1
|
Amaya-Garrido A, Klein J. The role of calprotectin in vascular calcification. Curr Opin Nephrol Hypertens 2025; 34:276-283. [PMID: 40152927 DOI: 10.1097/mnh.0000000000001075] [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: 03/30/2025]
Abstract
PURPOSE OF REVIEW Vascular calcification significantly contributes to cardiovascular morbidity and mortality, particularly in high-risk populations like chronic kidney disease (CKD) patients. Calprotectin, a heterodimeric protein with pro-inflammatory and pro-calcific properties, has emerged as a key molecule in vascular pathology. This review highlights the mechanisms linking calprotectin to vascular calcification, its clinical relevance, and its potential as a therapeutic target. RECENT FINDINGS Vascular calcification is an active, cell-mediated process involving vascular smooth muscle cell (VSMC) dysfunction, inflammation, matrix remodeling, and cellular senescence. Calprotectin is strongly associated with cardiovascular disease and vascular calcification, particularly in CKD. Mechanistic studies reveal that calprotectin promotes calcification through the activation of RAGE and TLR4 signaling pathways, driving inflammatory cascades. Preclinical studies demonstrate that pharmacological inhibition of calprotectin attenuates vascular calcification in animal models, supporting its potential as a therapeutic target. SUMMARY Calprotectin is emerging as a promising biomarker and therapeutic target in vascular calcification, particularly in CKD and aging-related vascular diseases. However, further studies are required to clarify its mechanisms and assess the long-term efficacy and safety of calprotectin-targeted therapies. A deeper understanding of calprotectin's multifaceted role could pave the way for innovative therapeutic strategies targeting both inflammation and mineralization in calcification-related vascular diseases.
Collapse
Affiliation(s)
- Ana Amaya-Garrido
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease
- Université Toulouse III Paul-Sabatier, Toulouse, France
| |
Collapse
|
2
|
Rangel-López A, Mata-Rocha M, Pérez-González OA, López-Romero R, López-Sánchez DM, Juárez-Méndez S, Villegas-Ruiz V, Méndez-Tenorio A, Mejía-Araguré JM, Orihuela-Rodríguez O, Álvarez-Aguilar C, Majluf-Cruz A, Amato D, Zavala-Vega S, Melchor-Doncel de la Torre S, Paniagua-Sierra R, Arellano-Galindo J. Gene Expression Profile of Cultured Human Coronary Arterial Endothelial Cells Exposed to Serum from Chronic Kidney Disease Patients: Role of MAPK Signaling Pathway. Int J Mol Sci 2025; 26:3732. [PMID: 40332370 PMCID: PMC12027878 DOI: 10.3390/ijms26083732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Patients with end-stage renal disease (ESRD) are at increased risk of cardiovascular disease (CVD), such as myocardial infarction (MI). Uremic toxins and endothelial dysfunction are central to this process. In this exploratory study, we used the Affymetrix GeneChip microarray to investigate the gene expression profile in uremic serum-induced human coronary arterial endothelial cells (HCAECs) from ESRD patients with and without MI (UWI and UWOI groups) as an approach to its underlying mechanism. We also explored which pathways are involved in this process. We found 100 differentially expressed genes (DEGs) among the conditions of interest by supervised principal component analysis and hierarchical cluster analysis. The expressions of four major DEGs were validated by quantitative RT-PCR. Pathway analysis and molecular network were used to analyze the interaction and expression patterns. Ten pathways were identified as the main enriched metabolic pathways according to the transcriptome profiling analysis, which were, among others, positive regulation of inflammatory response, positive regulation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) cascade, cardiac muscle cell development, highlighting positive regulation of mitogen-activated protein kinase (MAPK) activity (p = 0.00016). Up- and down-regulation of genes from HCAECs exposed to uremic serum could contribute to increased endothelial dysfunction and CVD in ESRD patients. Our study suggests that inflammation and the ERK-MAPK pathway are highly enriched in kidney disease patients with MI, suggesting their role in ESRD pathology. Further studies and approaches based on MAPK pathway interfering strategies are needed to confirm these data.
Collapse
Affiliation(s)
- Angélica Rangel-López
- Unidad de Investigación Médica en Enfermedades Nefrológicas, UMAE Hospital de Especialidades, Centro Médico Nacional (CMN) Siglo XXI (SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City 06720, Mexico;
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez-Secretaría de Salud (SS), Mexico City 06720, Mexico;
| | - Minerva Mata-Rocha
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría, CMN SXXI IMSS, Mexico City 06720, Mexico;
| | - Oscar Alberto Pérez-González
- Laboratorio de Oncología Experimental, Instituto Nacional de Pediatría-SS, Mexico City 04530, Mexico; (O.A.P.-G.); (S.J.-M.); (V.V.-R.)
| | - Ricardo López-Romero
- Unidad de Investigación en Biomedicina y Oncología Genómica, Hospital de Gineco-Pediatría 3A, IMSS, Mexico City 07760, Mexico;
| | - Dulce María López-Sánchez
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, SS, Mexico City 14080, Mexico;
- Laboratorio de Biotecnología y Bioinformática Genómica, ENCB-Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Sergio Juárez-Méndez
- Laboratorio de Oncología Experimental, Instituto Nacional de Pediatría-SS, Mexico City 04530, Mexico; (O.A.P.-G.); (S.J.-M.); (V.V.-R.)
| | - Vanessa Villegas-Ruiz
- Laboratorio de Oncología Experimental, Instituto Nacional de Pediatría-SS, Mexico City 04530, Mexico; (O.A.P.-G.); (S.J.-M.); (V.V.-R.)
| | - Alfonso Méndez-Tenorio
- Laboratorio de Biotecnología y Bioinformática Genómica, ENCB-Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Juan Manuel Mejía-Araguré
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, SS, Mexico City 14610, Mexico;
| | - Oscar Orihuela-Rodríguez
- Departamento Clínico de Cardiología-UMAE Hospital de Especialidades, CMN SXXI IMSS, Mexico City 06720, Mexico;
| | - Cleto Álvarez-Aguilar
- Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolas de Hidalgo, Morelia, Michoacán 58020, Mexico;
| | - Abraham Majluf-Cruz
- Unidad de Investigación Médica en Hemostasia, Trombosis y Aterogénesis, Hospital General Regional 1, IMSS, Mexico City 03103, Mexico;
| | - Dante Amato
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico;
| | - Sergio Zavala-Vega
- Laboratorio Clínico y Banco de Sangre, Instituto Nacional de Neurología y Neurocirugía, SS, Mexico City 14269, Mexico;
| | - Silvia Melchor-Doncel de la Torre
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez-Secretaría de Salud (SS), Mexico City 06720, Mexico;
| | - Ramón Paniagua-Sierra
- Unidad de Investigación Médica en Enfermedades Nefrológicas, UMAE Hospital de Especialidades, Centro Médico Nacional (CMN) Siglo XXI (SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City 06720, Mexico;
| | - José Arellano-Galindo
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez-Secretaría de Salud (SS), Mexico City 06720, Mexico;
| |
Collapse
|
3
|
Kelly DM, Kelleher EM, Rothwell PM. The Kidney-Immune-Brain Axis: The Role of Inflammation in the Pathogenesis and Treatment of Stroke in Chronic Kidney Disease. Stroke 2025; 56:1069-1081. [PMID: 39851054 PMCID: PMC11932449 DOI: 10.1161/strokeaha.124.047070] [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: 01/25/2025]
Abstract
Cardiovascular diseases such as stroke are a major cause of morbidity and mortality for patients with chronic kidney disease (CKD). The underlying mechanisms connecting CKD and cardiovascular disease are yet to be fully elucidated, but inflammation is proposed to play an important role based on genetic association studies, studies of inflammatory biomarkers, and clinical trials of anti-inflammatory drug targets. There are multiple sources of both endogenous and exogenous inflammation in CKD, including increased production and decreased clearance of proinflammatory cytokines, oxidative stress, metabolic acidosis, chronic and recurrent infections, dialysis access, changes in adipose tissue metabolism, and disruptions in intestinal microbiota. This review focuses on the mechanisms of inflammation in CKD, dialysis and associated therapies, its proposed impact on stroke pathogenesis and prognosis, and the potential role of anti-inflammatory agents in the prevention and treatment of stroke in patients with CKD.
Collapse
Affiliation(s)
- Dearbhla M. Kelly
- Wolfson Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (D.M.K., P.M.R.)
| | - Eoin M. Kelleher
- Nuffield Department of Clinical Neurosciences (E.M.K.), University of Oxford, United Kingdom
| | - Peter M. Rothwell
- Wolfson Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (D.M.K., P.M.R.)
| |
Collapse
|
4
|
Rahbar Saadat Y, Abbasi A, Hejazian SS, Hekmatshoar Y, Ardalan M, Farnood F, Zununi Vahed S. Combating chronic kidney disease-associated cachexia: A literature review of recent therapeutic approaches. BMC Nephrol 2025; 26:133. [PMID: 40069669 PMCID: PMC11895341 DOI: 10.1186/s12882-025-04057-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025] Open
Abstract
In 2008, the Society on Sarcopenia, Cachexia, and Wasting Disorders introduced a generic definition for all types of cachexia: "a complex metabolic syndrome associated with the underlying illness characterized by a loss of muscle, with or without fat loss". It is well-known that the presence of inflammatory burden in end-stage renal disease (ESRD) patients may lead to the evolution of cachexia. Since the etiology of cachexia in chronic kidney disease (CKD) is multifactorial, thus the successful treatment must involve several concomitant measures (nutritional interventions, appetite stimulants, and anti-inflammatory pharmacologic agents) to provide integrated effective therapeutic modalities to combat causative factors and alleviate the outcomes of patients. Given the high mortality rate associated with cachexia, developing new therapeutic modalities are prerequisite for ameliorating patients with CKD worldwide. The present review aims to discuss some therapeutic strategies and provide an update on advances in nutritional approaches to counteract cachexia.
Collapse
Affiliation(s)
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Sina Hejazian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Hekmatshoar
- Medical Biology Department, School of Medicine, Altinbas University, Istanbul, Türkiye
| | | | - Farahnoosh Farnood
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
5
|
Ho CY, Wu MY, Thammaphet J, Ahmad S, Ho C.S. J, Draganova L, Anderson G, Jonnalagadda US, Hayward R, Shroff R, Wen WTL, Verhulst A, Foo RSY, Shanahan CM. Mineral Stress Drives Loss of Heterochromatin: An Early Harbinger of Vascular Inflammaging and Calcification. Circ Res 2025; 136:379-399. [PMID: 39840455 PMCID: PMC11825498 DOI: 10.1161/circresaha.124.325374] [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: 08/08/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Vascular calcification is a detrimental aging pathology markedly accelerated in patients with chronic kidney disease. PLA (prelamin A) is a biomarker of vascular smooth muscle cell aging that accelerates calcification however the mechanisms remain undefined. METHODS Vascular smooth muscle cells were transduced with PLA using an adenoviral vector and epigenetic modifications were monitored using immunofluorescence and targeted polymerase chain reaction array. Epigenetic findings were verified in vivo using immunohistochemistry in human vessels, in a mouse model of inducible prelamin A expression, and in a rat model of chronic kidney disease-induced calcification. Transcriptomic and chromatin immunoprecipitation followed by sequencing analyses were used to identify gene targets impacted by changes in the epigenetic landscape. Molecular tools and antibody arrays were used to monitor the effects of mineral dysregulation on heterochromatin, inflammation, aging, and calcification. RESULTS Here, we report that depletion of the repressive heterochromatin marks, H3K9me3 (histone H3, lysine 9, trimethylation) and H3K27me3 (histone H3, lysine 27,trimethylation), is an early hallmark of vascular aging induced by both nuclear lamina dysfunction and dysregulated mineral metabolism, which act to modulate the expression of key epigenetic writers and erasers. Global analysis of H3K9me3 and H3K27me3 marks and pathway analysis revealed deregulation of insulin signaling and autophagy pathways as well as cross-talking DNA damage and NF-κB (nuclear factor κB) inflammatory pathways consistent with early activation of the senescence-associated secretory phenotype. Expression of PLA in vivo induced loss of heterochromatin and promoted inflammation and osteogenic differentiation which preceded aging indices, such as DNA damage and senescence. Vessels from children on dialysis and rats with chronic kidney disease showed prelamin A accumulation and accelerated loss of heterochromatin before the onset of calcification. CONCLUSIONS Dysregulated mineral metabolism drives changes in the epigenetic landscape and nuclear lamina dysfunction that together promote early induction of inflammaging pathways priming the vasculature for downstream pathological change.
Collapse
MESH Headings
- Animals
- Heterochromatin/metabolism
- Heterochromatin/pathology
- Heterochromatin/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Humans
- Epigenesis, Genetic
- Rats
- Mice
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cellular Senescence
- Inflammation/metabolism
- Inflammation/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Histones/metabolism
- Mice, Inbred C57BL
- Cells, Cultured
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Lamin Type A
Collapse
Affiliation(s)
- Chin Yee Ho
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Meng-Ying Wu
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Jirapath Thammaphet
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Sadia Ahmad
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - James Ho C.S.
- Nanyang Technological University, Singapore (J.H.C.S., U.S.J.)
| | - Lilia Draganova
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Grace Anderson
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | | | - Robert Hayward
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Rukshana Shroff
- Nephrology Unit, Great Ormond Street Hospital and University College London Institute of Child Health, United Kingdom (R.S.)
| | - Wilson Tan Lek Wen
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium (A.V.)
| | - Roger SY. Foo
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Catherine M. Shanahan
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| |
Collapse
|
6
|
Hejazian SM, Hejazian SS, Mostafavi SM, Hosseiniyan SM, Montazersaheb S, Ardalan M, Zununi Vahed S, Barzegari A. Targeting cellular senescence in kidney diseases and aging: A focus on mesenchymal stem cells and their paracrine factors. Cell Commun Signal 2024; 22:609. [PMID: 39696575 DOI: 10.1186/s12964-024-01968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Cellular senescence is a phenomenon distinguished by the halting of cellular division, typically triggered by DNA injury or numerous stress-inducing factors. Cellular senescence is implicated in various pathological and physiological processes and is a hallmark of aging. The presence of accumulated senescent cells, whether transiently (acute senescence) or persistently (chronic senescence) plays a dual role in various conditions such as natural kidney aging and different kidney disorders. Elevations in senescent cells and senescence-associated secretory phenotype (SASP) levels correlate with decreased kidney function, kidney ailments, and age-related conditions. Strategies involving senotherapeutic agents like senolytics, senomorphics, and senoinflammation have been devised to specifically target senescent cells. Mesenchymal stem cells (MSCs) and their secreted factors may also offer alternative approaches for anti-senescence interventions. The MSC-derived secretome compromises significant therapeutic benefits in kidney diseases by facilitating tissue repair via anti-inflammatory, anti-fibrosis, anti-apoptotic, and pro-angiogenesis effects, thereby improving kidney function and mitigating disease progression. Moreover, by promoting the clearance of senescent cells or modulating their secretory profiles, MSCs could potentially reverse some age-related declines in kidney function.This review article intends to shed light on the present discoveries concerning the role of cellular senescence in kidney aging and diseases. Furthermore, it outlines the role of senotherapeutics utilized to alleviate kidney damage and aging. It also highlights the possible impact of MSCs secretome on mitigating kidney injury and prolonging lifespan across various models of kidney diseases as a novel senotherapy.
Collapse
Affiliation(s)
| | - Seyyed Sina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyedeh Mina Mostafavi
- Ayatollah Taleghani Hospital, Research Development Unit, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
McCallion S, McLarnon T, Cooper E, English AR, Watterson S, Chemaly ME, McGeough C, Eakin A, Ahmed T, Gardiner P, Pendleton A, Wright G, McGuigan D, O’Kane M, Peace A, Kuan Y, Gibson DS, McClean PL, Kelly C, McGilligan V, Murray EK, McCarroll F, Bjourson AJ, Rai TS. Senescence Biomarkers CKAP4 and PTX3 Stratify Severe Kidney Disease Patients. Cells 2024; 13:1613. [PMID: 39404377 PMCID: PMC11475272 DOI: 10.3390/cells13191613] [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: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Cellular senescence is the irreversible growth arrest subsequent to oncogenic mutations, DNA damage, or metabolic insult. Senescence is associated with ageing and chronic age associated diseases such as cardiovascular disease and diabetes. The involvement of cellular senescence in acute kidney injury (AKI) and chronic kidney disease (CKD) is not fully understood. However, recent studies suggest that such patients have a higher-than-normal level of cellular senescence and accelerated ageing. METHODS This study aimed to discover key biomarkers of senescence in AKI and CKD patients compared to other chronic ageing diseases in controls using OLINK proteomics. RESULTS We show that senescence proteins CKAP4 (p-value < 0.0001) and PTX3 (p-value < 0.0001) are upregulated in AKI and CKD patients compared with controls with chronic diseases, suggesting the proteins may play a role in overall kidney disease development. CONCLUSIONS CKAP4 was found to be differentially expressed in both AKI and CKD when compared to UHCs; hence, this biomarker could be a prognostic senescence biomarker of both AKI and CKD.
Collapse
Affiliation(s)
- Sean McCallion
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Thomas McLarnon
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Eamonn Cooper
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Andrew R. English
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
- School of Health and Life Sciences, Teesside University, Campus Heart, Middlesbrough TS1 3BX, UK
| | - Steven Watterson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Melody El Chemaly
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Cathy McGeough
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Amanda Eakin
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Tan Ahmed
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Philip Gardiner
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Adrian Pendleton
- Belfast Health and Social Care Trust (BHSCT), Belfast City Hospital, Belfast BT9 7AB, UK
| | - Gary Wright
- Belfast Health and Social Care Trust (BHSCT), Belfast City Hospital, Belfast BT9 7AB, UK
| | - Declan McGuigan
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Maurice O’Kane
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Aaron Peace
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Ying Kuan
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - David S. Gibson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Paula L. McClean
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Catriona Kelly
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Victoria McGilligan
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Elaine K. Murray
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Frank McCarroll
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Anthony J. Bjourson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Taranjit Singh Rai
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| |
Collapse
|
8
|
Williams ZJ, Chow L, Dow S, Pezzanite LM. The potential for senotherapy as a novel approach to extend life quality in veterinary medicine. Front Vet Sci 2024; 11:1369153. [PMID: 38812556 PMCID: PMC11133588 DOI: 10.3389/fvets.2024.1369153] [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: 01/11/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Cellular senescence, a condition where cells undergo arrest and can assume an inflammatory phenotype, has been associated with initiation and perpetuation of inflammation driving multiple disease processes in rodent models and humans. Senescent cells secrete inflammatory cytokines, proteins, and matrix metalloproteinases, termed the senescence associated secretory phenotype (SASP), which accelerates the aging processes. In preclinical models, drug interventions termed "senotherapeutics" selectively clear senescent cells and represent a promising strategy to prevent or treat multiple age-related conditions in humans and veterinary species. In this review, we summarize the current available literature describing in vitro evidence for senotheraputic activity, preclinical models of disease, ongoing human clinical trials, and potential clinical applications in veterinary medicine. These promising data to date provide further justification for future studies identifying the most active senotherapeutic combinations, dosages, and routes of administration for use in veterinary medicine.
Collapse
Affiliation(s)
- Zoë J. Williams
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Steven Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lynn M. Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
9
|
Russo E, Zanetti V, Macciò L, Benizzelli G, Carbone F, La Porta E, Esposito P, Verzola D, Garibotto G, Viazzi F. SGLT2 inhibition to target kidney aging. Clin Kidney J 2024; 17:sfae133. [PMID: 38803397 PMCID: PMC11129592 DOI: 10.1093/ckj/sfae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Indexed: 05/29/2024] Open
Abstract
Anti-aging therapy is the latest frontier in the world of medical science, especially for widespread diseases such as chronic kidney disease (CKD). Both renal aging and CKD are characterized by increased cellular senescence, inflammation and oxidative stress. A variety of cellular signalling mechanisms are involved in these processes, which provide new potential targets for therapeutic strategies aimed at counteracting the onset and progression of CKD. At the same time, sodium-glucose co-transporter 2 inhibitors (SGLT2is) continuously demonstrate large beneficial effects at all stages of the cardiorenal metabolic continuum. The broad-spectrum benefits of SGLT2is have led to changes in several treatment guidelines and to growing scientific interest in the underlying working principles. Multiple mechanisms have been studied to explain these great renal benefits, but many things remain to be solved. With this in mind, we provide an overview of the experimental evidence for the effects of SGLT2is on the molecular pathway's ability to modulate senescence, aging and parenchymal damage, especially at the kidney level. We propose to shed some light on the role of SGLT2is in kidney care by focusing on their potential to reduce the progression of kidney disease across the spectrum of aging and dysregulation of senescence.
Collapse
Affiliation(s)
- Elisa Russo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Lucia Macciò
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Edoardo La Porta
- UO Nephrology Dialysis and Transplant, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- UOSD Dialysis IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Pasquale Esposito
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | - Francesca Viazzi
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
10
|
Schwarz A, Hernandez L, Arefin S, Sartirana E, Witasp A, Wernerson A, Stenvinkel P, Kublickiene K. Sweet, bloody consumption - what we eat and how it affects vascular ageing, the BBB and kidney health in CKD. Gut Microbes 2024; 16:2341449. [PMID: 38686499 PMCID: PMC11062370 DOI: 10.1080/19490976.2024.2341449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
In today's industrialized society food consumption has changed immensely toward heightened red meat intake and use of artificial sweeteners instead of grains and vegetables or sugar, respectively. These dietary changes affect public health in general through an increased incidence of metabolic diseases like diabetes and obesity, with a further elevated risk for cardiorenal complications. Research shows that high red meat intake and artificial sweeteners ingestion can alter the microbial composition and further intestinal wall barrier permeability allowing increased transmission of uremic toxins like p-cresyl sulfate, indoxyl sulfate, trimethylamine n-oxide and phenylacetylglutamine into the blood stream causing an array of pathophysiological effects especially as a strain on the kidneys, since they are responsible for clearing out the toxins. In this review, we address how the burden of the Western diet affects the gut microbiome in altering the microbial composition and increasing the gut permeability for uremic toxins and the detrimental effects thereof on early vascular aging, the kidney per se and the blood-brain barrier, in addition to the potential implications for dietary changes/interventions to preserve the health issues related to chronic diseases in future.
Collapse
Affiliation(s)
- Angelina Schwarz
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leah Hernandez
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Samsul Arefin
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Sartirana
- Department of Translational Medicine, Nephrology and Kidney Transplantation Unit, University of Piemonte Orientale, Novara, Italy
| | - Anna Witasp
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika Wernerson
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Karolina Kublickiene
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Leotta C, Hernandez L, Tothova L, Arefin S, Ciceri P, Cozzolino MG, Barany P, Chromek M, Stenvinkel P, Kublickiene K. Levels of Cell-Free DNA in Kidney Failure Patients before and after Renal Transplantation. Cells 2023; 12:2774. [PMID: 38132094 PMCID: PMC10741614 DOI: 10.3390/cells12242774] [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: 10/30/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
Circulating cell-free DNA (cfDNA) has diverse applications in oncological, prenatal, toxicological, cardiovascular, and autoimmune diseases, diagnostics, and organ transplantation. In particular, mitochondrial cfDNA (mt-cfDNA) is associated with inflammation and linked to early vascular ageing (EVA) in end-stage kidney failure (ESKF), which could be a noninvasive marker for graft rejection and organ damage. Plasma samples from 44 ESKF patients, of whom half (n = 22) underwent either conservative therapy (non-HD) or hemodialysis (HD) before kidney transplantation (KT). These samples were analyzed at baseline and two years after KT. cfDNA was extracted from plasma and quantified using the fluorometric method. qPCR was used to quantify and differentiate the fractions of mt-cfDNA and nuclear cfDNA (nc-cfDNA). mt-cfDNA levels in KT patients decreased significantly from baseline to two years post-KT (p < 0.0268), while levels of total cfDNA and nc-cfDNA did not differ. Depending on therapy modality (HD vs. non-HD) before KT, total cfDNA levels were higher in HD patients at both baseline (p = 0.0133) and two years post-KT (p = 0.0421), while nc-cfDNA levels were higher in HD only at baseline (p = 0.0079). Males showed a nonsignificant trend of higher cfDNA levels. Patients with assessed vascular fibrosis (p = 0.0068), either alone or in combination with calcification plus fibrosis, showed reduced mt-cfDNA post-KT (p = 0.0195). Changes in mt-cfDNA levels suggests the impact of KT on the inflammatory state of ESKF, as evidenced via its correlation with high sensitivity C-reactive protein after KT. Further studies are warranted to assess if cfDNA could serve as a noninvasive method for monitoring the response to organ transplantation and even for amelioration of EVA status per se.
Collapse
Affiliation(s)
- Chiara Leotta
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo Hospital Milan, University of Milan, 20142 Milan, Italy (M.G.C.)
| | - Leah Hernandez
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
| | - Lubomira Tothova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
| | - Samsul Arefin
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo Hospital Milan, University of Milan, 20142 Milan, Italy (M.G.C.)
| | - Mario Gennaro Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo Hospital Milan, University of Milan, 20142 Milan, Italy (M.G.C.)
| | - Peter Barany
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
| | - Milan Chromek
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
- Division of Pediatrics, Clinical Science, Intervention and Technology (CLINTEC), Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
| | - Karolina Kublickiene
- Division of Renal Medicine, Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden; (C.L.); (L.H.); (P.B.); (M.C.)
| |
Collapse
|
12
|
Tanriover C, Copur S, Mutlu A, Peltek IB, Galassi A, Ciceri P, Cozzolino M, Kanbay M. Early aging and premature vascular aging in chronic kidney disease. Clin Kidney J 2023; 16:1751-1765. [PMID: 37915901 PMCID: PMC10616490 DOI: 10.1093/ckj/sfad076] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Indexed: 11/03/2023] Open
Abstract
Aging is the progressive decline of body functions and a number of chronic conditions can lead to premature aging characterized by frailty, a diseased vasculature, osteoporosis, and muscle wasting. One of the major conditions associated with premature and accelerated aging is chronic kidney disease (CKD), which can also result in early vascular aging and the stiffening of the arteries. Premature vascular aging in CKD patients has been considered as a marker of prognosis of mortality and cardiovascular morbidity and therefore requires further attention. Oxidative stress, inflammation, advanced glycation end products, fructose, and an aberrant gut microbiota can contribute to the development of early aging in CKD patients. There are several key molecular pathways and molecules which play a role in aging and vascular aging including nuclear factor erythroid 2-related factor 2 (Nrf-2), AMP-activated protein kinase (AMPK), sirtuin 1 (SIRT1), and klotho. Potential therapeutic strategies can target these pathways. Future studies are needed to better understand the importance of premature aging and early vascular aging and to develop therapeutic alternatives for these conditions.
Collapse
Affiliation(s)
- Cem Tanriover
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | | | - Andrea Galassi
- Department of Health Sciences, Renal Division, University of Milan, Milan, Italy
| | - Paola Ciceri
- Department of Health Sciences, Renal Division, University of Milan, Milan, Italy
| | - Mario Cozzolino
- Department of Health Sciences, Renal Division, University of Milan, Milan, Italy
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
13
|
Ward LJ, Laucyte-Cibulskiene A, Hernandez L, Ripsweden J, Stenvinkel P, Kublickiene K. Coronary artery calcification and aortic valve calcification in patients with kidney failure: a sex-disaggregated study. Biol Sex Differ 2023; 14:48. [PMID: 37443048 PMCID: PMC10347725 DOI: 10.1186/s13293-023-00530-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is linked to an increased cardiovascular disease (CVD) burden. Albeit underappreciated, sex differences are evident in CKD with females being more prone to CKD development, but males progressing more rapidly to kidney failure (KF). Cardiovascular remodelling is a hallmark of CKD with increased arterial and valvular calcification contributing to CKD. However, little is known regarding sex differences in calcific cardiovascular remodelling in KF patients. Thus, we hypothesise that sex differences are present in coronary artery calcification (CAC) and aortic valve calcification (AVC) in patients with KF. METHODS KF patients, males (n = 214) and females (n = 107), that had undergone computer tomography (CT) assessment for CAC and AVC were selected from three CKD cohorts. All patients underwent non-contrast multi-detector cardiac CT scanning, with CAC and AVC scoring based on the Agatston method. Baseline biochemical measurements were retrieved from cohort databases, including plasma analyses for inflammation markers (IL-6, TNF, hsCRP) and oxidative stress by skin autofluorescence measuring advanced glycation end-products (AGE), amongst other variables. RESULTS Sex-disaggregated analyses revealed that CAC score was associated with age in both males and females (both p < 0.001). Age-adjusted analyses revealed that in males CAC was associated with diabetes mellitus (DM) (p = 0.018) and CVD (p = 0.011). Additionally, for females CAC associated with IL-6 (p = 0.005) and TNF (p = 0.004). In both females and males CAC associated with AGE (p = 0.042 and p = 0.05, respectively). CAC was associated with mortality for females (p = 0.015) independent of age. AVC in females was not reviewed due to low AVC-positive samples (n = 14). In males, in multivariable regression AVC was associated with age (p < 0.001) and inflammation, as measured by IL-6 (p = 0.010). CONCLUSIONS In female KF patients inflammatory burden and oxidative stress were associated with CAC. Whereas in male KF patients oxidative stress and inflammation were associated with CAC and AVC, respectively. Our findings suggest a sex-specific biomarker signature for cardiovascular calcification that may affect the development of cardiovascular complications in males and females with KF.
Collapse
Affiliation(s)
- Liam J Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86, Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Agne Laucyte-Cibulskiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86, Stockholm, Sweden
- Department of Nephrology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Jonaz Ripsweden
- Unit of Radiology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86, Stockholm, Sweden.
| |
Collapse
|
14
|
Fularski P, Krzemińska J, Lewandowska N, Młynarska E, Saar M, Wronka M, Rysz J, Franczyk B. Statins in Chronic Kidney Disease-Effects on Atherosclerosis and Cellular Senescence. Cells 2023; 12:1679. [PMID: 37443712 PMCID: PMC10340582 DOI: 10.3390/cells12131679] [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: 05/21/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Chronic kidney disease (CKD) is a serious health problem that can affect various systems in the human body. Renal failure promotes mechanisms of premature cellular aging and also features of generalized inflammation in the body, which translates into a close relationship between kidney dysfunction and cardiovascular disease (CVD). As kidney function deteriorates, cardiovascular risk and mortality increase in this group of patients. Oxidative stress and inflammation are two closely related processes that initiate a vicious cycle by activating each other. Together with aging, they represent the key factors that cause and exacerbate CVD in CKD. Patients with CKD are particularly vulnerable to the accumulation of aging endothelial cells, vascular smooth muscle and macrophages, increasing the risk of atherosclerosis. Several mechanisms are known that can lead to the progression of the aforementioned problems, such as the accumulation of uremic toxins, persistent inflammation, impaired lipid and electrolyte metabolism, nitric oxide (NO) deficiency, the increased production of reactive oxygen species (ROS) and damage to deoxyribonucleic acid (DNA) and mitochondria. According to research, we can distinguish a group of drugs that effectively counteract the negative effects of CKD-statins. This is a group of drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme-A (HMG-CoA) reductase and affect a number of cellular processes and pathways, resulting in the overall slowing of atherosclerosis and cellular aging.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Julia Krzemińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Natalia Lewandowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Maciej Saar
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Magdalena Wronka
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland;
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| |
Collapse
|
15
|
Hobson S, Arefin S, Witasp A, Hernandez L, Kublickiene K, Shiels PG, Stenvinkel P. Accelerated Vascular Aging in Chronic Kidney Disease: The Potential for Novel Therapies. Circ Res 2023; 132:950-969. [PMID: 37053277 DOI: 10.1161/circresaha.122.321751] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The pathophysiology of vascular disease is linked to accelerated biological aging and a combination of genetic, lifestyle, biological, and environmental risk factors. Within the scenario of uncontrolled artery wall aging processes, CKD (chronic kidney disease) stands out as a valid model for detailed structural, functional, and molecular studies of this process. The cardiorenal syndrome relates to the detrimental bidirectional interplay between the kidney and the cardiovascular system. In addition to established risk factors, this group of patients is subjected to a plethora of other emerging vascular risk factors, such as inflammation, oxidative stress, mitochondrial dysfunction, vitamin K deficiency, cellular senescence, somatic mutations, epigenetic modifications, and increased apoptosis. A better understanding of the molecular mechanisms through which the uremic milieu triggers and maintains early vascular aging processes, has provided important new clues on inflammatory pathways and emerging risk factors alike, and to the altered behavior of cells in the arterial wall. Advances in the understanding of the biology of uremic early vascular aging opens avenues to novel pharmacological and nutritional therapeutic interventions. Such strategies hold promise to improve future prevention and treatment of early vascular aging not only in CKD but also in the elderly general population.
Collapse
Affiliation(s)
- S Hobson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - S Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - A Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - L Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - K Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - P G Shiels
- School of Molecular Biosciences, MVLS, University of Glasgow, United Kingdom (P.G.S.)
| | - P Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| |
Collapse
|
16
|
Baaten CCFMJ, Vondenhoff S, Noels H. Endothelial Cell Dysfunction and Increased Cardiovascular Risk in Patients With Chronic Kidney Disease. Circ Res 2023; 132:970-992. [PMID: 37053275 PMCID: PMC10097498 DOI: 10.1161/circresaha.123.321752] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The endothelium is considered to be the gatekeeper of the vessel wall, maintaining and regulating vascular integrity. In patients with chronic kidney disease, protective endothelial cell functions are impaired due to the proinflammatory, prothrombotic and uremic environment caused by the decline in kidney function, adding to the increase in cardiovascular complications in this vulnerable patient population. In this review, we discuss endothelial cell functioning in healthy conditions and the contribution of endothelial cell dysfunction to cardiovascular disease. Further, we summarize the phenotypic changes of the endothelium in chronic kidney disease patients and the relation of endothelial cell dysfunction to cardiovascular risk in chronic kidney disease. We also review the mechanisms that underlie endothelial changes in chronic kidney disease and consider potential pharmacological interventions that can ameliorate endothelial health.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| |
Collapse
|
17
|
Hernandez L, Ward LJ, Arefin S, Barany P, Wennberg L, Söderberg M, Bruno S, Cantaluppi V, Stenvinkel P, Kublickiene K. Blood–Brain Barrier Biomarkers before and after Kidney Transplantation. Int J Mol Sci 2023; 24:ijms24076628. [PMID: 37047601 PMCID: PMC10095132 DOI: 10.3390/ijms24076628] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Kidney transplantation (KT) may improve the neurological status of chronic kidney disease (CKD) patients, reflected by the altered levels of circulating BBB-specific biomarkers. This study compares the levels of neuron specific enolase (NSE), brain-derived neurotrophic factor (BDNF), neurofilament light chain (NfL), and circulating plasma extracellular vesicles (EVs) in kidney-failure patients before KT and at a two-year follow up. Using ELISA, NSE, BDNF, and NfL levels were measured in the plasma of 74 living-donor KT patients. Plasma EVs were isolated with ultracentrifugation, and characterized for concentration/size and surface protein expression using flow cytometry from a subset of 25 patients. Lower NSE levels, and higher BDNF and NfL were observed at the two-year follow-up compared to the baseline (p < 0.05). Male patients had significantly higher BDNF levels compared to those of females. BBB biomarkers correlated with the baseline lipid profile and with glucose, vitamin D, and inflammation markers after KT. BBB surrogate marker changes in the microcirculation of early vascular aging phenotype patients with calcification and/or fibrosis were observed only in NSE and BDNF. CD31+ microparticles from endothelial cells expressing inflammatory markers such as CD40 and integrins were significantly reduced after KT. KT may, thus, improve the neurological status of CKD patients, as reflected by changes in BBB-specific biomarkers.
Collapse
Affiliation(s)
- Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Liam J. Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, 587 58 Linköping, Sweden
| | - Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Barany
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Magnus Söderberg
- Department of Pathology, Clinical Pharmacology and Safety Sciences, R&D AstraZeneca, 431 83 Gothenburg, Sweden
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, 10124 Torino, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplant Unit, Department of Translational Medicine (DIMET), University of Piemonte Orientale (UPO), “Maggiore della Carita” University Hospital, 28100 Novara, Italy
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
18
|
Branco P, Calça R, Martins AR, Mateus C, Jervis MJ, Gomes DP, Azeredo-Lopes S, De Melo Junior AF, Sousa C, Civantos E, Mas-Fontao S, Gaspar A, Ramos S, Morello J, Nolasco F, Rodrigues A, Pereira SA. Fibrosis of Peritoneal Membrane, Molecular Indicators of Aging and Frailty Unveil Vulnerable Patients in Long-Term Peritoneal Dialysis. Int J Mol Sci 2023; 24:5020. [PMID: 36902451 PMCID: PMC10002940 DOI: 10.3390/ijms24055020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Peritoneal membrane status, clinical data and aging-related molecules were investigated as predictors of long-term peritoneal dialysis (PD) outcomes. A 5-year prospective study was conducted with the following endpoints: (a) PD failure and time until PD failure, (b) major cardiovascular event (MACE) and time until MACE. A total of 58 incident patients with peritoneal biopsy at study baseline were included. Peritoneal membrane histomorphology and aging-related indicators were assessed before the start of PD and investigated as predictors of study endpoints. Fibrosis of the peritoneal membrane was associated with MACE occurrence and earlier MACE, but not with the patient or membrane survival. Serum α-Klotho bellow 742 pg/mL was related to the submesothelial thickness of the peritoneal membrane. This cutoff stratified the patients according to the risk of MACE and time until MACE. Uremic levels of galectin-3 were associated with PD failure and time until PD failure. This work unveils peritoneal membrane fibrosis as a window to the vulnerability of the cardiovascular system, whose mechanisms and links to biological aging need to be better investigated. Galectin-3 and α-Klotho are putative tools to tailor patient management in this home-based renal replacement therapy.
Collapse
Affiliation(s)
- Patrícia Branco
- Nephrology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2790-134 Lisboa, Portugal
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Rita Calça
- Nephrology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2790-134 Lisboa, Portugal
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Ana Rita Martins
- Nephrology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2790-134 Lisboa, Portugal
| | - Catarina Mateus
- Nephrology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2790-134 Lisboa, Portugal
| | - Maria João Jervis
- Surgery Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2740-134 Lisboa, Portugal
| | - Daniel Pinto Gomes
- Pathology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2740-134 Lisboa, Portugal
| | - Sofia Azeredo-Lopes
- CHRC, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Department of Statistics and Operational Research, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Antonio Ferreira De Melo Junior
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Cátia Sousa
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Ester Civantos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Sebastian Mas-Fontao
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Augusta Gaspar
- Nephrology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2790-134 Lisboa, Portugal
| | - Sância Ramos
- Pathology Department, Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental (CHLO), 2740-134 Lisboa, Portugal
| | - Judit Morello
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Fernando Nolasco
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Anabela Rodrigues
- UMIB—Unidade Multidisciplinar de Investigação Biomédica, ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
- Departamento de Nefrologia, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Centro Hospitalar Universitário do Porto (CHUdsA), 4050-345 Porto, Portugal
| | - Sofia Azeredo Pereira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| |
Collapse
|
19
|
Huang Y, Wang B, Hassounah F, Price SR, Klein J, Mohamed TMA, Wang Y, Park J, Cai H, Zhang X, Wang XH. The impact of senescence on muscle wasting in chronic kidney disease. J Cachexia Sarcopenia Muscle 2023; 14:126-141. [PMID: 36351875 PMCID: PMC9891952 DOI: 10.1002/jcsm.13112] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/29/2022] [Accepted: 09/19/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Muscle wasting is a common complication of chronic kidney disease (CKD) that is associated with higher mortality. Although the mechanisms of myofibre loss in CKD has been widely studied, the contribution of muscle precursor cell (MPC) senescence remains poorly understood. Senescent MPCs no longer proliferate and can produce proinflammatory factors or cytokines. In this study, we tested the hypothesis that the senescence associated secretory phenotype (SASP) of MPCs contributes to CKD-induced muscle atrophy and weakness. METHODS CKD was induced in mice by 5/6th nephrectomy. Kidney function, muscle size, and function were measured, and markers of atrophy, inflammation, and senescence were evaluated using immunohistochemistry, immunoblots, or qPCR. To study the impact of senescence, a senolytics cocktail of dasatinib + quercetin (D&Q) was given orally to mice for 8 weeks. To investigate CKD-induced senescence at the cellular level, primary MPCs were incubated with serum from CKD or control subjects. The roles of specific proteins in MPC senescence were studied using adenoviral transduction, siRNA, and plasmid transfection. RESULTS In the hindlimb muscles of CKD mice, (i) the senescence biomarker SA-β-gal was sharply increased (~30-fold); (ii) the DNA damage response marker γ-H2AX was increased 1.9-fold; and (iii) the senescence pathway markers p21 and p16INK4a were increased 1.99-fold and 2.82-fold, respectively (all values, P < 0.05), whereas p53 was unchanged. γ-H2AX, p21, and p16INK4A were negatively correlated at P < 0.05 with gastrocnemius weight, suggesting a causal relationship with muscle atrophy. Administration of the senolytics cocktail to CKD mice for 8 weeks eliminated the disease-related elevation of p21, p16INK4a , and γ-H2AX, abolished positive SA-β-gal, and depressed the high levels of the SASP cytokines, TNF-α, IL-6, IL-1β, and IFN (all values, P < 0.05). Skeletal muscle weight, myofibre cross-sectional area, and grip function were improved in CKD mice receiving D&Q. Markers of protein degradation, inflammation, and MPCs dysfunction were also attenuated by D&Q treatment compared with the vehicle treatment in 5/6th nephrectomy mice (all values, P < 0.05). Uraemic serum induced senescence in cultured MPCs. Overexpression of FoxO1a in MPCs increased the number of p21+ senescent cells, and p21 siRNA prevented uraemic serum-induced senescence (P < 0.05). CONCLUSIONS Senescent MPCs are likely to contribute to the development of muscle wasting during CKD by producing inflammatory cytokines. Limiting senescence with senolytics ameliorated muscle wasting and improved muscle strength in vivo and restored cultured MPC functions. These results suggest potential new therapeutic targets to improve muscle health and function in CKD.
Collapse
Affiliation(s)
- Ying Huang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease, Changsha, China
| | - Bin Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Institute of Nephrology, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Faten Hassounah
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - S Russ Price
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Janet Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, USA
| | - Yanhua Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Jeanie Park
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Nephrology Section, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Nephrology Section, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Xuemei Zhang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Williams MJ, White SC, Joseph Z, Hruska KA. Updates in the chronic kidney disease-mineral bone disorder show the role of osteocytic proteins, a potential mechanism of the bone-Vascular paradox, a therapeutic target, and a biomarker. Front Physiol 2023; 14:1120308. [PMID: 36776982 PMCID: PMC9909112 DOI: 10.3389/fphys.2023.1120308] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The chronic kidney disease-mineral bone disorder (CKD-MBD) is a complex multi-component syndrome occurring during kidney disease and its progression. Here, we update progress in the components of the syndrome, and synthesize recent investigations, which suggest a potential mechanism of the bone-vascular paradox. The discovery that calcified arteries in chronic kidney disease inhibit bone remodeling lead to the identification of factors produced by the vasculature that inhibit the skeleton, thus providing a potential explanation for the bone-vascular paradox. Among the factors produced by calcifying arteries, sclerostin secretion is especially enlightening. Sclerostin is a potent inhibitor of bone remodeling and an osteocyte specific protein. Its production by the vasculature in chronic kidney disease identifies the key role of vascular cell osteoblastic/osteocytic transdifferentiation in vascular calcification and renal osteodystrophy. Subsequent studies showing that inhibition of sclerostin activity by a monoclonal antibody improved bone remodeling as expected, but stimulated vascular calcification, demonstrate that vascular sclerostin functions to brake the Wnt stimulation of the calcification milieu. Thus, the target of therapy in the chronic kidney disease-mineral bone disorder is not inhibition of sclerostin function, which would intensify vascular calcification. Rather, decreasing sclerostin production by decreasing the vascular osteoblastic/osteocytic transdifferentiation is the goal. This might decrease vascular calcification, decrease vascular stiffness, decrease cardiac hypertrophy, decrease sclerostin production, reduce serum sclerostin and improve skeletal remodeling. Thus, the therapeutic target of the chronic kidney disease-mineral bone disorder may be vascular osteoblastic transdifferentiation, and sclerostin levels may be a useful biomarker for the diagnosis of the chronic kidney disease-mineral bone disorder and the progress of its therapy.
Collapse
Affiliation(s)
- Matthew J. Williams
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Sarah C. White
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Zachary Joseph
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Keith A. Hruska
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
- Departments of Medicine and Cell Biology, Washington University, Saint Louis, MO, United States
| |
Collapse
|
21
|
Figuer A, Alique M, Valera G, Serroukh N, Ceprían N, de Sequera P, Morales E, Carracedo J, Ramírez R, Bodega G. New mechanisms involved in the development of cardiovascular disease in chronic kidney disease. Nefrologia 2023; 43:63-80. [PMID: 37268501 DOI: 10.1016/j.nefroe.2023.05.014] [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: 11/15/2021] [Accepted: 03/02/2022] [Indexed: 06/04/2023] Open
Abstract
Chronic kidney disease (CKD) is a pathology with a high worldwide incidence and an upward trend affecting the elderly. When CKD is very advanced, the use of renal replacement therapies is required to prolong its life (dialysis or kidney transplantation). Although dialysis improves many complications of CKD, the disease does not reverse completely. These patients present an increase in oxidative stress, chronic inflammation and the release of extracellular vesicles (EVs), which cause endothelial damage and the development of different cardiovascular diseases (CVD). CKD patients develop premature diseases associated with advanced age, such as CVD. EVs play an essential role in developing CVD in patients with CKD since their number increases in plasma and their content is modified. The EVs of patients with CKD cause endothelial dysfunction, senescence and vascular calcification. In addition, miRNAs free or transported in EVs together with other components carried in these EVs promote endothelial dysfunction, thrombotic and vascular calcification in CKD, among other effects. This review describes the classic factors and focuses on the role of new mechanisms involved in the development of CVD associated with CKD, emphasizing the role of EVs in the development of cardiovascular pathologies in the context of CKD. Moreover, the review summarized the EVs' role as diagnostic and therapeutic tools, acting on EV release or content to avoid the development of CVD in CKD patients.
Collapse
Affiliation(s)
- Andrea Figuer
- Departamento de Biología de Sistemas, Universidad de Alcalá (IRYCIS), Alcalá de Henares (Madrid), Spain
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá (IRYCIS), Alcalá de Henares (Madrid), Spain.
| | - Gemma Valera
- Departamento de Biología de Sistemas, Universidad de Alcalá (IRYCIS), Alcalá de Henares (Madrid), Spain
| | - Nadia Serroukh
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (IMAS12), Madrid, Spain
| | - Noemí Ceprían
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (IMAS12), Madrid, Spain
| | - Patricia de Sequera
- Sección de Nefrología, Hospital Universitario Infanta Leonor, Universidad Complutense de Madrid, Madrid, Spain
| | - Enrique Morales
- Sección de Nefrología, Hospital 12 de Octubre, Madrid, Spain
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (IMAS12), Madrid, Spain
| | - Rafael Ramírez
- Departamento de Biología de Sistemas, Universidad de Alcalá (IRYCIS), Alcalá de Henares (Madrid), Spain
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá, Alcalá de Henares (Madrid), Spain
| |
Collapse
|
22
|
Greco A, Herrmann J, Babic M, Gummi MR, van der Giet M, Tölle M, Schuchardt M. Molecular Imaging and Quantification of Smooth Muscle Cell and Aortic Tissue Calcification In Vitro and Ex Vivo with a Fluorescent Hydroxyapatite-Specific Probe. Biomedicines 2022; 10:biomedicines10092271. [PMID: 36140372 PMCID: PMC9496085 DOI: 10.3390/biomedicines10092271] [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: 06/30/2022] [Revised: 08/22/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Vessel calcification is characterized by the precipitation of hydroxyapatite (HAP) in the vasculature. Currently, no causal therapy exists to reduce or prevent vessel calcification. Studying the underlying pathways within vascular smooth muscle cells and testing pharmacological intervention is a major challenge in the vascular research field. This study aims to establish a rapid and efficient working protocol for specific HAP detection in cells and tissue using the synthetic bisphosphonate fluorescence dye OsteoSense™. This protocol facilitates especially early quantification of the fluorescence signal and permits co-staining with other markers of interest, enabling smaller experimental set-ups with lesser primary cells consumption and fast workflows. The fluorescence-based detection of vascular calcification with OsteoSense™ combines a high specificity with improved sensitivity. Therefore, this methodology can improve research of the pathogenesis of vascular calcification, especially for testing the therapeutic benefit of inhibitors in the case of in vitro and ex vivo settings.
Collapse
|
23
|
Zhao JL, Qiao XH, Mao JH, Liu F, Fu HD. The interaction between cellular senescence and chronic kidney disease as a therapeutic opportunity. Front Pharmacol 2022; 13:974361. [PMID: 36091755 PMCID: PMC9459105 DOI: 10.3389/fphar.2022.974361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/03/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasingly serious public health problem in the world, but the effective therapeutic approach is quite limited at present. Cellular senescence is characterized by the irreversible cell cycle arrest, senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Renal senescence shares many similarities with CKD, including etiology, mechanism, pathological change, phenotype and outcome, however, it is difficult to judge whether renal senescence is a trigger or a consequence of CKD, since there is a complex correlation between them. A variety of cellular signaling mechanisms are involved in their interactive association, which provides new potential targets for the intervention of CKD, and then extends the researches on senotherapy. Our review summarizes the common features of renal senescence and CKD, the interaction between them, the strategies of senotherapy, and the open questions for future research.
Collapse
Affiliation(s)
- Jing-Li Zhao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Jian-Hua Mao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- *Correspondence: Jian-Hua Mao,
| | - Fei Liu
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hai-Dong Fu
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
24
|
Uremic mouse model to study vascular calcification and "inflamm-aging". J Mol Med (Berl) 2022; 100:1321-1330. [PMID: 35916902 PMCID: PMC9402761 DOI: 10.1007/s00109-022-02234-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/19/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022]
Abstract
Calcification and chronic inflammation of the vascular wall is a high-risk factor for cardiovascular mortality, especially in patients with chronic uremia. For the reduction or prevention of rapid disease progression, no specific treatment options are currently available. This study aimed to evaluate an adenine-based uremic mouse model for studying medial vessel calcification and senescence-associated secretory phenotype (SASP) changes of aortic tissue to unravel molecular pathogenesis and provide a model for therapy testing. The dietary adenine administration induced a stable and similar degree of chronic uremia in DBA2/N mice with an increase of uremia blood markers such as blood urea nitrogen, calcium, creatinine, alkaline phosphatase, and parathyroid hormone. Also, renal fibrosis and crystal deposits were detected upon adenine feeding. The uremic condition is related to a moderate to severe medial vessel calcification and subsequent elastin disorganization. In addition, expression of osteogenic markers as Bmp-2 and its transcription factor Sox-9 as well as p21 as senescence marker were increased in uremic mice compared to controls. Pro-inflammatory uremic proteins such as serum amyloid A, interleukin (Il)-1β, and Il-6 increased. This novel model of chronic uremia provides a simple method for investigation of signaling pathways in vascular inflammation and calcification and therefore offers an experimental basis for the development of potential therapeutic intervention studies.
Collapse
|
25
|
Lipid Profile Is Negatively Associated with Uremic Toxins in Patients with Kidney Failure-A Tri-National Cohort. Toxins (Basel) 2022; 14:toxins14060412. [PMID: 35737073 PMCID: PMC9231137 DOI: 10.3390/toxins14060412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/05/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Patients with kidney failure (KF) have a high incidence of cardiovascular (CV) disease, partly driven by insufficient clearance of uremic toxins. Recent investigations have questioned the accepted effects of adverse lipid profile and CV risk in uremic patients. Therefore, we related a panel of uremic toxins previously associated with CV morbidity/mortality to a full lipid profile in a large, tri-national, cross-sectional cohort. Total, high-density lipoprotein (HDL), non-HDL, low-density lipoprotein (LDL), and remnant cholesterol, as well as triglyceride, levels were associated with five uremic toxins in a cohort of 611 adult KF patients with adjustment for clinically relevant covariates and other patient-level variables. Univariate analyses revealed negative correlations of total, non-HDL, and LDL cholesterol with all investigated uremic toxins. Multivariate linear regression analyses confirmed independent, negative associations of phenylacetylglutamine with total, non-HDL, and LDL cholesterol, while indole-3 acetic acid associated with non-HDL and LDL cholesterol. Furthermore, trimethylamine-N-Oxide was independently and negatively associated with non-HDL cholesterol. Sensitivity analyses largely confirmed findings in the entire cohort. In conclusion, significant inverse associations between lipid profile and distinct uremic toxins in KF highlight the complexity of the uremic milieu, suggesting that not all uremic toxin interactions with conventional CV risk markers may be pathogenic.
Collapse
|
26
|
Erlandsson H, Qureshi AR, Ripsweden J, Haugen Löfman I, Söderberg M, Wennberg L, Lundgren T, Bruchfeld A, Brismar TB, Stenvinkel P. Scoring of medial arterial calcification predicts cardiovascular events and mortality after kidney transplantation. J Intern Med 2022; 291:813-823. [PMID: 35112417 PMCID: PMC9306575 DOI: 10.1111/joim.13459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Progression of vascular calcification causes cardiovascular disease, which is the most common cause of death in chronic kidney failure and after kidney transplantation (KT). The prognostic impact of the extent of medial vascular calcification at KT is unknown. METHODS In this prospective cohort study, we investigated the impact of medial calcification compared to a mix of intimal and medial calcification represented by coronary artery calcification (CAC score) and aortic valve calcification in 342 patients starting on kidney failure replacement therapy. The primary outcomes were cardiovascular events (CVE) and death. The median follow-up time was 6.4 years (interquartile range 3.7-9.6 years). Exposure was CAC score and arteria epigastrica medial calcification scored as none, mild, moderate, or severe by a pathologist at time of KT (n = 200). We divided the patients according to kidney failure replacement therapy during follow-up, that is, living donor KT, deceased donor KT, or dialysis. RESULTS Moderate to severe medial calcification in the arteria epigastrica was associated with higher mortality (p = 0.001), and the hazard ratio for CVE was 3.1 (95% confidence interval [CI] 1.12-9.02, p < 0.05) compared to no or mild medial calcification. The hazard ratio for 10-year mortality in the dialysis group was 33.6 (95% CI, 10.0-113.0, p < 0.001) compared to living donor recipients, independent of Framingham risk score and prevalent CAC. CONCLUSION Scoring of medial calcification in the arteria epigastrica identified living donor recipients as having 3.1 times higher risk of CVE, independent of traditional risk factors. The medial calcification score could be a reliable method to identify patients with high and low risk of CVE and mortality following KT.
Collapse
Affiliation(s)
- Helen Erlandsson
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Jonaz Ripsweden
- Department of Radiology, Karolinska University Hospital, Stockholm, Sweden.,Unit of radiology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ida Haugen Löfman
- Section of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Söderberg
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Wennberg
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Torkel B Brismar
- Department of Radiology, Karolinska University Hospital, Stockholm, Sweden.,Unit of radiology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Ren SC, Mao N, Yi S, Ma X, Zou JQ, Tang X, Fan JM. Vascular Calcification in Chronic Kidney Disease: An Update and Perspective. Aging Dis 2022; 13:673-697. [PMID: 35656113 PMCID: PMC9116919 DOI: 10.14336/ad.2021.1024] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a devastating condition resulting from irreversible loss of nephron numbers and function and leading to end-stage renal disease and mineral disorders. Vascular calcification, an ectopic deposition of calcium-phosphate salts in blood vessel walls and heart valves, is an independent risk factor of cardiovascular morbidity and mortality in chronic kidney disease. Moreover, aging and related metabolic disorders are essential risk factors for chronic kidney disease and vascular calcification. Marked progress has been recently made in understanding and treating vascular calcification in chronic kidney disease. However, there is a paucity of systematic reviews summarizing this progress, and investigating unresolved issues is warranted. In this systematic review, we aimed to overview the underlying mechanisms of vascular calcification in chronic kidney diseases and discuss the impact of chronic kidney disease on the pathophysiology of vascular calcification. Additionally, we summarized potential clinical diagnostic biomarkers and therapeutic applications for vascular calcification with chronic kidney disease. This review may offer new insights into the pathogenesis, diagnosis, and therapeutic intervention of vascular calcification.
Collapse
Affiliation(s)
- Si-Chong Ren
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
- Center for Translational Medicine, Sichuan Academy of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Mao
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Si Yi
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| | - Xin Ma
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Jia-Qiong Zou
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| |
Collapse
|
28
|
Updated Pathways in Cardiorenal Continuum after Kidney Transplantation. TRANSPLANTOLOGY 2022. [DOI: 10.3390/transplantology3020017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) remains one of the leading causes for increased morbidity and mortality in chronic kidney disease (CKD). Kidney transplantation is the preferred treatment option for CKD G5. Improved perioperative and postoperative care, personalized immunosuppressive regimes, and refined matching procedures of kidney transplants improves cardiovascular health in the early posttransplant period. However, the long-term burden of CVD is considerable. Previously underrecognized, the role of the complement system alongside innate immunity, inflammaging, structural changes in the glomerular filtration barrier and early vascular ageing also seem to play an important role in the posttransplant management. This review provides up-to-date knowledge on these pathways that may influence the cardiovascular and renal continuum and identifies potential targets for future therapies. Arterial destiffening strategies and the applicability of sodium-glucose cotransporter 2 inhibitors and their role in cardiovascular health after kidney transplantation are also addressed.
Collapse
|
29
|
Induction of Accelerated Aging in a Mouse Model. Cells 2022; 11:cells11091418. [PMID: 35563724 PMCID: PMC9102583 DOI: 10.3390/cells11091418] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
With the global increase of the elderly population, the improvement of the treatment for various aging-related diseases and the extension of a healthy lifespan have become some of the most important current medical issues. In order to understand the developmental mechanisms of aging and aging-related disorders, animal models are essential to conduct relevant studies. Among them, mice have become one of the most prevalently used model animals for aging-related studies due to their high similarity to humans in terms of genetic background and physiological structure, as well as their short lifespan and ease of reproduction. This review will discuss some of the common and emerging mouse models of accelerated aging and related chronic diseases in recent years, with the aim of serving as a reference for future application in fundamental and translational research.
Collapse
|
30
|
Figuer A, Alique M, Valera G, Serroukh N, Ceprían N, de Sequera P, Morales E, Carracedo J, Ramírez R, Bodega G. Nuevos mecanismos implicados en el desarrollo de la enfermedad cardiovascular en la enfermedad renal crónica. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
31
|
Hernandez L, Ward LJ, Arefin S, Ebert T, Laucyte-Cibulskiene A, Heimbürger O, Barany P, Wennberg L, Stenvinkel P, Kublickiene K. Blood-brain barrier and gut barrier dysfunction in chronic kidney disease with a focus on circulating biomarkers and tight junction proteins. Sci Rep 2022; 12:4414. [PMID: 35292710 PMCID: PMC8924178 DOI: 10.1038/s41598-022-08387-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/28/2022] [Indexed: 11/09/2022] Open
Abstract
Kidney failure and associated uraemia have implications for the cardiovascular system, brain, and blood–brain barrier (BBB). We aim to examine BBB disruption, by assessing brain-derived neurotropic factor (BDNF), neuron-specific enolase (NSE) levels, and gut-blood barrier (GBB) disruption by trimethylamine N-oxide (TMAO), in chronic kidney disease (CKD) patients. Additionally, endothelial tight-junction protein expressions and modulation via TMAO were assessed. Serum from chronic kidney disease (CKD) female and male haemodialysis (HD) patients, and controls, were used to measure BDNF and NSE by enzyme-linked immunosorbent assays, and TMAO by mass spectrometry. Immunofluorescent staining of subcutaneous fat biopsies from kidney transplant recipients, and controls, were used to measure microvascular expression of tight-junction proteins (claudin-5, occludin, JAM-1), and control microvasculature for TMAO effects. HD patients versus controls, had significantly lower and higher serum levels of BDNF and NSE, respectively. In CKD biopsies versus controls, reduced expression of claudin-5, occludin, and JAM-1 were observed. Incubation with TMAO significantly decreased expression of all tight-junction proteins in the microvasculature. Uraemia affects BBB and GBB resulting in altered levels of circulating NSE, BDNF and TMAO, respectively, and it also reduces expression of tight-junction proteins that confer BBB maintenance. TMAO serves as a potential candidate to alter BBB integrity in CKD.
Collapse
Affiliation(s)
- Leah Hernandez
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Liam J Ward
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Samsul Arefin
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Thomas Ebert
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Agne Laucyte-Cibulskiene
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden.,Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Olof Heimbürger
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Peter Barany
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Karolina Kublickiene
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, 14186, Stockholm, Sweden.
| |
Collapse
|
32
|
Molinari P, Alfieri CM, Mattinzoli D, Campise M, Cervesato A, Malvica S, Favi E, Messa P, Castellano G. Bone and Mineral Disorder in Renal Transplant Patients: Overview of Pathology, Clinical, and Therapeutic Aspects. Front Med (Lausanne) 2022; 9:821884. [PMID: 35360722 PMCID: PMC8960161 DOI: 10.3389/fmed.2022.821884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/08/2022] [Indexed: 12/25/2022] Open
Abstract
Renal transplantation (RTx) allows us to obtain the resolution of the uremic status but is not frequently able to solve all the metabolic complications present during end-stage renal disease. Mineral and bone disorders (MBDs) are frequent since the early stages of chronic kidney disease (CKD) and strongly influence the morbidity and mortality of patients with CKD. Some mineral metabolism (MM) alterations can persist in patients with RTx (RTx-p), as well as in the presence of complete renal function recovery. In those patients, anomalies of calcium, phosphorus, parathormone, fibroblast growth factor 23, and vitamin D such as bone and vessels are frequent and related to both pre-RTx and post-RTx specific factors. Many treatments are present for the management of post-RTx MBD. Despite that, the guidelines that can give clear directives in MBD treatment of RTx-p are still missed. For the future, to obtain an ever-greater individualisation of therapy, an increase of the evidence, the specificity of international guidelines, and more uniform management of these anomalies worldwide should be expected. In this review, the major factors related to post-renal transplant MBD (post-RTx-MBD), the main mineral metabolism biochemical anomalies, and the principal treatment for post-RTx MBD will be reported.
Collapse
Affiliation(s)
- Paolo Molinari
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Carlo Maria Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Deborah Mattinzoli
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Mariarosaria Campise
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Angela Cervesato
- Department of Nephrology, Clinical and Translational Sciences, Università degli Studi della Campania L.Vanvitelli, Naples, Italy
| | - Silvia Malvica
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Evaldo Favi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of General Surgery, Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Piergiorgio Messa
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
33
|
Ramírez R, Ceprian N, Figuer A, Valera G, Bodega G, Alique M, Carracedo J. Endothelial Senescence and the Chronic Vascular Diseases: Challenges and Therapeutic Opportunities in Atherosclerosis. J Pers Med 2022; 12:jpm12020215. [PMID: 35207703 PMCID: PMC8874678 DOI: 10.3390/jpm12020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is probably one of the paradigms of disease linked to aging. Underlying the physiopathology of atherosclerosis are cellular senescence, oxidative stress, and inflammation. These factors are increased in the elderly and from chronic disease patients. Elevated levels of oxidative stress affect cellular function and metabolism, inducing senescence. This senescence modifies the cell phenotype into a senescent secretory phenotype. This phenotype activates immune cells, leading to chronic systemic inflammation. Moreover, due to their secretory phenotype, senescence cells present an increased release of highlighted extracellular vesicles that will change nearby/neighborhood cells and paracrine signaling. For this reason, searching for specific senescent cell biomarkers and therapies against the development/killing of senescent cells has become relevant. Recently, senomorphic and senolityc drugs have become relevant in slowing down or eliminating senescence cells. However, even though they have shown promising results in experimental studies, their clinical use is still yet to be determined.
Collapse
Affiliation(s)
- Rafael Ramírez
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
| | - Noemi Ceprian
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
| | - Andrea Figuer
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
| | - Gemma Valera
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain;
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
- Correspondence: (M.A.); (J.C.)
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
- Correspondence: (M.A.); (J.C.)
| |
Collapse
|
34
|
Li Q, Deng Y, Liu L, Zhang C, Cai Y, Zhang T, Han M, Xu G. Sympathetic Denervation Ameliorates Renal Fibrosis via Inhibition of Cellular Senescence. Front Immunol 2022; 12:823935. [PMID: 35140713 PMCID: PMC8818683 DOI: 10.3389/fimmu.2021.823935] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Objective Continuous overactivation of the renal sympathetic nerve is considered to be an important cause of renal fibrosis. Accumulated senescent cells in the damaged kidney have metabolic activities and secrete amounts of proinflammatory factors as part of the SASP (the senescence-associated secretory phenotype), which induce chronic inflammation and fibrosis. It is still unclear whether renal sympathetic nerves affect renal inflammation and fibrosis by regulating cellular senescence. Therefore, we hypothesize that sympathetic activation in the injured kidney induces cellular senescence, which contributes to progressive renal inflammation and fibrosis. Methods Renal denervation was performed 2 days before the UUO (unilateral ureteral obstruction) and UIRI (unilateral ischemia-reperfusion injury) models. The effects of renal denervation on renal fibrosis and cellular senescence were observed. In vitro, cellular senescence was induced in renal proximal tubular epithelial cell lines (TKPTS cells) by treatment with norepinephrine (NE). The selective α2A-adrenergic receptor (α2A-AR) antagonists BRL44408 and β-arrestin2 siRNA, were administered to inhibit NE-induced cellular senescence. A significantly altered pathway was identified through immunoblotting, immunofluorescence, immunocytochemistry, and functional assays involved in mitochondrial function. Results Renal fibrosis and cellular senescence were significantly increased in UUO and UIRI models, which were partially reversed by renal denervation. In vitro, NE induced epithelial cells secreting proinflammatory cytokines and promoted cell senescence by activating α2A-AR. Importantly, the effects of NE during cellular senescence were blocked by α2A-AR selective antagonist and β-arrestin2 (downstream of α2A-AR) siRNA. Conclusion Renal sympathetic activation and cellular senescence are important neurometabolic and neuroimmune mechanisms in the development of renal fibrosis. Renal sympathetic neurotransmitter NE acting on the α2A-AR of epithelial cells promotes cellular senescence through the downstream β-arrestin2 signaling, which is a potential preventive target for renal fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Han
- *Correspondence: Gang Xu, ; Min Han,
| | - Gang Xu
- *Correspondence: Gang Xu, ; Min Han,
| |
Collapse
|
35
|
Phosphate Burden and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:7-13. [DOI: 10.1007/978-3-030-91623-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
36
|
Cheng FF, Liu YL, Du J, Lin JT. Metformin's Mechanisms in Attenuating Hallmarks of Aging and Age-Related Disease. Aging Dis 2022; 13:970-986. [PMID: 35855344 PMCID: PMC9286921 DOI: 10.14336/ad.2021.1213] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/13/2021] [Indexed: 11/01/2022] Open
Affiliation(s)
- Fang-Fang Cheng
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Yan-Li Liu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jun-Tang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
- Correspondence should be addressed to: Dr. Jun-Tang Lin, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
37
|
Mas-Bargues C, Alique M, Barrús-Ortiz MT, Borrás C, Rodrigues-Díez R. Exploring New Kingdoms: The Role of Extracellular Vesicles in Oxi-Inflamm-Aging Related to Cardiorenal Syndrome. Antioxidants (Basel) 2021; 11:78. [PMID: 35052582 PMCID: PMC8773353 DOI: 10.3390/antiox11010078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of age associated chronic diseases has increased in recent years. Although several diverse causes produce these phenomena, abundant evidence shows that oxidative stress plays a central role. In recent years, numerous studies have focused on elucidating the role of oxidative stress in the development and progression of both aging and chronic diseases, opening the door to the discovery of new underlying mechanisms and signaling pathways. Among them, senolytics and senomorphics, and extracellular vesicles offer new therapeutic strategies to slow the development of aging and its associated chronic diseases by decreasing oxidative stress. In this review, we aim to discuss the role of extracellular vesicles in human cardiorenal syndrome development and their possible role as biomarkers, targets, or vehicles of drugs to treat this syndrome.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Madrid, Spain;
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - María Teresa Barrús-Ortiz
- Área de Fisiología, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Univesidad Rey Juan Carlos, Avenida de Atenas s/n, 28922 Madrid, Spain
| | - Consuelo Borrás
- Grupo de Investigación Freshage, Departmento de Fisiología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (C.M.-B.); (C.B.)
- Instituto Sanitario de Investigación INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), 28029 Madrid, Spain
| | - Raquel Rodrigues-Díez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Hospital La Paz (IdiPAZ), 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 08036 Barcelona, Spain
| |
Collapse
|
38
|
Inserra F, Forcada P, Castellaro A, Castellaro C. Chronic Kidney Disease and Arterial Stiffness: A Two-Way Path. Front Med (Lausanne) 2021; 8:765924. [PMID: 34888327 PMCID: PMC8650118 DOI: 10.3389/fmed.2021.765924] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
The kidney-heart relationship has raised interest for the medical population since its vast and complex interaction significantly impacts health. Chronic kidney disease (CKD) generates vascular structure and function changes, with significant hemodynamic effects. The early arterial stiffening in CKD patients is a consequence of the interaction between oxidative stress and chronic vascular inflammation, leading to an accelerated deterioration of left ventricular function and alteration in tissue perfusion. CKD amplifies the inflammatory cascade's activation and is responsible for altering the endothelium function, increasing the vascular tone, wall thickening, and favors calcium deposits in the arterial wall. Simultaneously, the autonomic imbalance, and alteration in other hormonal systems, also favor the overactivation of inflammatory and fibrotic mediators. Thus, hormonal disarrangement also contributes to structural and functional lesions throughout the arterial wall. On the other hand, a rise in arterial stiffening and volume overload generates high left ventricular afterload. It increases the left ventricular burden with consequent myocardial remodeling, development of left ventricular hypertrophy and, in turn, heart failure. It is noteworthy that reduction in glomerular mass of renal diseases generates a compensatory glomerular filtration overdriven associated with large-arteries stiffness and high cardiovascular events. Furthermore, we consider that the consequent alterations of the arterial system's mechanical properties are crucial for altering tissue perfusion, mainly in low resistance. Thus, increasing the knowledge of these processes may help the reader to integrate them from a pathophysiological perspective, providing a comprehensive idea of this two-way path between arterial stiffness and renal dysfunction and their impact at the cardiovascular level.
Collapse
Affiliation(s)
- Felipe Inserra
- Advisor of Academic Vice-Rectory Department, Maimonides University, Buenos Aires, Argentina.,Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina
| | - Pedro Forcada
- Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina.,Non-Invasive Vascular Labs, CardioArenales and Diagnóstico Integral Médico (DIM) Prevención Cardiovascular, Buenos Aires, Argentina
| | - Agustina Castellaro
- Pediatric Medicine of Prof. Dr. Juan P Garrahan Hospital, Buenos Aires, Argentina
| | - Carlos Castellaro
- Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina.,Department of Nephrology, Centro de Educación Médica e Investigaciones Clínicas Norberto Quirno (CEMIC) Hospital, Buenos Aires, Argentina
| |
Collapse
|
39
|
Ebert T, Neytchev O, Witasp A, Kublickiene K, Stenvinkel P, Shiels PG. Inflammation and Oxidative Stress in Chronic Kidney Disease and Dialysis Patients. Antioxid Redox Signal 2021; 35:1426-1448. [PMID: 34006115 DOI: 10.1089/ars.2020.8184] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chronic kidney disease (CKD) can be regarded as a burden of lifestyle disease that shares common underpinning features and risk factors with the aging process; it is a complex constituted by several adverse components, including chronic inflammation, oxidative stress, early vascular aging, and cellular senescence. Recent Advances: A systemic approach to tackle CKD, based on mitigating the associated inflammatory, cell stress, and damage processes, has the potential to attenuate the effects of CKD, but it also preempts the development and progression of associated morbidities. In effect, this will enhance health span and compress the period of morbidity. Pharmacological, nutritional, and potentially lifestyle-based interventions are promising therapeutic avenues to achieve such a goal. Critical Issues: In the present review, currents concepts of inflammation and oxidative damage as key patho-mechanisms in CKD are addressed. In particular, potential beneficial but also adverse effects of different systemic interventions in patients with CKD are discussed. Future Directions: Senotherapeutics, the nuclear factor erythroid 2-related factor 2-kelch-like ECH-associated protein 1 (NRF2-KEAP1) signaling pathway, the endocrine klotho axis, inhibitors of the sodium-glucose cotransporter 2 (SGLT2), and live bio-therapeutics have the potential to reduce the burden of CKD and improve quality of life, as well as morbidity and mortality, in this fragile high-risk patient group. Antioxid. Redox Signal. 35, 1426-1448.
Collapse
Affiliation(s)
- Thomas Ebert
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ognian Neytchev
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Anna Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
40
|
Passage Number-Induced Replicative Senescence Modulates the Endothelial Cell Response to Protein-Bound Uremic Toxins. Toxins (Basel) 2021; 13:toxins13100738. [PMID: 34679030 PMCID: PMC8538293 DOI: 10.3390/toxins13100738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial aging may be induced early in pathological situations. The uremic toxins indoxyl sulfate (IS) and p-cresol (PC) accumulate in the plasma of chronic kidney disease (CKD) patients, causing accelerated endothelial aging, increased cardiovascular events and mortality. However, the mechanisms by which uremic toxins exert their deleterious effects on endothelial aging are not yet fully known. Thus, the aim of the present study is to determine the effects of IS and PC on endothelial damage and early senescence in cultured human umbilical vein endothelial cells (HUVECs). Hence, we establish an in vitro model of endothelial damage mediated by different passages of HUVECs and stimulated with different concentrations of IS and PC to evaluate functional effects on the vascular endothelium. We observe that cell passage-induced senescence is associated with apoptosis, ROS production and decreased endothelial proliferative capacity. Similarly, we observe that IS and PC cause premature aging in a dose-dependent manner, altering HUVECs' regenerative capacity, and decreasing their cell migration and potential to form vascular structures in vitro. In conclusion, IS and PC cause accelerated aging in HUVECs, thus contributing to endothelial dysfunction associated with CKD progression.
Collapse
|
41
|
Clark D, Kotronia E, Ramsay SE. Frailty, aging, and periodontal disease: Basic biologic considerations. Periodontol 2000 2021; 87:143-156. [PMID: 34463998 PMCID: PMC8771712 DOI: 10.1111/prd.12380] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aging is associated with the development of disease. Periodontal disease is one of the many diseases and conditions that increase in prevalence with age. In addition to the traditional focus on individual age-related conditions, there is now a greater recognition that multisystem conditions such as frailty play an important role in the health of older populations. Frailty is a clinical condition in older adults that increases the risk of adverse health outcomes. Both frailty and periodontal disease are common chronic conditions in older populations and share several risk factors. There is likely a bidirectional relationship between periodontal disease and frailty. Comorbid systemic diseases, poor physical functioning, and limited ability to self-care in frail older people have been implicated as underlying the association between frailty and periodontal disease. In addition, both frailty and periodontal disease also have strong associations with inflammatory dysregulation and other age-related pathophysiologic changes that may similarly underlie their development and progression. Investigating age-related changes in immune cells that regulate inflammation may lead to a better understanding of age-related disease and could lead to therapeutic targets for the improved management of frailty and periodontal disease.
Collapse
Affiliation(s)
- Daniel Clark
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Eftychia Kotronia
- Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Sheena E Ramsay
- Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
42
|
Premature Aging in Chronic Kidney Disease: The Outcome of Persistent Inflammation beyond the Bounds. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18158044. [PMID: 34360333 PMCID: PMC8345753 DOI: 10.3390/ijerph18158044] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023]
Abstract
Over the last hundred years, life expectancy in developed countries has increased because of healthier living habits and the treatment of chronic pathologies causing premature aging. Aging is an inexorable, time-dependent, multifactorial process characterized by a series of progressive and irreversible physiological changes associated with loss of functional, psychological, and social capabilities. Numerous factors, such as oxidative stress, inflammation, and cellular senescence, and an irreversible geriatric syndrome known as frailty, contribute to human body deterioration in aging. The speed of aging may differ between individuals depending on the presence or absence of multiple factors (genetic and/or environment) and the subsequent misbalance of homeostasis, together with the increase of frailty, which also plays a key role in developing chronic diseases. In addition, pathological circumstances have been reported to precipitate or accelerate the aging process. This review investigated the mechanisms involved in the developing pathologies, particularly chronic kidney disease, associated with aging.
Collapse
|
43
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
44
|
Leal-Alegre G, Lerma C, Leal-Escobar G, Moguel-González B, Martínez-Vázquez KB, Cano-Escobar KB. Relationship between Vascular Calcification, Protein-Energy Wasting Syndrome, and Sarcopenia in Maintenance Automated Peritoneal Dialysis. Life (Basel) 2021; 11:666. [PMID: 34357038 PMCID: PMC8306624 DOI: 10.3390/life11070666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/28/2021] [Accepted: 07/03/2021] [Indexed: 01/29/2023] Open
Abstract
Vascular calcifications affect 80% to 90% of chronic kidney disease patients and are a predictive factor of cardiovascular mortality. Sarcopenia and protein-energy wasting syndrome are also associated with mortality. The aim was to assess the relationship between vascular calcification, sarcopenia, and protein-energy wasting syndrome (PEW) in automated peritoneal dialysis patients. Fifty-one maintenance automated peritoneal dialysis patients were included (27 were male, mean age 39 ± 14 years). Vascular calcification was assessed based on abdomen, pelvis, and hand radiographs. Sarcopenia was assessed with bioimpedance analysis and a hand grip strength test. The Malnutrition-Inflammation Score and the presence of PEW were also assessed. Vascular calcification was present in 21 patients (41.2%). Univariate logistic regression analysis showed that age (p = 0.001), Malnutrition-Inflammation Score (p = 0.022), PEW (p = 0.049), sarcopenia (p = 0.048), and diabetes (p = 0.010) were associated with vascular calcification. Multivariate logistic regression analysis showed that age (p = 0.006) was the only variable associated independently with vascular calcification. In conclusion, there is association between vascular calcification, PEW, and sarcopenia in patients with maintenance automated peritoneal dialysis. These associations are not independent of age. This demonstrates the importance of nutritional status in the prevention of vascular calcification.
Collapse
Affiliation(s)
- Gustavo Leal-Alegre
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico; (G.L.-A.); (G.L.-E.); (B.M.-G.); (K.B.M.-V.)
| | - Claudia Lerma
- Department of Electromechanical Instrumentation, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico or (C.L.)
| | - Gabriela Leal-Escobar
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico; (G.L.-A.); (G.L.-E.); (B.M.-G.); (K.B.M.-V.)
| | - Bernardo Moguel-González
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico; (G.L.-A.); (G.L.-E.); (B.M.-G.); (K.B.M.-V.)
| | - Karen Belén Martínez-Vázquez
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico; (G.L.-A.); (G.L.-E.); (B.M.-G.); (K.B.M.-V.)
| | - Karla Berenice Cano-Escobar
- Department of Electromechanical Instrumentation, Instituto Nacional de Cardiología Ignacio Chávez, Mexico 04480, Mexico or (C.L.)
| |
Collapse
|
45
|
Higgins CE, Tang J, Higgins SP, Gifford CC, Mian BM, Jones DM, Zhang W, Costello A, Conti DJ, Samarakoon R, Higgins PJ. The Genomic Response to TGF-β1 Dictates Failed Repair and Progression of Fibrotic Disease in the Obstructed Kidney. Front Cell Dev Biol 2021; 9:678524. [PMID: 34277620 PMCID: PMC8284093 DOI: 10.3389/fcell.2021.678524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis is a common and diagnostic hallmark of a spectrum of chronic renal disorders. While the etiology varies as to the causative nature of the underlying pathology, persistent TGF-β1 signaling drives the relentless progression of renal fibrotic disease. TGF-β1 orchestrates the multifaceted program of kidney fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery or re-differentiation, capillary collapse and subsequent interstitial fibrosis eventually leading to chronic and ultimately end-stage disease. An increasing complement of non-canonical elements function as co-factors in TGF-β1 signaling. p53 is a particularly prominent transcriptional co-regulator of several TGF-β1 fibrotic-response genes by complexing with TGF-β1 receptor-activated SMADs. This cooperative p53/TGF-β1 genomic cluster includes genes involved in cellular proliferative control, survival, apoptosis, senescence, and ECM remodeling. While the molecular basis for this co-dependency remains to be determined, a subset of TGF-β1-regulated genes possess both p53- and SMAD-binding motifs. Increases in p53 expression and phosphorylation, moreover, are evident in various forms of renal injury as well as kidney allograft rejection. Targeted reduction of p53 levels by pharmacologic and genetic approaches attenuates expression of the involved genes and mitigates the fibrotic response confirming a key role for p53 in renal disorders. This review focuses on mechanisms underlying TGF-β1-induced renal fibrosis largely in the context of ureteral obstruction, which mimics the pathophysiology of pediatric unilateral ureteropelvic junction obstruction, and the role of p53 as a transcriptional regulator within the TGF-β1 repertoire of fibrosis-promoting genes.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - David M. Jones
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| |
Collapse
|
46
|
Chao CT, Lin SH. Uremic Toxins and Frailty in Patients with Chronic Kidney Disease: A Molecular Insight. Int J Mol Sci 2021; 22:ijms22126270. [PMID: 34200937 PMCID: PMC8230495 DOI: 10.3390/ijms22126270] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
The accumulation of uremic toxins (UTs) is a prototypical manifestation of uremic milieu that follows renal function decline (chronic kidney disease, CKD). Frailty as a potential outcome-relevant indicator is also prevalent in CKD. The intertwined relationship between uremic toxins, including small/large solutes (phosphate, asymmetric dimethylarginine) and protein-bound ones like indoxyl sulfate (IS) and p-cresyl sulfate (pCS), and frailty pathogenesis has been documented recently. Uremic toxins were shown in vitro and in vivo to induce noxious effects on many organ systems and likely influenced frailty development through their effects on multiple preceding events and companions of frailty, such as sarcopenia/muscle wasting, cognitive impairment/cognitive frailty, osteoporosis/osteodystrophy, vascular calcification, and cardiopulmonary deconditioning. These organ-specific effects may be mediated through different molecular mechanisms or signal pathways such as peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), mitogen-activated protein kinase (MAPK) signaling, aryl hydrocarbon receptor (AhR)/nuclear factor-κB (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), Runt-related transcription factor 2 (RUNX2), bone morphogenic protein 2 (BMP2), osterix, Notch signaling, autophagy effectors, microRNAs, and reactive oxygen species induction. Anecdotal clinical studies also suggest that frailty may further accelerate renal function decline, thereby augmenting the accumulation of UTs in affected individuals. Judging from these threads of evidence, management strategies aiming for uremic toxin reduction may be a promising approach for frailty amelioration in patients with CKD. Uremic toxin lowering strategies may bear the potential of improving patients’ outcomes and restoring their quality of life, through frailty attenuation. Pathogenic molecule-targeted therapeutics potentially disconnect the association between uremic toxins and frailty, additionally serving as an outcome-modifying approach in the future.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100255, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Shih-Hua Lin
- Nephrology Division, Department of Internal Medicine, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: or
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW This review examines the current knowledge and recent developments in the field of vascular calcification focusing on the emerging role of senescence and inflammation in driving this disorder and exploring the overlap and relevance of these pathways to calcinosis in rheumatic disease. RECENT FINDINGS Vascular calcification is an age-associated disorder. Recent studies have identified DNA damage, cellular senescence and consequent inflammation as key drivers of vascular smooth muscle cell osteogenic change and mineralization. Similar ageing and inflammatory factors are associated with calcinosis in rheumatic disease and some are targets of experimental drugs currently undergoing clinical trials. SUMMARY Calcinosis in the vascular system and in rheumatic disease share similarities in terms of biomineralization and cardiovascular outcomes. Although research into the role of senescence and inflammation has recently been advanced in vascular calcification, little is known about the mechanistic role of inflammation in calcinosis in rheumatic disease. This review explores whether lessons from one calcinosis can be transferred and applied to the other to provide further insights and inform treatment strategies.
Collapse
|
48
|
Calciphylaxis in end-stage kidney disease: outcome data from the United Kingdom Calciphylaxis Study. J Nephrol 2021; 34:1537-1545. [PMID: 33548054 PMCID: PMC8494680 DOI: 10.1007/s40620-020-00908-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/10/2020] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Calciphylaxis is a rare condition associated with very high mortality in patients with end-stage kidney disease. Data from country-based registries have been an invaluable resource for a better understanding of the natural history and management for this condition. This study aimed to investigate the current management strategies and outcomes of patients enrolled in the United Kingdom Calciphylaxis study (UKCS). METHODS The study was conducted on 89 patients registered in the UKCS since 2012. The initial analysis included a description of the baseline characteristics, management strategies and outcomes on follow-up until May 2020. Further analysis included a comparison of the mortality outcome of the UKCS patients who were receiving haemodialysis with a propensity score matched cohort of haemodialysis patients from the Chronic Renal Insufficiency Standards Implementation Study- Haemodialysis (CRISIS-HD). RESULTS Median age of the cohort was 59 years, with a predominance of females (61%) and Caucasian (95%) ethnicity. About 54% of the patients were diabetic and 70% were receiving haemodialysis at study entry. The skin lesions were mostly distributed in the lower extremities (48%). Sodium thiosulphate and calcimimetic were the most widely used management strategies. The mortality rate was 72 deaths per hundred patient-years (50 deaths observed in 69.5 patient years). Complete wound healing was noted in 17% and bacteraemia was reported in 26% of patients. In a comparative analysis of the matched haemodialysis patients, the presence of calciphylaxis in 62 patients showed a strong association with all-cause mortality (HR 6.96; p < 0.001), with annual mortality 67% versus 10.2% in haemodialysis patients without calciphylaxis. CONCLUSIONS This UK wide study strengthens the evidence that calciphylaxis is a strong and independent risk factor associated with all-cause mortality; no significant benefit was shown with any individual treatment modality. Until further evidence becomes available, a multifaceted approach would be the appropriate treatment strategy in the management of this extremely serious condition.
Collapse
|
49
|
Papacocea RI, Timofte D, Tanasescu MD, Balcangiu-Stroescu AE, Balan DG, Tulin A, Stiru O, Vacaroiu IA, Mihai A, Popa CC, Cosconel CI, Enyedi M, Miricescu D, Raducu L, Ionescu D. Kidney aging process and the management of the elderly patient with renal impairment (Review). Exp Ther Med 2021; 21:266. [PMID: 33603873 PMCID: PMC7851660 DOI: 10.3892/etm.2021.9697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
As life expectancy increases, a rise in the number of chronically ill patients is observed due to the aging population. Among the various diseases, chronic kidney disease is at present one of the main causes of morbidity and, due to its typical complications, it is also one of the most important causes of mortality in the general population. For these reasons, the understanding of the kidney aging process, its consequences and its adequate management are essential. The judicious use of certain types of drugs, the prevention of episodes of renal injury either by toxic mechanisms or by dehydration are important aspects and are part of the apropriate approach for elderly patients. The most effective treatment of various types of conditions with a negative impact on renal function and for which an increased incidence is known as we age should also be considered. Thus, in the case of elderly patients, in order to protect the kidneys, an integrative approach is recommended, one that includes both elements of prevention and the appropriate treatment of existing diseases.
Collapse
Affiliation(s)
- Raluca Ioana Papacocea
- Discipline of Physiology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Delia Timofte
- Department of Dialysis, Emergency University Hospital, 050098 Bucharest, Romania
| | - Maria-Daniela Tanasescu
- Department of Medical Semiology, Discipline of Internal Medicine I and Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Nephrology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Andra-Elena Balcangiu-Stroescu
- Department of Dialysis, Emergency University Hospital, 050098 Bucharest, Romania.,Discipline of Physiology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Gabriela Balan
- Discipline of Physiology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Adrian Tulin
- Department of Anatomy, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of General Surgery, 'Prof. Dr. Agrippa Ionescu' Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Ovidiu Stiru
- Department of Cardiovascular Surgery, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Cardiovascular Surgery, 'Prof. Dr. C.C. Iliescu' Emergency Institute for Cardiovascular Diseases, 022322 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology and Dialysis, St. John Emergency Clinical Hospital, 042122 Bucharest, Romania.,Department of Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Romania
| | - Andrada Mihai
- Discipline of Diabetes, Nutrition and Metabolic Diseases-N. Paulescu National Institute, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department II of Diabetes, Prof. N. Paulescu, Bucharest, Nutrition and Metabolic Diseases National Institute of Diabetes, Nutrition and Metabolic Disease, 020474 Bucharest, Romania
| | - Cristian Constantin Popa
- Department of Surgery, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Surgery, Emergency University Hospital, 050098 Bucharest, Romania
| | - Cristina-Ileana Cosconel
- Discipline of Foreign Languages, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihaly Enyedi
- Department of Anatomy, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Radiology, 'Victor Babes' Private Medical Clinic, 030303 Bucharest, Romania
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Raducu
- Discipline of Plastic and Reconstructive Surgery, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Plastic and Reconstructive Surgery 'Prof. Dr. Agrippa Ionescu' Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Dorin Ionescu
- Department of Dialysis, Emergency University Hospital, 050098 Bucharest, Romania.,Department of Medical Semiology, Discipline of Internal Medicine I and Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
50
|
Wang Y, Wang Y, Yang M, Ma X. Implication of cellular senescence in the progression of chronic kidney disease and the treatment potencies. Biomed Pharmacother 2021; 135:111191. [PMID: 33418306 DOI: 10.1016/j.biopha.2020.111191] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is an increasing major public health problem worldwide. And CKD shares numerous phenotypic similarities with kidney as well as systemic ageing. Cellular senescence is mainly characterized by a stable cell cycle arrest, senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Herein, the regulations and the internal mechanisms of cellular senescence will be discussed. Meanwhile, efforts are made to give a comprehensive overview of the recent advances of the implication of cellular senescence in CKD. To date, numerous studies have focused on the effects of ageing risk factors in kidney and thereby trying to interrupt the kidney ageing processes with senolytics. Interestingly, some of them showed enormous clinical application potentials. Therefore, senotherapeutics can be applied as novel potential strategies for the treatment of CKD.
Collapse
Affiliation(s)
- Yao Wang
- Department of Nephrology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Wang
- Department of Endocrinology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ming Yang
- Department of Nephrology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xingjie Ma
- Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|