Inflammation, Aging, and Cardiovascular Disease: JACC Review Topic of the Week

Luca Liberale, Lina Badimon, Fabrizio Montecucco, Thomas F Lüscher, Peter Libby, Giovanni G Camici, Luca Liberale, Lina Badimon, Fabrizio Montecucco, Thomas F Lüscher, Peter Libby, Giovanni G Camici

Abstract

Aging and inflammation both contribute pivotally to cardiovascular (CV) and cerebrovascular disease, the leading causes of death and disability worldwide. The concept of inflamm-aging recognizes that low-grade inflammatory pathways observed in the elderly contribute to CV risk. Understanding the mechanisms that link inflammation and aging could reveal new therapeutic targets and offer options to cope with the growing aging population worldwide. This review reports recent scientific advances in the pathways through which inflamm-aging mediates age-dependent decline in CV function and disease onset and considers critically the translational potential of such concepts into everyday clinical practice.

Keywords: cardiovascular disease; inflamm-aging; inflammation; vascular aging.

Conflict of interest statement

Funding Support and Author Disclosures Dr Camici has received support for this work from the Swiss National Science Foundation (310030_175546), the Swiss Heart Foundation, Alfred and Annemarie von Sick Grants for Translational and Clinical Research Cardiology and Oncology, and the Foundation for Cardiovascular Research–Zurich Heart House; and is the recipient of a Sheikh Khalifa's Foundation Assistant Professorship at the Faculty of Medicine, University of Zurich. Dr Liberale has received support from the Swiss Heart Foundation and the Novartis Foundation for Medical-Biological Research. Dr Libby has received funding support from the National Heart, Lung, and Blood Institute (1R01HL134892), the American Heart Association (18CSA34080399), the RRM Charitable Fund, and the Simard Fund. Dr Montecucco has received support from the “Rete Cardiologica” of Italian Ministry of Health (#2754291). Drs Liberale, Libby, and Camici are coinventors on the International Patent WO/2020/226993 filed in April 2020; the patent relates to the use of antibodies which specifically bind interleukin-1α to reduce various sequelae of ischemia-reperfusion injury to the central nervous system. Dr Liberale has received speaker fees outside of this work from Daichi-Sankyo. Dr Badimon is founder of 2 spin-offs (Glycardial Diagnostics and Ivestatin Therapeutics); has several patents; has received consultancy fees from Sanofi and Novartis; and has received speaker fees from Lilly, Pfizer, and AstraZeneca (all unrelated to this work). Dr Lüscher has received educational and research grants outside this work from Abbott, Amgen, AstraZeneca, Boehringer Ingelheim, Daichi-Sankyo, Novartis, Servier, Sanofi, and Vifor; and has received speaker fees from Amgen and Daiichi-Sankyo. Dr Libby has served as an unpaid consultant to or been involved in clinical trials for Amgen, AstraZeneca, Baim Institute, Beren Therapeutics, Esperion Therapeutics, Genentech, Kancera, Kowa Pharmaceuticals, Medimmune, Merck, Norvo Nordisk, Novartis, Pfizer, and Sanofi-Regeneron; has served as a member of scientific advisory board for Amgen, Caristo, Cartesian, Corvidia Therapeutics, CSL Behring, DalCor Pharmaceuticals, Dewpoint, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Novartis, PlaqueTec, and XBiotech; his laboratory has received research funding in the last 2 years from Novartis; has served on the Board of Directors of XBiotech, Inc; has a financial interest in XBiotech, a company developing therapeutic human antibodies; and his interests were reviewed and are managed by Brigham and Women’s Hospital and Partners HealthCare in accordance with their conflict-of-interest policies. Dr Montecucco has reported that he has no relationships relevant to the contents of this paper to disclose.

Copyright © 2022 American College of Cardiology Foundation. All rights reserved.

Figures

Figure 1.. Immunosenescence and inflamm-aging.
Figure 1.. Immunosenescence and inflamm-aging.
“Immunosenescence” describes the effect of aging on immune cell distribution and characteristics and sustains inflamm-aging.
Figure 2.. Garbaging.
Figure 2.. Garbaging.
Misplaced and misfolded proteins accumulate with age and are recognized as damage-associated molecular patterns by common receptors eventually fueling inflamm-aging with implications for age-related cardiovascular and cerebrovascular conditions.
Central illustration:. Targeting inflamm-aging to cope with…
Central illustration:. Targeting inflamm-aging to cope with age-related cardio- and cerebrovascular diseases.
Different interventions targeting inflamm-aging-related pathways are now being tested in the context of age-related conditions. CV: cardiovascular; CBV: cerebrovascular.

References

    1. Liberale L, Montecucco F, Schwarz L, Luscher TF, Camici GG. Inflammation and cardiovascular diseases: lessons from seminal clinical trials. Cardiovasc Res 2021;117:411–422.
    1. Libby P Inflammation in Atherosclerosis-No Longer a Theory. Clin Chem 2021;67:131–142.
    1. Poulain M, Pes GM, Grasland C et al. Identification of a geographic area characterized by extreme longevity in the Sardinia island: the AKEA study. Exp Gerontol 2004;39:1423–9.
    1. Frasca D, Blomberg BB. Inflammaging decreases adaptive and innate immune responses in mice and humans. Biogerontology 2016;17:7–19.
    1. Alpert A, Pickman Y, Leipold M et al. A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat Med 2019;25:487–495.
    1. Franceschi C, Capri M, Monti D et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev 2007;128:92–105.
    1. Morrisette-Thomas V, Cohen AA, Fulop T et al. Inflamm-aging does not simply reflect increases in pro-inflammatory markers. Mech Ageing Dev 2014;139:49–57.
    1. Cubedo J, Padro T, Formiga F et al. Inflammation and hemostasis in older octogenarians: implication in 5-year survival. Transl Res 2017;185:34–46 e9.
    1. Sayed N, Huang X, Nguyen K et al. An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging. Nat Aging 2021;1:598–615.
    1. Chiva-Blanch G, Vilella-Figuerola A, Padro T, Formiga F, Ferrer A, Badimon L. Functional and Cognitive Decline Is Associated With Increased Endothelial Cell Inflammation and Platelet Activation: Liquid Biopsy of Microvesicles in Community-Dwelling Octogenarians. Front Cell Dev Biol 2021;9:716435.
    1. Liberale L, Kraler S, Camici GG, Luscher TF. Ageing and longevity genes in cardiovascular diseases. Basic Clin Pharmacol Toxicol 2020;127:120–131.
    1. Walaszczyk A, Dookun E, Redgrave R et al. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019;18:e12945.
    1. Correia-Melo C, Birch J, Fielder E et al. Rapamycin improves healthspan but not inflammaging in nfkappab1(−/−) mice. Aging Cell 2019;18:e12882.
    1. Pawelec G Immune signatures associated with mortality differ in elderly populations from different birth cohorts and countries even within northern Europe. Mech Ageing Dev 2019;177:182–185.
    1. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016;15:428–35.
    1. Maier R, Bawamia B, Bennaceur K et al. Telomerase Activation to Reverse Immunosenescence in Elderly Patients With Acute Coronary Syndrome: Protocol for a Randomized Pilot Trial. JMIR Res Protoc 2020;9:e19456.
    1. Franceschi C, Garagnani P, Vitale G, Capri M, Salvioli S. Inflammaging and ‘Garbaging’. Trends Endocrinol Metab 2017;28:199–212.
    1. Diteepeng T, Del Monte F, Luciani M. The long and winding road to target protein misfolding in cardiovascular diseases. Eur J Clin Invest 2021;51:e13504.
    1. Lynn EG, Lhotak S, Lebeau P et al. 4-Phenylbutyrate protects against atherosclerotic lesion growth by increasing the expression of HSP25 in macrophages and in the circulation of Apoe(−/−) mice. FASEB J 2019;33:8406–8422.
    1. Ye S, Luo W, Khan ZA et al. Celastrol Attenuates Angiotensin II-Induced Cardiac Remodeling by Targeting STAT3. Circ Res 2020;126:1007–1023.
    1. Quarles E, Basisty N, Chiao YA et al. Rapamycin persistently improves cardiac function in aged, male and female mice, even following cessation of treatment. Aging Cell 2020;19:e13086.
    1. Wang N, Li R, Lin H et al. Enriched taxa were found among the gut microbiota of centenarians in East China. PLoS One 2019;14:e0222763.
    1. Biagi E, Franceschi C, Rampelli S et al. Gut Microbiota and Extreme Longevity. Curr Biol 2016;26:1480–5.
    1. Sovran B, Hugenholtz F, Elderman M et al. Age-associated Impairment of the Mucus Barrier Function is Associated with Profound Changes in Microbiota and Immunity. Sci Rep 2019;9:1437.
    1. Liberale L, Holy EW, Akhmedov A et al. Interleukin-1beta Mediates Arterial Thrombus Formation via NET-Associated Tissue Factor. J Clin Med 2019;8.
    1. Bonetti NR, Liberale L, Akhmedov A et al. Long-term dietary supplementation with plant-derived omega-3 fatty acid improves outcome in experimental ischemic stroke. Atherosclerosis 2021;325:89–98.
    1. Saeedi Saravi SS, Bonetti NR, Pugin B et al. Lifelong dietary omega-3 fatty acid suppresses thrombotic potential through gut microbiota alteration in aged mice. iScience 2021;24:102897.
    1. Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020;40:1135–1147.
    1. Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 2018;14:576–590.
    1. Arai Y, Kamide K, Hirose N. Adipokines and Aging: Findings From Centenarians and the Very Old. Front Endocrinol (Lausanne) 2019;10:142.
    1. Bik W, Baranowska-Bik A, Wolinska-Witort E et al. Assessment of adiponectin and its isoforms in Polish centenarians. Exp Gerontol 2013;48:401–7.
    1. Van Berendoncks AM, Garnier A, Beckers P et al. Functional adiponectin resistance at the level of the skeletal muscle in mild to moderate chronic heart failure. Circ Heart Fail 2010;3:185–94.
    1. Xu XM, Ning YC, Wang WJ et al. Anti-Inflamm-Aging Effects of Long-Term Caloric Restriction via Overexpression of SIGIRR to Inhibit NF-kappaB Signaling Pathway. Cell Physiol Biochem 2015;37:1257–70.
    1. Martucci M, Ostan R, Biondi F et al. Mediterranean diet and inflammaging within the hormesis paradigm. Nutr Rev 2017;75:442–455.
    1. Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J 2020;41:2974–2982.
    1. Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 2020;17:137–144.
    1. Jaiswal S, Natarajan P, Silver AJ et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med 2017;377:111–121.
    1. Dorsheimer L, Assmus B, Rasper T et al. Association of Mutations Contributing to Clonal Hematopoiesis With Prognosis in Chronic Ischemic Heart Failure. JAMA Cardiol 2019;4:25–33.
    1. Pascual-Figal DA, Bayes-Genis A, Diez-Diez M et al. Clonal Hematopoiesis and Risk of Progression of Heart Failure With Reduced Left Ventricular Ejection Fraction. J Am Coll Cardiol 2021;77:1747–1759.
    1. Abplanalp WT, Mas-Peiro S, Cremer S, John D, Dimmeler S, Zeiher AM. Association of Clonal Hematopoiesis of Indeterminate Potential With Inflammatory Gene Expression in Patients With Severe Degenerative Aortic Valve Stenosis or Chronic Postischemic Heart Failure. JAMA Cardiol 2020;5:1170–1175.
    1. Liberale L, Ministrini S, Carbone F, Camici GG, Montecucco F. Cytokines as therapeutic targets for cardio- and cerebrovascular diseases. Basic Res Cardiol 2021;116:23.
    1. Abplanalp WT, Cremer S, John D et al. Clonal Hematopoiesis-Driver DNMT3A Mutations Alter Immune Cells in Heart Failure. Circ Res 2021;128:216–228.
    1. Wolach O, Sellar RS, Martinod K et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med 2018;10.
    1. Rauch PJ, Silver AJ, Gopakumar J et al. Loss-of-Function Mutations in Dnmt3a and Tet2 Lead to Accelerated Atherosclerosis and Convergent Macrophage Phenotypes in Mice. Blood 2018;132.
    1. Sano S, Oshima K, Wang Y, Katanasaka Y, Sano M, Walsh K. CRISPR-Mediated Gene Editing to Assess the Roles of Tet2 and Dnmt3a in Clonal Hematopoiesis and Cardiovascular Disease. Circ Res 2018;123:335–341.
    1. Fuster JJ, MacLauchlan S, Zuriaga MA et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017;355:842–847.
    1. Higa KC, Goodspeed A, Chavez JS et al. Chronic interleukin-1 exposure triggers selection for Cebpa-knockout multipotent hematopoietic progenitors. J Exp Med 2021;218.
    1. Zeng Y, Nie C, Min J et al. Novel loci and pathways significantly associated with longevity. Sci Rep 2016;6:21243.
    1. Bonafe M, Olivieri F, Cavallone L et al. A gender--dependent genetic predisposition to produce high levels of IL-6 is detrimental for longevity. Eur J Immunol 2001;31:2357–61.
    1. Varadhan R, Yao W, Matteini A et al. Simple biologically informed inflammatory index of two serum cytokines predicts 10 year all-cause mortality in older adults. J Gerontol A Biol Sci Med Sci 2014;69:165–73.
    1. Georgakis MK, Malik R, Gill D et al. Interleukin-6 Signaling Effects on Ischemic Stroke and Other Cardiovascular Outcomes: A Mendelian Randomization Study. Circ Genom Precis Med 2020;13:e002872.
    1. Theriault S, Dina C, Messika-Zeitoun D et al. Genetic Association Analyses Highlight IL6, ALPL, and NAV1 As 3 New Susceptibility Genes Underlying Calcific Aortic Valve Stenosis. Circ Genom Precis Med 2019;12:e002617.
    1. Agrawal A, Tay J, Yang GE, Agrawal S, Gupta S. Age-associated epigenetic modifications in human DNA increase its immunogenicity. Aging (Albany NY) 2010;2:93–100.
    1. Jylhava J, Nevalainen T, Marttila S, Jylha M, Hervonen A, Hurme M. Characterization of the role of distinct plasma cell-free DNA species in age-associated inflammation and frailty. Aging Cell 2013;12:388–97.
    1. Bacalini MG, D’Aquila P, Marasco E et al. The methylation of nuclear and mitochondrial DNA in ageing phenotypes and longevity. Mech Ageing Dev 2017;165:156–161.
    1. Nachun D, Lu AT, Bick AG et al. Clonal hematopoiesis associated with epigenetic aging and clinical outcomes. Aging Cell 2021;20:e13366.
    1. Nicolia V, Cavallaro RA, Lopez-Gonzalez I et al. DNA Methylation Profiles of Selected Pro-Inflammatory Cytokines in Alzheimer Disease. J Neuropathol Exp Neurol 2017;76:27–31.
    1. Chen BH, Marioni RE, Colicino E et al. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging (Albany NY) 2016;8:1844–1865.
    1. Bergsma T, Rogaeva E. DNA Methylation Clocks and Their Predictive Capacity for Aging Phenotypes and Healthspan. Neurosci Insights 2020;15:2633105520942221.
    1. Wasiak S, Gilham D, Tsujikawa LM et al. Downregulation of the Complement Cascade In Vitro, in Mice and in Patients with Cardiovascular Disease by the BET Protein Inhibitor Apabetalone (RVX-208). J Cardiovasc Transl Res 2017;10:337–347.
    1. Tsujikawa LM, Fu L, Das S et al. Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenetics 2019;11:102.
    1. Ray KK, Nicholls SJ, Buhr KA et al. Effect of Apabetalone Added to Standard Therapy on Major Adverse Cardiovascular Events in Patients With Recent Acute Coronary Syndrome and Type 2 Diabetes: A Randomized Clinical Trial. JAMA 2020;323:1565–1573.
    1. Fahy GM, Brooke RT, Watson JP et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 2019;18:e13028.
    1. Jylhava J, Pedersen NL, Hagg S. Biological Age Predictors. EBioMedicine 2017;21:29–36.
    1. Vogel B, Acevedo M, Appelman Y et al. The Lancet women and cardiovascular disease Commission: reducing the global burden by 2030. Lancet 2021;397:2385–2438.
    1. Yousefzadeh MJ, Zhao J, Bukata C et al. Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice. Aging Cell 2020;19:e13094.
    1. Dulken B, Brunet A. Stem Cell Aging and Sex: Are We Missing Something? Cell Stem Cell 2015;16:588–90.
    1. Jenkins EC, Shah N, Gomez M et al. Proteasome mapping reveals sexual dimorphism in tissue-specific sensitivity to protein aggregations. EMBO Rep 2020;21:e48978.
    1. Pomatto LCD, Wong S, Carney C, Shen B, Tower J, Davies KJA. The age- and sex-specific decline of the 20s proteasome and the Nrf2/CncC signal transduction pathway in adaption and resistance to oxidative stress in Drosophila melanogaster. Aging (Albany NY) 2017;9:1153–1185.
    1. Horvath S, Gurven M, Levine ME et al. An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol 2016;17:171.
    1. Li X, Ploner A, Wang Y et al. Longitudinal trajectories, correlations and mortality associations of nine biological ages across 20-years follow-up. Elife 2020;9.
    1. Yusipov I, Bacalini MG, Kalyakulina A et al. Age-related DNA methylation changes are sex-specific: a comprehensive assessment. Aging (Albany NY) 2020;12:24057–24080.
    1. Honigberg MC, Zekavat SM, Niroula A et al. Premature Menopause, Clonal Hematopoiesis, and Coronary Artery Disease in Postmenopausal Women. Circulation 2021;143:410–423.
    1. Marquez EJ, Chung CH, Marches R et al. Sexual-dimorphism in human immune system aging. Nat Commun 2020;11:751.
    1. Kane AE, Bisset ES, Heinze-Milne S, Keller KM, Grandy SA, Howlett SE. Maladaptive Changes Associated With Cardiac Aging Are Sex-Specific and Graded by Frailty and Inflammation in C57BL/6 Mice. J Gerontol A Biol Sci Med Sci 2021;76:233–243.

Source: PubMed

3
Subscribe