO Mol Med 2023; 15:e18526. [PMID: 37971164 PMCID: PMC10701600 DOI: 10.15252/emmm.202318526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/21/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
Aging results from the accumulation of molecular damage that impairs normal biochemical processes. We previously reported that age-linked damage to amino acid sequence NGR (Asn-Gly-Arg) results in "gain-of-function" conformational switching to isoDGR (isoAsp-Gly-Arg). This integrin-binding motif activates leukocytes and promotes chronic inflammation, which are characteristic features of age-linked cardiovascular disorders. We now report that anti-isoDGR immunotherapy mitigates lifespan reduction of Pcmt1-/- mouse. We observed extensive accumulation of isoDGR and inflammatory cytokine expression in multiple tissues from Pcmt1-/- and naturally aged WT animals, which could also be induced via injection of isoDGR-modified plasma proteins or synthetic peptides into young WT animals. However, weekly injection of anti-isoDGR mAb (1 mg/kg) was sufficient to significantly reduce isoDGR-protein levels in body tissues, decreased pro-inflammatory cytokine concentrations in blood plasma, improved cognition/coordination metrics, and extended the average lifespan of Pcmt1-/- mice. Mechanistically, isoDGR-mAb mediated immune clearance of damaged isoDGR-proteins via antibody-dependent cellular phagocytosis (ADCP). These results indicate that immunotherapy targeting age-linked protein damage may represent an effective intervention strategy in a range of human degenerative disorders.
Collapse
Affiliation(s)
| | | | - SoFong Cam Ngan
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Ranjith Iyappan
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Evelin Melekh
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Tian Lu
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life SciencesWestlake UniversityHangzhouChina
| | - Gan Wei Zien
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Bhargy Sharma
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Tiannan Guo
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life SciencesWestlake UniversityHangzhouChina
| | - Adam J MacNeil
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Rebecca EK MacPherson
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Evangelia Litsa Tsiani
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Deborah D O'Leary
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Kah Leong Lim
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - I Hsin Su
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Yong‐Gui Gao
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - A Mark Richards
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Department of CardiologyUniversity of OtagoChristchurchNew Zealand
| | - Raj N Kalaria
- Institute of Neuroscience, Campus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
| | - Christopher P Chen
- Memory, Aging and Cognition CentreNational University Health SystemSingaporeSingapore
| | - Neil E McCarthy
- Centre for Immunobiology, The Blizard Institute, Bart's and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Siu Kwan Sze
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesONCanada
| |
Collapse
|
43
|
Luan Y, Luan Y, Jiao Y, Liu H, Huang Z, Feng Q, Pei J, Yang Y, Ren K. Broadening Horizons: Exploring mtDAMPs as a Mechanism and Potential Intervention Target in Cardiovascular Diseases. Aging Dis 2023:AD.2023.1130. [PMID: 38270118 DOI: 10.14336/ad.2023.1130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Cardiovascular diseases (CVDs) have been recognized as the leading cause of premature mortality and morbidity worldwide despite significant advances in therapeutics. Inflammation is a key factor in CVD progression. Once stress stimulates cells, they release cellular compartments known as damage-associated molecular patterns (DAMPs). Mitochondria can release mitochondrial DAMPs (mtDAMPs) to initiate an immune response when stimulated with cellular stress. Investigating the molecular mechanisms underlying the DAMPs that regulate CVD progression is crucial for improving CVDs. Herein, we discuss the composition and mechanism of DAMPs, the significance of mtDAMPs in cellular inflammation, the presence of mtDAMPs in different types of cells, and the main signaling pathways associated with mtDAMPs. Based on this, we determined the role of DAMPs in CVDs and the effects of mtDAMP intervention on CVD progression. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of DAMPs, this review seeks to provide important theoretical foundations for developing drugs targeting CVDs.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Qi Feng
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People's Hospital, Zhengzhou, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Fumagalli S, Ricciardi G, Di Serio C, La Marca G, Pieraccini G, Franci Montorzi R, Santamaria E, Spanalatte G, Marchetti F, Corti G, Pinton L, Marchionni N. Inflammation, mitochondrial dysfunction and physical performance: a possible association in older patients with persistent atrial fibrillation-the results of a preliminary study. Aging Clin Exp Res 2023; 35:2831-2837. [PMID: 37733227 DOI: 10.1007/s40520-023-02558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with chronic inflammation, a hallmark of ageing process. The aim of this study was to determine interleukin-6 (IL-6)-associated variables, also exploring acylcarnitines, expression of mitochondrial abnormalities. METHODS We evaluated 22 controls and 50 patients with persistent AF. IL-6 and acylcarnitines were measured with ELISA kits and mass spectrometry techniques. RESULTS IL-6 concentration (mean: 3.9 ± 3.1 pg/mL) was lower in controls and increased in AF patients, especially with heart failure. The CHA2DS2-VASc, the MMSE and the SPPB scores were 3.8 ± 1.6, 28 ± 2 and 9.4 ± 2.1. Thirteen acylcanitines correlated with IL-6. At multivariable analysis, IL-6 was directly associated with C4-OH-a short-chain acylcarnitine, fibrinogen and alanine aminotransferase values, and with hyperuricemia. An inverse association existed with calcium concentration and SPPB score. CONCLUSIONS In older AF patients, IL-6 correlated with acylcarnitines and lower physical performance. Alterations in energy production, reduced physical function and inflammation could contribute to frailty development.
Collapse
Affiliation(s)
- Stefano Fumagalli
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy.
| | - Giulia Ricciardi
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Claudia Di Serio
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Giancarlo La Marca
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', Newborn Screening, Clinical Chemistry and Pharmacology Lab, Meyer Children's Hospital and University of Florence, Florence, Italy
| | - Giuseppe Pieraccini
- Department of Health Sciences, CISM Mass Spectrometry Centre, University of Florence, Florence, Italy
| | - Riccardo Franci Montorzi
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Emanuele Santamaria
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Giulia Spanalatte
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Francesca Marchetti
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Ginevra Corti
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Laura Pinton
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Niccolò Marchionni
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
45
|
Komatsu S, Yutani C, Takahashi S, Takewa M, Iwa N, Ohara T, Kodama K. Cholesterol Crystals as the Main Trigger of Interleukin-6 Production through Innate Inflammatory Response in Human Spontaneously Ruptured Aortic Plaques. J Atheroscler Thromb 2023; 30:1715-1726. [PMID: 37081615 PMCID: PMC10627768 DOI: 10.5551/jat.64098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/13/2023] [Indexed: 04/22/2023] Open
Abstract
AIM This study aimed to clarify whether cholesterol crystals (CCs) are the main trigger of innate inflammation in human spontaneously ruptured aortic plaques (SRAPs). METHODS This study included 260 SRAPs collected during nonobstructive general angioscopy (NOGA) from 126 patients with confirmed or suspected coronary artery disease. Interleukin (IL)-6 levels in SRAPs were measured. IL-6 levels in the Valsalva sinus and femoral or brachial arteries were measured. IL-6 ratios (the IL-6 level in SRAPs and arteries divided by the IL-6 level at the Valsalva sinus at the beginning of the aorta) were calculated. Quantitative analysis of CCs was performed from SRAPs. The correlation between the count of CCs and IL-6 levels in SRAPs and that between the counts of CCs and IL-6 ratios in SRAPs were analyzed. RESULTS The IL-6 levels in SRAPs were 3.4 [2.1, 7.2] pg/mL, and the IL-6 ratio (median [interquartile range]) in SRAPs was 1.10 [1.00, 1.26]. CCs were detected in 94 of 260 SRAPs (36%). The count of CCs was 11,590 (95% confidence interval, 2,386-30,113) per 10 mL in CC-positive samples. There was a moderate correlation between the counts of CCs and IL-6 ratios in SRAPs (r=0.49, r<0.0001), whereas there was no correlation between the count of CCs and IL-6 levels in SRAPs. The IL-6 ratios of the brachial and femoral arteries were 1.06 (95% CI, 0.99-1.20) and 1.11 (95% CI, 1.04-1.20), respectively. CONCLUSIONS CC is the main trigger of IL-6 production through innate inflammatory response in human SRAPs in situ.
Collapse
Affiliation(s)
- Sei Komatsu
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Chikao Yutani
- Division of Pathology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
- Department of Medical Technology, Morinomiya University of Medical Sciences, Osaka, Japan
| | - Satoru Takahashi
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Mitsuhiko Takewa
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Nobuzo Iwa
- Division of Pathology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Tomoki Ohara
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Kazuhisa Kodama
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| |
Collapse
|
46
|
Preda A, Carbone F, Tirandi A, Montecucco F, Liberale L. Obesity phenotypes and cardiovascular risk: From pathophysiology to clinical management. Rev Endocr Metab Disord 2023; 24:901-919. [PMID: 37358728 PMCID: PMC10492705 DOI: 10.1007/s11154-023-09813-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/27/2023]
Abstract
Obesity epidemic reached the dimensions of a real global health crisis with more than one billion people worldwide living with obesity. Multiple obesity-related mechanisms cause structural, functional, humoral, and hemodynamic alterations with cardiovascular (CV) deleterious effects. A correct assessment of the cardiovascular risk in people with obesity is critical for reducing mortality and preserving quality of life. The correct identification of the obesity status remains difficult as recent evidence suggest that different phenotypes of obesity exist, each one associated with different degrees of CV risk. Diagnosis of obesity cannot depend only on anthropometric parameters but should include a precise assessment of the metabolic status. Recently, the World Heart Federation and World Obesity Federation provided an action plan for management of obesity-related CV risk and mortality, stressing for the instauration of comprehensive structured programs encompassing multidisciplinary teams. In this review we aim at providing an updated summary regarding the different obesity phenotypes, their specific effects on CV risk and differences in clinical management.
Collapse
Affiliation(s)
| | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Amedeo Tirandi
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy.
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy.
| | - Luca Liberale
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| |
Collapse
|
47
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
48
|
Vasunilashorn SM, Dillon ST, Marcantonio ER, Libermann TA. Application of Multiple Omics to Understand Postoperative Delirium Pathophysiology in Humans. Gerontology 2023; 69:1369-1384. [PMID: 37722373 PMCID: PMC10711777 DOI: 10.1159/000533789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/23/2023] [Indexed: 09/20/2023] Open
Abstract
Delirium, an acute change in cognition, is common, morbid, and costly, particularly among hospitalized older adults. Despite growing knowledge of its epidemiology, far less is known about delirium pathophysiology. Initial work understanding delirium pathogenesis has focused on assaying single or a limited subset of molecules or genetic loci. Recent technological advances at the forefront of biomarker and drug target discovery have facilitated application of multiple "omics" approaches aimed to provide a more complete understanding of complex disease processes such as delirium. At its basic level, "omics" involves comparison of genes (genomics, epigenomics), transcripts (transcriptomics), proteins (proteomics), metabolites (metabolomics), or lipids (lipidomics) in biological fluids or tissues obtained from patients who have a certain condition (i.e., delirium) and those who do not. Multi-omics analyses of these various types of molecules combined with machine learning and systems biology enable the discovery of biomarkers, biological pathways, and predictors of delirium, thus elucidating its pathophysiology. This review provides an overview of the most recent omics techniques, their current impact on identifying delirium biomarkers, and future potential in enhancing our understanding of delirium pathogenesis. We summarize challenges in identification of specific biomarkers of delirium and, more importantly, in discovering the mechanisms underlying delirium pathophysiology. Based on mounting evidence, we highlight a heightened inflammatory response as one common pathway in delirium risk and progression, and we suggest other promising biological mechanisms that have recently emerged. Advanced multiple omics approaches coupled with bioinformatics methodologies have great promise to yield important discoveries that will advance delirium research.
Collapse
Affiliation(s)
- Sarinnapha M. Vasunilashorn
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Simon T. Dillon
- Harvard Medical School, Boston, MA, USA
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, BIDMC, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, BIDMC, Boston, MA, USA
| | - Edward R. Marcantonio
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Gerontology, Department of Medicine, BIDMC, Boston, MA, USA
| | - Towia A. Libermann
- Harvard Medical School, Boston, MA, USA
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, BIDMC, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, BIDMC, Boston, MA, USA
| |
Collapse
|
49
|
Uddin MM, Saadatagah S, Niroula A, Yu B, Hornsby W, Ganesh S, Lannery K, Shuermans A, Honigberg MC, Bick AG, Libby P, Ebert BL, Ballantyne CM, Natarajan P. Long-term longitudinal analysis of 4,187 participants reveals new insights into determinants of incident clonal hematopoiesis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.05.23295093. [PMID: 37732181 PMCID: PMC10508802 DOI: 10.1101/2023.09.05.23295093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Clonal hematopoiesis (CH), characterized by blood cells predominantly originating from a single mutated hematopoietic stem cell, is linked to diverse aging-related diseases, including hematologic malignancy and atherosclerotic cardiovascular disease (ASCVD). While CH is common among older adults, the underlying factors driving its development are largely unknown. To address this, we performed whole-exome sequencing on 8,374 blood DNA samples collected from 4,187 Atherosclerosis Risk in Communities Study (ARIC) participants over a median follow-up of 21 years. During this period, 735 participants developed incident CH. We found that age at baseline, sex, and dyslipidemia significantly influence the incidence of CH, while ASCVD and other traditional risk factors for ASCVD did not exhibit such associations. Our study also revealed associations between germline genetic variants and incident CH, prioritizing genes in CH development. Our comprehensive longitudinal assessment yields novel insights into the factors contributing to incident CH in older adults.
Collapse
Affiliation(s)
- Md Mesbah Uddin
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Seyedmohammad Saadatagah
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Abhishek Niroula
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Whitney Hornsby
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Shriienidhie Ganesh
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Kim Lannery
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Art Shuermans
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Michael C. Honigberg
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Libby
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Benjamin L. Ebert
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Pradeep Natarajan
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Lai Z, Liu Y, Huang M, Li L, Li Z, Su J, Pan G, Li B, Gao S, Yu C. Associations Between Atherosclerosis and Elevated Serum Alkaline Phosphatase in Patients With Coronary Artery Disease in an Inflammatory State. Heart Lung Circ 2023; 32:1096-1106. [PMID: 37550157 DOI: 10.1016/j.hlc.2023.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND AND AIM Serum alkaline phosphatase (ALP) has been shown to be associated with cardiovascular disease (CVD) risk. Inflammation is the initiator of atherosclerosis, throughout the life of atherosclerosis. This study investigated the relationship between serum ALP and atherosclerosis in patients with coronary artery disease (CAD) in an inflammatory state. METHODS This was a multicentre retrospective study including 22,989 patients with CAD. Serum alkaline phosphatase was converted into the quartiles. C-reactive protein (CRP) was assayed as a marker of systemic inflammation. The atherosclerosis index (AI) was used to assess the degree of atherosclerosis. Binary logistic regression was used to analyse the relationship between ALP and AI. Stratified analysis was performed according to sex and age. RESULTS Elevated serum ALP was associated with the risk of atherosclerosis in patients with CAD, and after quartiling ALP, the OR for Q4 was 1.17 (95% CI 1.08-1.26; p<0.001) when using Q1 as reference. The odds ratio (OR) for ALP and risk of atherosclerosis was higher in patients aged ≤60 years (OR 1.33, 95% CI 1.15-1.53; p<0.001) than in patients aged >60 years (OR 1.11, 95% CI 1.01-1.23; p<0.05), and higher in males (OR 1.21, 95% CI 1.09-1.35; p<0.001) than in females (OR 1.16, 95% CI 1.03-1.31; p<0.05). Q4 (ALP >83.00 U/L) was significantly associated with increased risk of atherosclerosis in the inflammatory state (OR 1.48, 95% CI 1.18-1.86; p<0.001), and it remained after stratified analysis according to sex and age. CONCLUSIONS The risk of atherosclerosis tended to increase with increasing ALP levels and the correlation between ALP and the degree of atherosclerosis was significantly stronger when ALP was >83.00 U/L. This relationship was more pronounced in inflammatory states, and there were sex and age differences. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04026724.
Collapse
Affiliation(s)
- Ziqin Lai
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yijia Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengnan Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinyu Su
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guangwei Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|