1
|
Goyal K, Afzal M, Altamimi ASA, Babu MA, Ballal S, Kaur I, Kumar S, Kumar MR, Chauhan AS, Ali H, Shahwan M, Gupta G. Chronic kidney disease and aging: dissecting the p53/p21 pathway as a therapeutic target. Biogerontology 2024; 26:32. [PMID: 39725742 DOI: 10.1007/s10522-024-10173-z] [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: 11/09/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney diseases (CKD) are a group of multi-factorial disorders that markedly impair kidney functions with progressive renal deterioration. Aging contributes to age-specific phenotypes in kidneys, which undergo several structural and functional alterations, such as a decline in regenerative capacity and increased fibrosis, inflammation, and tubular atrophy, all predisposing them to disease and increasing their susceptibility to injury while impeding their recovery. A central feature of these age-related processes is the activation of the p53/p21 pathway signaling. The pathway is a key player in cellular senescence, apoptosis, and cell cycle regulation, which are all key to maintaining the health of the kidney. P53 is a transcription factor and a tumor suppressor protein that responds to cell stress and damage. Persistent activation of cell p53 can lead to the expression of p21, an inhibitor of the cell cycle known as a cyclin-dependent kinase. This causes cells to cease dividing and leads to senescence, where cells can no longer increase. The accumulation of senescent cells in the aging kidney impairs kidney function by altering the microenvironment. As the number of senescent cells increases, the capacity of the kidney to recover from injury decreases, accelerating the progression of end-stage renal disease. This article review extensively explores the relationship between the p53/p21 pathway and cellular senescence within an aging kidney and the emerging therapeutic strategies that target it to overcome the impacts of cellular senescence on CKD.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, 140307, India
| | - Ashish Singh Chauhan
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Haider Ali
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
2
|
Beridze G, Dai L, Carrero JJ, Marengoni A, Vetrano DL, Calderón-Larrañaga A. Associations between multimorbidity and kidney function decline in old age: A population-based cohort study. J Am Geriatr Soc 2024. [PMID: 39690840 DOI: 10.1111/jgs.19298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/07/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Individual chronic conditions have been linked to kidney function decline; however, the role of multimorbidity (the presence of ≥2 conditions) and multimorbidity patterns remains unclear. METHODS A total of 3094 individuals from the Swedish National study on Aging and Care in Kungsholmen (SNAC-K) were followed for 15 years. Multimorbidity was operationalized as the number of chronic conditions and multimorbidity patterns identified using latent class analysis (LCA). Joint models and Cox regression models were used to explore the associations between multimorbidity, and subsequent absolute and relative (≥25% decline from baseline) changes, respectively, in the estimated glomerular filtration rate (eGFR) calculated using the creatinine-based Berlin Initiative Study equation. RESULTS Mean age of the sample was 73.9, and 87% had multimorbidity. There was an independent dose-response relationship between the number of chronic conditions, and absolute (β [95% confidence interval, CI] = -0.05 [-0.07; -0.03]) and relative (hazard ratio, HR [95% CI] = 1.23 [1.17; 1.29]) declines in eGFR. Five patterns of multimorbidity were identified. The Unspecific, low burden pattern had the lowest morbidity burden and was used as the reference category. The Unspecific, high burden, and Cardiometabolic patterns showed accelerated absolute (β [95% CI] = -0.15 [-0.26; -0.05] and -0.77 [-0.98; -0.55], respectively) and relative (HR [95% CI] = 1.45 [1.09; 1.92] and 3.45 [2.27; 5.23], respectively) declines. Additionally, the Cognitive and Sensory pattern showed accelerated relative decline (HR [95% CI] = 1.53 [1.02; 2.31]). No associations were found for the Psychiatric and Respiratory pattern. CONCLUSION Multimorbidity is strongly associated with accelerated kidney function decline in older age. Individuals with cardiometabolic multimorbidity exhibit a particularly increased risk. Increased monitoring and timely interventions may preserve kidney function and reduce cardiovascular risks in individuals presenting with conditions that are characteristic of high-risk multimorbidity patterns.
Collapse
Affiliation(s)
- Giorgi Beridze
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Lu Dai
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Juan-Jesús Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alessandra Marengoni
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Davide L Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| | - Amaia Calderón-Larrañaga
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| |
Collapse
|
3
|
Athish KK, Nayak Rao S, Marimuthu VH, Krishna V, Arun A. Tenofovir-Associated Kidney Dysfunction and Bone Fracture: A Case Report and Literature Review. Cureus 2024; 16:e61562. [PMID: 38962632 PMCID: PMC11220731 DOI: 10.7759/cureus.61562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2024] [Indexed: 07/05/2024] Open
Abstract
Tenofovir is an integral part of antiretroviral therapy used to treat HIV. Long-term use of tenofovir has been associated with decreased glomerular filtration rate, leading to chronic kidney disease, as well as acidosis, electrolyte imbalances, and tubular dysfunction. Tenofovir can also disrupt bone health by decreasing renal phosphate absorption, contributing to osteomalacia. This leads to disruption in mineral metabolism, elevated parathyroid hormone levels, and ultimately, low bone mineral density. Replacing tenofovir with alternative antiretroviral therapy can improve kidney function if done early in the course of the disease. Here, we discuss a case of a 65-year-old woman with HIV who presented with advanced renal failure and hypophosphatemia-induced bone fracture attributed to long-term use of tenofovir. We conclude monitoring kidney function and considering alternative antiretroviral therapy is important to prevent and manage these side effects in patients on long-term tenofovir therapy.
Collapse
Affiliation(s)
- K K Athish
- Department of Internal Medicine, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | - Shobhana Nayak Rao
- Department of Nephrology, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | - Venkat H Marimuthu
- Department of Emergency Medicine, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | - Vamsi Krishna
- Department of Internal Medicine, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | - Anwadevi Arun
- Department of Internal Medicine, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| |
Collapse
|
4
|
Nauffal V, Klarqvist MDR, Hill MC, Pace DF, Di Achille P, Choi SH, Rämö JT, Pirruccello JP, Singh P, Kany S, Hou C, Ng K, Philippakis AA, Batra P, Lubitz SA, Ellinor PT. Noninvasive assessment of organ-specific and shared pathways in multi-organ fibrosis using T1 mapping. Nat Med 2024; 30:1749-1760. [PMID: 38806679 DOI: 10.1038/s41591-024-03010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fibrotic diseases affect multiple organs and are associated with morbidity and mortality. To examine organ-specific and shared biologic mechanisms that underlie fibrosis in different organs, we developed machine learning models to quantify T1 time, a marker of interstitial fibrosis, in the liver, pancreas, heart and kidney among 43,881 UK Biobank participants who underwent magnetic resonance imaging. In phenome-wide association analyses, we demonstrate the association of increased organ-specific T1 time, reflecting increased interstitial fibrosis, with prevalent diseases across multiple organ systems. In genome-wide association analyses, we identified 27, 18, 11 and 10 independent genetic loci associated with liver, pancreas, myocardial and renal cortex T1 time, respectively. There was a modest genetic correlation between the examined organs. Several loci overlapped across the examined organs implicating genes involved in a myriad of biologic pathways including metal ion transport (SLC39A8, HFE and TMPRSS6), glucose metabolism (PCK2), blood group antigens (ABO and FUT2), immune function (BANK1 and PPP3CA), inflammation (NFKB1) and mitosis (CENPE). Finally, we found that an increasing number of organs with T1 time falling in the top quintile was associated with increased mortality in the population. Individuals with a high burden of fibrosis in ≥3 organs had a 3-fold increase in mortality compared to those with a low burden of fibrosis across all examined organs in multivariable-adjusted analysis (hazard ratio = 3.31, 95% confidence interval 1.77-6.19; P = 1.78 × 10-4). By leveraging machine learning to quantify T1 time across multiple organs at scale, we uncovered new organ-specific and shared biologic pathways underlying fibrosis that may provide therapeutic targets.
Collapse
Affiliation(s)
- Victor Nauffal
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Danielle F Pace
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cody Hou
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | - Anthony A Philippakis
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Lubitz
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
5
|
Patel L, Roy A, Alvior AMB, Yuan M, Baig S, Bunting KV, Hodson J, Gehmlich K, Lord JM, Geberhiwot T, Steeds RP. Phenoage and longitudinal changes on transthoracic echocardiography in Alström syndrome: a disease of accelerated ageing? GeroScience 2024; 46:1989-1999. [PMID: 37782438 PMCID: PMC10828353 DOI: 10.1007/s11357-023-00959-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023] Open
Abstract
Alström syndrome (AS) is an ultra-rare disorder characterised by early-onset multi-organ dysfunction, such as insulin resistance, obesity, dyslipidaemia, and renal and cardiovascular disease. The objective is to explore whether AS is a disease of accelerated ageing and whether changes over time on echocardiography could reflect accelerated cardiac ageing. Cross-sectional measurement of Phenoage and retrospective analysis of serial echocardiography were performed between March 2012 and November 2022. The setting is a single national tertiary service jointly run by health service and patient charity. Forty-five adult patients aged over 16 years were included, 64% were male and 67% of White ethnicity. The median Phenoage was 48 years (interquartile range [IQR]: 35-72) in the 34 patients for whom this was calculable, which was significantly higher than the median chronological age of 29 years (IQR: 22-39, p<0.001). Phenoage was higher than chronological age in 85% (N=29) of patients, with a median difference of +18 years (IQR: +4, +34). On echocardiography, significant decreases were observed over time in left ventricular (LV) size at end-diastole (average of 0.046 cm per year, p<0.001) and end-systole (1.1% per year, p=0.025), with significant increase in posterior wall thickness at end-diastole (0.009 cm per year, p=0.008). LV systolic function measured by global longitudinal strain reduced (0.34 percentage points per year, p=0.020) and E/e'lat increased (2.5% per year, p=0.019). Most AS patients display a higher Phenoage compared to chronological age. Cardiac changes in AS patients were also reflective of accelerated ageing, with a reduction in LV size and increased wall thickening. AS may be a paradigm disease for premature ageing.
Collapse
Affiliation(s)
- Leena Patel
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Ashwin Roy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK.
| | - Amor Mia B Alvior
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Mengshi Yuan
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Shanat Baig
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Karina V Bunting
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - James Hodson
- Research Development and Innovation, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Janet M Lord
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Tarekegn Geberhiwot
- Department of Endocrinology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, UK
| | - Richard P Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| |
Collapse
|
6
|
Sheshadri A, Lai M, Hsu FC, Bauer SR, Chen SH, Tse W, Jotwani V, Tranah GJ, Lai JC, Hallan S, Fielding RA, Liu C, Ix JH, Coca SG, Shlipak MG. Structured Moderate Exercise and Biomarkers of Kidney Health in Sedentary Older Adults: The Lifestyle Interventions and Independence for Elders Randomized Clinical Trial. Kidney Med 2023; 5:100721. [PMID: 37915963 PMCID: PMC10616412 DOI: 10.1016/j.xkme.2023.100721] [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] [Indexed: 11/03/2023] Open
Abstract
Rationale & Objective In the Lifestyle Interventions and Independence for Elders (LIFE) trial, a structured exercise intervention slowed kidney function decline in sedentary older adults. Biomarkers of kidney health could distinguish potential mechanisms for this beneficial effect. Study Design Randomized controlled trial. Setting & Population A total of 1,381 sedentary adults aged 70-89 years enrolled in the LIFE trial. Intervention Structured, 2-year, moderate-intensity exercise intervention versus health education. Outcomes Physical activity was measured by step count. Primary outcomes were changes in 14 serum and urine biomarkers of kidney health collected at baseline, year 1, and year 2. We determined the effect of randomization on changes in kidney measures and then evaluated observational associations of achieved activity on each measure. Results Participants assigned to exercise walked on average 291 more steps per day than participants assigned to health education. The intervention was not significantly associated with changes in biomarkers of kidney health. In observational analyses, persons in the highest versus lowest quartile of activity (≥3,470 vs <1,568 steps/day) had significant improvement in urine albumin (mean, -0.22 mg albumin/g urine creatinine [interquartile range (IQR), -0.37 to -0.06]), alpha-1-microglobulin (-0.18 mg/L [-0.28 to -0.08]), trefoil factor-3 (-0.24 pg/mL [-0.35 to -0.13]), epidermal growth factor (0.19 pg/mL [0.06-0.32]), uromodulin (0.06 pg/mL [0.00-0.12]), interleukin 18 (-0.09 pg/mL [-0.15 to -0.03]), neutrophil gelatinase-associated lipocalin (-0.16 pg/mL [-0.24 to -0.07]), monocyte chemoattractant protein-1 (-0.25 pg/mL [-0.36 to -0.14]), clusterin (-0.16 pg/mL [-0.30 to -0.02]), serum tumor necrosis factor receptor-1 (-0.25 mg/dL [-0.39 to -0.11]) and tumor necrosis factor receptor-2 (-0.30 mg/dL [-0.44 to -0.16]). In sensitivity analyses, incremental changes in activity were most impactful on urine interleukin 18 and serum tumor necrosis factor-1. Limitations The original study was not designed to assess the impact on kidney health. Non-white individuals and patients with advanced chronic kidney disease are underrepresented. Conclusions Randomization to structured exercise did not improve kidney health at a group level. However, higher exercise was associated with concurrent improvements in biomarkers of glomerular injury, tubular function/repair, tubular injury, generalized inflammation, and tubulointerstitial repair/fibrosis. Plain-Language Summary In the Lifestyle Interventions For Elders (LIFE) study, randomization to an exercise and physical activity intervention improved the slope of estimated glomerular filtration rate over 2 years compared with health education among older adults. In this study, we sought to determine whether there were specific biomarkers of kidney health that were affected by the exercise and physical activity intervention to investigate potential mechanisms for this positive impact on kidney decline. We found that randomization to the intervention did not improve any of the 14 measures of kidney tubule health. However, in observational analyses, higher activity was independently associated with improvements in several domains, especially tubular injury and generalized inflammation. These results help to clarify the impact of physical activity on kidney health.
Collapse
Affiliation(s)
- Anoop Sheshadri
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA
| | - Mason Lai
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Scott R. Bauer
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA
| | - Shyh-Huei Chen
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Warren Tse
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA
| | - Vasantha Jotwani
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA
| | | | - Jennifer C. Lai
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Roger A. Fielding
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Christine Liu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
- Geriatric Research Education and Clinical Center, Palo Alto VA Health Care System, Palo Alto, CA
| | - Joachim H. Ix
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Steven G. Coca
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael G. Shlipak
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA
| |
Collapse
|
7
|
Perazza LR, Gower AC, Brown-Borg HM, Pajevic PD, Thompson LV. Protectin DX as a therapeutic strategy against frailty in mice. GeroScience 2023; 45:2601-2627. [PMID: 37059838 PMCID: PMC10651819 DOI: 10.1007/s11357-023-00789-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023] Open
Abstract
Frailty in aging is driven by the dysregulation of multiple biological pathways. Protectin DX (PDX) is a docosahexaenoic acid (DHA)-derived molecule that alleviates many chronic inflammatory disorders, but its potential effects on frailty remain unknown. Our goal is to identify age-related impairments in metabolic systems and to evaluate the therapeutic potential of PDX on frailty, physical performance, and health parameters. A set of 22-month-old C57BL/6 male and female mice were assigned to vehicle (Old) or PDX daily gavage treatment for 9 weeks, whereas 6-month-old (Adult) mice received only vehicle. Forelimb and hindlimb strength, endurance, voluntary wheel activity and walking speed determined physical performance and were combined with a frailty index score and body weight loss to determine frailty status. Our data shows that old vehicle-treated mice from both sexes had body weight loss paralleling visceromegaly, and Old females also had impaired insulin clearance as compared to the Adult group. Aging was associated with physical performance decline together with higher odds of frailty development. There was also age-driven mesangial expansion and glomerular hypertrophy as well as bone mineral density loss. All of the in vivo and in vitro impairments observed with aging co-occurred with upregulation of inflammatory pathways and Myc signaling as well as downregulation of genes related to adipogenesis and oxidative phosphorylation in liver. PDX attenuated the age-driven physical performance (strength, exhaustion, walking speed) decline, promoted robustness, prevented bone losses and partially reversed changes in hepatic expression of Myc targets and metabolic genes. In conclusion, our data provides evidence of the beneficial therapeutic effect of PDX against features of frailty in mice. Further studies are warranted to investigate the mechanisms of action and the potential for human translation.
Collapse
Affiliation(s)
- Laís R Perazza
- Department of Physical Therapy, Boston University, Boston, MA, USA.
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University, Boston, MA, USA
| | - Holly M Brown-Borg
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Paola Divieti Pajevic
- Department of Translational Dental Medicine, Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | | |
Collapse
|
8
|
Luo P, Zhang H, Liang Y, Li X, Wen Z, Xia C, Lan X, Yang Y, Xiong Y, Huang J, Ling X, Zhou S, Miao J, Shen W, Hou FF, Liu Y, Zhou L, Liang M. Pentraxin 3 plays a key role in tubular cell senescence and renal fibrosis through inducing β-catenin signaling. Biochim Biophys Acta Mol Basis Dis 2023:166807. [PMID: 37453582 DOI: 10.1016/j.bbadis.2023.166807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Renal fibrosis is the common pathological feature of various chronic kidney diseases (CKD). Tubular cell senescence plays a key role in the progression of renal fibrosis. However, the underlying mechanisms are still in mystery. In this study, we identified, Pentraxin 3 (PTX3), belonging to the Pentraxin family, is a new fibrogenic factor. PTX3 was increased in various CKD models. PTX3 was primarily localized in tubular epithelial cells and upregulated, accompanied by mitochondrial dysfunction and cellular senescence. Overexpression of PTX3 aggravated mitochondrial damage and accelerated cell senescence in tubular cells, leading to more severe fibrogenesis in kidneys. However, knockout of PTX3 significantly preserved mitochondrial homeostasis, and blocked cellular senescence in primary cultured tubular cells. Furthermore, KYA1797K, a destabilizer of β-catenin, greatly inhibited PTX3-induced mitochondrial homeostasis, tubular cell senescence, and renal fibrosis. Overexpression of PTX3 triggered nuclear translocation of β-catenin, an activating form of β-catenin. PTX3-induced mitochondrial dysfunction and tubular cell senescence were also significantly inhibited by knockdown of p16INK4A, a senescence-related protein. In a clinical cohort, we found PTX3 was increased in urine and serum in clinical patients with CKD. Urinary PTX3 negatively correlated with the decline of eGFR. PTX3 also increased gradually following the severity of diseases, triggering the fibrogenesis. Taken together, our results provide strong evidences that PTX3 is a new fibrogenic factor in the development of renal fibrosis through β-catenin-induced mitochondrial dysfunction and cell senescence. This study further suggests PTX3 is a new diagnostic factor to renal fibrosis and provides a new therapeutic target against renal fibrosis.
Collapse
Affiliation(s)
- Pei Luo
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Haixia Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Ye Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Xiaolong Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Zhen Wen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Chaoying Xia
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Xiaolei Lan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yaya Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yabing Xiong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jiewu Huang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Xian Ling
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Shan Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jinhua Miao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Weiwei Shen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China.
| | - Min Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China.
| |
Collapse
|
9
|
Yu X, Wang Y, Song Y, Gao X, Deng H. AP-1 is a regulatory transcription factor of inflammaging in the murine kidney and liver. Aging Cell 2023; 22:e13858. [PMID: 37154113 PMCID: PMC10352569 DOI: 10.1111/acel.13858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/07/2023] [Indexed: 05/10/2023] Open
Abstract
Aging is characterized by chronic low-grade inflammation in multiple tissues, also termed "inflammaging", which represents a significant risk factor for many aging-related chronic diseases. However, the mechanisms and regulatory networks underlying inflammaging across different tissues have not yet been fully elucidated. Here, we profiled the transcriptomes and epigenomes of the kidney and liver from young and aged mice and found that activation of the inflammatory response is a conserved signature in both tissues. Moreover, we revealed links between transcriptome changes and chromatin dynamics through integrative analysis and identified AP-1 and ETS family transcription factors (TFs) as potential regulators of inflammaging. Further in situ validation showed that c-JUN (a member of the AP-1 family) was mainly activated in aged renal and hepatic cells, while increased SPI1 (a member of the ETS family) was mostly induced by elevated infiltration of macrophages, indicating that these TFs have different mechanisms in inflammaging. Functional data demonstrated that genetic knockdown of Fos, a major member of the AP-1 family, significantly attenuated the inflammatory response in aged kidneys and livers. Taken together, our results revealed conserved signatures and regulatory TFs of inflammaging in the kidney and liver, providing novel targets for the development of anti-aging interventions.
Collapse
Affiliation(s)
- Xiaojie Yu
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Yuting Wang
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Yifan Song
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Xianda Gao
- School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - Hongkui Deng
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| |
Collapse
|
10
|
Long-Term Evaluation of Changes in Kidney Function after Switching from Tenofovir Disoproxil Fumarate to Tenofovir Alafenamide in Patients Living with HIV. PHARMACY 2022; 10:pharmacy10060164. [PMID: 36548320 PMCID: PMC9781640 DOI: 10.3390/pharmacy10060164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Tenofovir is one of the most widely used medications for HIV treatment and is administered as either tenofovir disoproxil fumarate (TDF) or tenofovir alafenamide (TAF). Use of TAF is preferred as it is associated with fewer negative impacts on renal function; however, long-term follow-up beyond 96 weeks is limited. A retrospective chart review of patients ≥18 years who received TDF-containing anti-retroviral therapy (ART) for ≥6 months and then switched to a TAF-containing regimen between 1 December 2015 and 1 January 2020 is presented. The primary objective was to evaluate changes in kidney function as measured by eGFR and Scr. The secondary objective was to evaluate changes in lipids. Among the 142 patients identified, the median age was 66 years old with a median follow-up of 3.6 years. The change in kidney function was a median increase in Scr of 0.1 mg/dL and a decrease in eGFR of -8 mL/min/1.73 m2. The change in lipid panels at the end of the medication use evaluation endpoint was a decrease in total cholesterol, LDL, HDL, and triglycerides of -2.5, -0.1, -0.6, and -9 mmol/L, respectively. There was no clinically meaningful difference in kidney function as measured by eGFR or Scr, nor was there any clinically meaningful difference in lipid panels in patients switched from TDF to TAF-containing ART. Our observations suggest that the favorable impact of TAF on kidney function is sustained for at least 44 months after conversion from TDF.
Collapse
|
11
|
Liu F, Chen J, Li Z, Meng X. Recent Advances in Epigenetics of Age-Related Kidney Diseases. Genes (Basel) 2022; 13:genes13050796. [PMID: 35627181 PMCID: PMC9142069 DOI: 10.3390/genes13050796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/03/2023] Open
Abstract
Renal aging has attracted increasing attention in today’s aging society, as elderly people with advanced age are more susceptible to various kidney disorders such as acute kidney injury (AKI) and chronic kidney disease (CKD). There is no clear-cut universal mechanism for identifying age-related kidney diseases, and therefore, they pose a considerable medical and public health challenge. Epigenetics refers to the study of heritable modifications in the regulation of gene expression that do not require changes in the underlying genomic DNA sequence. A variety of epigenetic modifiers such as histone deacetylases (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors have been proposed as potential biomarkers and therapeutic targets in numerous fields including cardiovascular diseases, immune system disease, nervous system diseases, and neoplasms. Accumulating evidence in recent years indicates that epigenetic modifications have been implicated in renal aging. However, no previous systematic review has been performed to systematically generalize the relationship between epigenetics and age-related kidney diseases. In this review, we aim to summarize the recent advances in epigenetic mechanisms of age-related kidney diseases as well as discuss the application of epigenetic modifiers as potential biomarkers and therapeutic targets in the field of age-related kidney diseases. In summary, the main types of epigenetic processes including DNA methylation, histone modifications, non-coding RNA (ncRNA) modulation have all been implicated in the progression of age-related kidney diseases, and therapeutic targeting of these processes will yield novel therapeutic strategies for the prevention and/or treatment of age-related kidney diseases.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Zhenqiong Li
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Correspondence: (Z.L.); (X.M.)
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (Z.L.); (X.M.)
| |
Collapse
|
12
|
Arroyo E, Troutman AD, Moorthi RN, Avin KG, Coggan AR, Lim K. Klotho: An Emerging Factor With Ergogenic Potential. FRONTIERS IN REHABILITATION SCIENCES 2022; 2:807123. [PMID: 36188832 PMCID: PMC9397700 DOI: 10.3389/fresc.2021.807123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022]
Abstract
Sarcopenia and impaired cardiorespiratory fitness are commonly observed in older individuals and patients with chronic kidney disease (CKD). Declines in skeletal muscle function and aerobic capacity can progress into impaired physical function and inability to perform activities of daily living. Physical function is highly associated with important clinical outcomes such as hospitalization, functional independence, quality of life, and mortality. While lifestyle modifications such as exercise and dietary interventions have been shown to prevent and reverse declines in physical function, the utility of these treatment strategies is limited by poor widespread adoption and adherence due to a wide variety of both perceived and actual barriers to exercise. Therefore, identifying novel treatment targets to manage physical function decline is critically important. Klotho, a remarkable protein with powerful anti-aging properties has recently been investigated for its role in musculoskeletal health and physical function. Klotho is involved in several key processes that regulate skeletal muscle function, such as muscle regeneration, mitochondrial biogenesis, endothelial function, oxidative stress, and inflammation. This is particularly important for older adults and patients with CKD, which are known states of Klotho deficiency. Emerging data support the existence of Klotho-related benefits to exercise and for potential Klotho-based therapeutic interventions for the treatment of sarcopenia and its progression to physical disability. However, significant gaps in our understanding of Klotho must first be overcome before we can consider its potential ergogenic benefits. These advances will be critical to establish the optimal approach to future Klotho-based interventional trials and to determine if Klotho can regulate physical dysfunction.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ashley D. Troutman
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Ranjani N. Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Keith G. Avin
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Andrew R. Coggan
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
13
|
Wang X, Zu Q, Lu J, Zhang L, Zhu Q, Sun X, Dong J. Effects of Donor-Recipient Age Difference in Renal Transplantation, an Investigation on Renal Function and Fluid Proteome. Clin Interv Aging 2021; 16:1457-1470. [PMID: 34349505 PMCID: PMC8326938 DOI: 10.2147/cia.s314587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022] Open
Abstract
Introduction Our previous study revealed that a young internal environment ameliorated kidney aging by virtue of an animal model of heterochronic parabiosis and a model of heterochronic renal transplantation. In this research, we used proteome to investigate the effects of donor-recipient age difference in clinical renal transplantation. Methods This study included 10 pairs of renal transplantation donors and recipients with an age difference of greater than 20 years to their corresponding recipients/donors. All recipients have received transplantation more than 3 years ago. Renal function and the serum/urine proteomes of the donors and recipients were analyzed. Results The renal function was similar between the young recipients and the old donors. In contrast, the renal function of the young donors was significantly superior to that of the old recipients. Furthermore, 497 and 975 proteins were identified in the serum and urine proteomes, respectively. The content of SLC3A2 in the blood was found to be related to aging, while the contents of SERPINA1 and SERPINA3 in the urine were related to immune functions after renal transplantation. Conclusion This study demonstrated that, in the human body, a younger internal environment could ameliorate kidney aging and provided not only clinical evidence for increasing the age limit of kidney transplant donors but also new information for kidney aging research.
Collapse
Affiliation(s)
- Xinning Wang
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Qiang Zu
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jinshan Lu
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lei Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Qiang Zhu
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xuefeng Sun
- Department of Nephrology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jun Dong
- Department of Urology, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
14
|
Samarakoon R, Higgins PJ. Editorial: Premature Aging and Senescence in Renal Fibrosis. Front Pharmacol 2021; 12:734892. [PMID: 34381371 PMCID: PMC8352495 DOI: 10.3389/fphar.2021.734892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Rohan Samarakoon
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J Higgins
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
15
|
Campbell RA, Docherty MH, Ferenbach DA, Mylonas KJ. The Role of Ageing and Parenchymal Senescence on Macrophage Function and Fibrosis. Front Immunol 2021; 12:700790. [PMID: 34220864 PMCID: PMC8248495 DOI: 10.3389/fimmu.2021.700790] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
In this review, we examine senescent cells and the overlap between the direct biological impact of senescence and the indirect impact senescence has via its effects on other cell types, particularly the macrophage. The canonical roles of macrophages in cell clearance and in other physiological functions are discussed with reference to their functions in diseases of the kidney and other organs. We also explore the translational potential of different approaches based around the macrophage in future interventions to target senescent cells, with the goal of preventing or reversing pathologies driven or contributed to in part by senescent cell load in vivo.
Collapse
Affiliation(s)
- Ross A. Campbell
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie-Helena Docherty
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - David A. Ferenbach
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Katie J. Mylonas
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Genetic Background and Clinicopathologic Features of Adult-onset Nephronophthisis. Kidney Int Rep 2021; 6:1346-1354. [PMID: 34013113 PMCID: PMC8116764 DOI: 10.1016/j.ekir.2021.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction Recently, nephronophthisis (NPH) has been considered a monogenic cause of end-stage renal disease (ESRD) in adults. However, adult-onset NPH is difficult to accurately diagnose and has not been reported in a cohort study. In this study, we assessed the genetic background and clinicopathologic features of adult NPH. Methods We investigated 18 sporadic adult patients who were suspected as having NPH by renal biopsy. We analyzed 69 genes that cause hereditary cystic kidney disease and compared clinicopathologic findings between patients with and without pathogenic mutations in NPH-causing genes. Results Seven of 18 patients had pathogenic NPH-causing mutations in NPHP1, NPHP3, NPHP4, or CEP164. Compared with patients without pathogenic mutations, those with pathogenic mutations were significantly younger but did not significantly differ in the classic NPH pathologic findings, such as tubular cysts. On the other hand, the number of tubules with thick tubular basement membrane (TBM) duplication, which was defined as >10-μm thickness, was significantly higher in patients with genetically proven adult NPH than in those without pathogenic mutations. α-Smooth muscle actin (α-SMA)-positive myofibroblasts were detected inside thick TBM duplication. Conclusions In adult patients with NPH, thick TBM duplication was the specific finding. Our analysis also suggested that older patients tended to have no pathogenic mutations, even when they were suspected to have NPH by renal biopsy. These findings could be the novel clinical clue for the diagnosis of NPH in adult patients.
Collapse
|
17
|
Liu JR, Cai GY, Ning YC, Wang JC, Lv Y, Guo YN, Fu B, Hong Q, Sun XF, Chen XM. Caloric restriction alleviates aging-related fibrosis of kidney through downregulation of miR-21 in extracellular vesicles. Aging (Albany NY) 2020; 12:18052-18072. [PMID: 32963130 PMCID: PMC7585074 DOI: 10.18632/aging.103591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/27/2020] [Indexed: 01/24/2023]
Abstract
Glomerulosclerosis and renal interstitial fibrosis occur with the aging kidney. In this study, we examined the expression of miR-21, peroxisome proliferator-activated receptor(PPARα), hypoxia-inducible factor(HIF-1α) in the kidney of 3-month-old rats fed ad libitum (YAL), 24-month-old rats fed ad libitum (OAL) and 24-month-old rats subjected to a 70% calorie-restricted diet for 8 months (OCR). We found long-term caloric restriction (CR) ameliorated aging and aging-related fibrosis. CR ameliorated the increment of miR-21 and HIF-1α, as well as the decrement of PPARα in old ad libitum group. Human proximal tubular cells (HPTCs) presented phenotypes of senescence and epithelial to mesenchymal transition (EMT) under high-glucose conditions, in which senescence occurred earlier than EMT. Senescent cells secreted extracellular vesicles (EVs) which contained miR-21 into the recipient cells. Inhibiting miR-21 of donor cells prevented the occurrence of EMT in recipient cells. In addition, miR-21 induced EMT through targeting PPARα protein and consequently enhancing HIF-1α expression, although other pathways cannot be ruled out. These findings demonstrated that miR-21-containing EVs derived from the senescent cells could facilitate EMT of HPTCs via PPARα-HIF-1α signaling pathway. Long-term caloric restriction and caloric restriction mimetics alleviated aging-related-fibrosis of kidney through downregulation of miR-21.
Collapse
Affiliation(s)
- Jin-rui Liu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China,Renal Transplant Division, Department of Nephrology, Zhengzhou No. 7 People's Hospital, Zhengzhou 450017, Henan, China
| | - Guang-yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yi-chun Ning
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing-chao Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yang Lv
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Ya-nan Guo
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xue-feng Sun
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiang-mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
18
|
Elbaset MA, Zahran MH, Elrefaie E, Elgamal M, M.A. S, Ezzat O, Elmeniar AM, Badawy M, Osman Y. Functional outcomes after pyeloplasty in solitary kidneys: structured analysis with the implication of Acute Kidney Injury Network (AKIN) staging criteria to predict long‐term renal function recoverability. BJU Int 2020; 126:502-508. [DOI: 10.1111/bju.15142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Mohamed A. Elbaset
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Mohamad H. Zahran
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Eman Elrefaie
- Nephrology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Mostafa Elgamal
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Sharaf M.A.
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Osama Ezzat
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Ali M. Elmeniar
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Mohamed Badawy
- Radiology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| | - Yasser Osman
- Urology Department, Urology and Nephrology Center Mansoura University Mansoura Egypt
| |
Collapse
|
19
|
Ban TH, Kim EN, Kim MY, Lim JH, Lee JH, Kim HD, Yoon HE, Park CW, Choi BS. Renoprotective Effect of a Dipeptidyl Peptidase-4 Inhibitor on Aging Mice. Aging Dis 2020; 11:588-602. [PMID: 32489704 PMCID: PMC7220286 DOI: 10.14336/ad.2019.0620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP-4) inhibitors exert pleiotropic effects beyond glycemic control. We investigated the renoprotective effects of DPP-4 inhibitors on aging mice mediated by the renin-angiotensin system (RAS). C57BL/6 mice were divided into three groups: the two-month-old mice (YM group), the eighteen-month-old mice (AM group) and the eighteen-month-old, linagliptin-treated mice (AM + LIN group). Renal function was improved, based on serum creatinine and cystatin-C levels (p < 0.05 compared with the AM group for both parameters). Fibrotic areas and the levels of proteins related to fibrosis improved in the AM + LIN group (p < 0.001 compared with the AM group for all parameters). In the AM + LIN group, the DPP-4-positive area and activity and expressions of DPP-4 were decreased (p < 0.05 compared with the AM group for all parameters). The levels of proteins related to the RAS, including prorenin receptor, angiotensin-converting enzyme, angiotensin II and angiotensin 1 receptor, were decreased in the AM + LIN group (p < 0.05, p < 0.01, p < 0.05, and p < 0.01 compared with the AM group, respectively). NADPH oxidase 2 and NADPH oxidase 4 levels decreased in the AM + LIN group (p < 0.001 compared with the AM group for both proteins), whereas the levels of endothelial nitric oxide synthase (eNOS) phosphorylated at serine1177 and superoxide dismutase 1 were increased (p < 0.01 compared with the AM group for both proteins). DPP-4 inhibitors may exert renoprotective effects via prorenin receptor/angiotensin-converting enzyme/angiotensin II/angiotensin 1 receptor axis.
Collapse
Affiliation(s)
- Tae H Ban
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun N Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Y Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji H Lim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong H Lee
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung D Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye E Yoon
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol W Park
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Bum S Choi
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
20
|
Lim K, Halim A, Lu TS, Ashworth A, Chong I. Klotho: A Major Shareholder in Vascular Aging Enterprises. Int J Mol Sci 2019; 20:E4637. [PMID: 31546756 PMCID: PMC6770519 DOI: 10.3390/ijms20184637] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Accelerated vascular aging is a condition that occurs as a complication of several highly prevalent inflammatory conditions such as chronic kidney disease, cancer, HIV infection and diabetes. Age-associated vascular alterations underlie a continuum of expression toward clinically overt cardiovascular disease. This has contributed to the striking epidemiologic transition whereby such noncommunicable diseases have taken center stage as modern-day global epidemics and public health problems. The identification of α-Klotho, a remarkable protein that confers powerful anti-aging properties has stimulated significant interest. In fact, emerging data have provided fundamental rationale for Klotho-based therapeutic intervention for vascular diseases and multiple other potential indications. However, the application of such discoveries in Klotho research remains fragmented due to significant gaps in our molecular understanding of Klotho biology, as well as hurdles in clinical research and experimental barriers that must first be overcome. These advances will be critical to establish the scientific platform from which future Klotho-based interventional trials and therapeutic enterprises can be successfully launched.
Collapse
Affiliation(s)
- Kenneth Lim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- MGH Renal Associates, 165 Cambridge Street, Suite 302, Boston, MA 02114, USA
| | - Arvin Halim
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Tzong-shi Lu
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (UCSF), 1450 3rd St, San Francisco, CA 94158, USA;
| | - Irene Chong
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK;
| |
Collapse
|
21
|
Xu J, Yu J, Xu X, Shen B, Wang Y, Jiang W, Lv W, Fang Y, Luo Z, Wang C, Teng J, Ding X. Preoperative hidden renal dysfunction add an age dependent risk of progressive chronic kidney disease after cardiac surgery. J Cardiothorac Surg 2019; 14:151. [PMID: 31438993 PMCID: PMC6704689 DOI: 10.1186/s13019-019-0977-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/15/2019] [Indexed: 01/08/2023] Open
Abstract
Background To study different value of estimated glomerular filtration rate with normal serum creatinine whether is a risk factor for hidden renal function of cardiac surgery outcomes. Methods A total of 1744 cardiac surgery patients with serum creatinine ≤1.2 mg/dL (female)/1.5 mg/dL (male) were divided into 3 groups: estimated glomerular filtration rate ≥ 90 mL/min/1.73 m2 (no renal dysfunction, n = 829), 60 ≤ estimated glomerular filtration rate < 90 mL/min/1.73 m2 (hidden renal dysfunction, n = 857), estimated glomerular filtration rate < 60 mL/min/1.73 m2 (known renal dysfunction, n = 58) and followed up for 3 years. Multivariate regression analyses for risk factors of postoperative acute kidney injury. Results The proportion of preoperative hidden renal dysfunction was 67.1% among patients ≥ 65 years old and 44.1% among patients < 65 years old. Multivariate Cox regression analyses showed that for patients < 65 years, known renal dysfunction was a risk factor for postoperative acute kidney injury (P < 0.01) and progressive chronic kidney disease (P = 0.018), while hidden renal dysfunction was a risk factor for progressive chronic kidney disease (P = 0.024). For patients ≥ 65 years, only known renal dysfunction was a risk factors for 3-year mortality (P = 0.022) and progressive chronic kidney disease (P < 0.01). Conclusion Hidden renal dysfunction was common in patients with normal serum creatinine for cardiac surgery, with a prevalence of 49.1%. For patients < 65 years old, hidden renal dysfunction was an independent risk factor for progressive chronic kidney disease.
Collapse
Affiliation(s)
- Jiarui Xu
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Jiawei Yu
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yimei Wang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Wuhua Jiang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Wenlv Lv
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China.,Hemodialysis Quality of Control Center of Shanghai, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China. .,Hemodialysis Quality of Control Center of Shanghai, No. 180 Fenglin Road, Shanghai, 200032, China. .,Department of Nephrology, Xiamen Branch, Zhongshan Hospital, Fudan University, No. 668 Jinhu Road, Xiamen, 361015, Fujian, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Medical Center of Kidney, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Institute for Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China. .,Hemodialysis Quality of Control Center of Shanghai, No. 180 Fenglin Road, Shanghai, 200032, China. .,Department of Nephrology, Xiamen Branch, Zhongshan Hospital, Fudan University, No. 668 Jinhu Road, Xiamen, 361015, Fujian, China.
| |
Collapse
|
22
|
Hypertension exaggerates renovascular resistance via miR-122-associated stress response in aging. J Hypertens 2018; 36:2226-2236. [DOI: 10.1097/hjh.0000000000001770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
23
|
Kanasaki K. The role of renal dipeptidyl peptidase-4 in kidney disease: renal effects of dipeptidyl peptidase-4 inhibitors with a focus on linagliptin. Clin Sci (Lond) 2018; 132:489-507. [PMID: 29491123 PMCID: PMC5828949 DOI: 10.1042/cs20180031] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggests that dipeptidyl peptidase-4 (DPP-4) inhibitors used to treat type 2 diabetes may have nephroprotective effects beyond the reduced renal risk conferred by glycemic control. DPP-4 is a ubiquitous protein with exopeptidase activity that exists in cell membrane-bound and soluble forms. The kidneys contain the highest levels of DPP-4, which is increased in diabetic nephropathy. DPP-4 inhibitors are a chemically heterogeneous class of drugs with important pharmacological differences. Of the globally marketed DPP-4 inhibitors, linagliptin is of particular interest for diabetic nephropathy as it is the only compound that is not predominantly excreted in the urine. Linagliptin is also the most potent DPP-4 inhibitor, has the highest affinity for this protein, and has the largest volume of distribution; these properties allow linagliptin to penetrate kidney tissue and tightly bind resident DPP-4. In animal models of kidney disease, linagliptin elicited multiple renoprotective effects, including reducing albuminuria, glomerulosclerosis, and tubulointerstitial fibrosis, independent of changes in glucagon-like peptide-1 (GLP-1) and glucose levels. At the molecular level, linagliptin prevented the pro-fibrotic endothelial-to-mesenchymal transition by disrupting the interaction between membrane-bound DPP-4 and integrin β1 that enhances signaling by transforming growth factor-β1 and vascular endothelial growth factor receptor-1. Linagliptin also increased stromal cell derived factor-1 levels, ameliorated endothelial dysfunction, and displayed unique antioxidant effects. Although the nephroprotective effects of linagliptin are yet to be translated to the clinical setting, the ongoing Cardiovascular and Renal Microvascular Outcome Study with Linagliptin in Patients with Type 2 Diabetes Mellitus (CARMELINA®) study will definitively assess the renal effects of this DPP-4 inhibitor. CARMELINA® is the only clinical trial of a DPP-4 inhibitor powered to evaluate kidney outcomes.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
24
|
Wang XX, Luo Y, Wang D, Adorini L, Pruzanski M, Dobrinskikh E, Levi M. A dual agonist of farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5, INT-767, reverses age-related kidney disease in mice. J Biol Chem 2017; 292:12018-12024. [PMID: 28596381 DOI: 10.1074/jbc.c117.794982] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/07/2017] [Indexed: 11/06/2022] Open
Abstract
Even in healthy individuals, renal function gradually declines during aging. However, an observed variation in the rate of this decline has raised the possibility of slowing or delaying age-related kidney disease. One of the most successful interventional measures that slows down and delays age-related kidney disease is caloric restriction. We undertook the present studies to search for potential factors that are regulated by caloric restriction and act as caloric restriction mimetics. Based on our prior studies with the bile acid-activated nuclear hormone receptor farnesoid X receptor (FXR) and G protein-coupled membrane receptor TGR5 that demonstrated beneficial effects of FXR and TGR5 activation in the kidney, we reasoned that FXR and TGR5 could be excellent candidates. We therefore determined the effects of aging and caloric restriction on the expression of FXR and TGR5 in the kidney. We found that FXR and TGR5 expression levels are decreased in the aging kidney and that caloric restriction prevents these age-related decreases. Interestingly, in long-lived Ames dwarf mice, renal FXR and TGR5 expression levels were also increased. A 2-month treatment of 22-month-old C57BL/6J mice with the FXR-TGR5 dual agonist INT-767 induced caloric restriction-like effects and reversed age-related increases in proteinuria, podocyte injury, fibronectin accumulation, TGF-β expression, and, most notably, age-related impairments in mitochondrial biogenesis and mitochondrial function. Furthermore, in podocytes cultured in serum obtained from old mice, INT-767 prevented the increases in the proinflammatory markers TNF-α, toll-like receptor 2 (TLR2), and TLR4. In summary, our results indicate that FXR and TGR5 may play an important role in modulation of age-related kidney disease.
Collapse
Affiliation(s)
- Xiaoxin X Wang
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver Veterans Affairs Medical Center and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Yuhuan Luo
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver Veterans Affairs Medical Center and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Dong Wang
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver Veterans Affairs Medical Center and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | | | | | - Evgenia Dobrinskikh
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver Veterans Affairs Medical Center and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Moshe Levi
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver Veterans Affairs Medical Center and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045.
| |
Collapse
|
25
|
Dinkler JM, Sugar CA, Escarce JJ, Ong MK, Mangione CM. Does Age Matter? Association Between Usual Source of Care and Hypertension Control in the US Population: Data From NHANES 2007-2012. Am J Hypertens 2016; 29:934-40. [PMID: 26884134 PMCID: PMC5006109 DOI: 10.1093/ajh/hpw010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/15/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The positive role of having a usual source of care (USOC) on the receipt of preventative services is known. However, associations between USOC and hypertension control and the differential association across age groups is unknown in the US population. METHODS We used data from the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2012. Multivariable logistic regression was used to evaluate the association between having a USOC and hypertension control. The differential effect of USOC on hypertension control by age was assessed using predicted marginal effects across age groups in the multivariable logistic model. RESULTS In adjusted analyses, those with a USOC had higher odds of hypertension control (odds ratio = 3.89, 95% confidence interval (CI): 2.15-6.98). The marginal effect of having a USOC is associated with a 30 percentage point higher probability of controlled blood pressure compared to those without a USOC (marginal probability = 0.30, 95% CI: 0.19-0.41). The marginal effect of USOC on hypertension control varied by age groups, with a statistically significantly lower marginal effect of USOC on hypertension seen among those older than 74 years of age (marginal probability = 0.27, 95% CI: 0.18-0.36) and younger than 35 years of age (marginal probability = 0.23, 95% CI: 0.14-0.33). CONCLUSION Having a USOC is significantly associated with improved hypertension control in the US population. The variation in the association across age groups has important implications in targeting age-specific antihypertensive strategies to reduce the burden of hypertension in the US population.
Collapse
Affiliation(s)
- John M Dinkler
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, California, USA; Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, California, USA;
| | - Catherine A Sugar
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - José J Escarce
- Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, California, USA; Division of General Internal Medicine and Health Services Research, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| | - Michael K Ong
- Division of General Internal Medicine and Health Services Research, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA; VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Carol M Mangione
- Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, California, USA; Division of General Internal Medicine and Health Services Research, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
26
|
Oral Administration of N-Acetyl-seryl-aspartyl-lysyl-proline Ameliorates Kidney Disease in Both Type 1 and Type 2 Diabetic Mice via a Therapeutic Regimen. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9172157. [PMID: 27088094 PMCID: PMC4818806 DOI: 10.1155/2016/9172157] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/14/2016] [Indexed: 01/03/2023]
Abstract
Kidney fibrosis is the final common pathway of progressive kidney diseases including diabetic nephropathy. Here, we report that the endogenous antifibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), the substrate of angiotensin-converting enzyme (ACE), is an orally available peptide drug used to cure kidney fibrosis in diabetic mice. We utilized two mouse models of diabetic nephropathy, streptozotocin- (STZ-) induced type 1 diabetic CD-1 mice and type 2 diabetic nephropathy model db/db mice. Intervention with the ACE inhibitor imidapril, oral AcSDKP, or imidapril + oral AcSDKP combination therapy increased urine AcSDKP levels. AcSDKP levels were significantly higher in the combination group compared to those of the other groups. AcSDKP oral administration, either AcSDKP alone or in addition to imidapril, ameliorated glomerulosclerosis and tubulointerstitial fibrosis. Plasma cystatin C levels were higher in both models, at euthanasia, and were restored by all the treatment groups. The levels of antifibrotic miRs, such as miR-29 or let-7, were suppressed in the kidneys of both models; all treatments, especially the combination of imidapril + oral AcSDKP, restored the antifibrotic miR levels to a normal value or even higher. AcSDKP may be an oral antifibrotic peptide drug that would be relevant to combating fibroproliferative kidney diseases such as diabetic nephropathy.
Collapse
|
27
|
Ng CF, Luke S, Chiu PKF, Teoh JYC, Wong KT, Hou SSM. The effect of renal cortical thickness on the treatment outcomes of kidney stones treated with shockwave lithotripsy. Korean J Urol 2015; 56:379-85. [PMID: 25964839 PMCID: PMC4426510 DOI: 10.4111/kju.2015.56.5.379] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/04/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Because the shock wave passes through various body tissues before reaching the stone, stone composition may affect the treatment efficacy of shock wave lithotripsy (SWL). We investigated the effect of various tissue components along the shock wave path on the success of SWL. MATERIALS AND METHODS From October 2008 to August 2010, a total of 206 patients with kidney stones sized 5 to 20 mm were prospectively recruited for a study of the factors that affect the outcome of treatment with a Sonolith Vision lithotripter. Successful SWL was defined as either stone-free status or residual fragments <4 mm at 12 weeks. Logistic regression analysis was performed to assess the factors that predicted treatment outcomes. Potential predictors included the patient's age, shock wave delivery rate, stone volume (SV), mean stone density (MSD), skin-to-stone distance (SSD), and the mean thickness of the three main components along the shock wave path: renal cortical thickness (KT), muscle thickness (MT), and soft-tissue thickness (ST). RESULTS The mean age of the patients was 53.8 years (range, 25-82 years). The overall treatment success rate after one session of SWL was 43.2%. The mean KT, MT, and ST were 26.9, 16.6, and 40.8 mm, respectively. The logistic regression results showed that a slower shock wave delivery rate, smaller SV, a lower MSD, and a thicker KT were found to be significant predictors for successful SWL. SSD, MT, and ST were not predictors of successful treatment. CONCLUSIONS Among the main tissue components along the shock wave path, a thicker KT was a favorable factor for successful SWL after adjustment for SV, MSD, and the shock wave delivery rate.
Collapse
Affiliation(s)
- Chi-Fai Ng
- The S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Sylvia Luke
- The S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Peter K F Chiu
- The S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeremy Y C Teoh
- The S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka-Tak Wong
- Department of Imaging and Intervention Radiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Simon S M Hou
- The S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
28
|
|
29
|
Abstract
PURPOSE OF REVIEW To describe the effect of ageing on kidney function and to summarize the benefits of advocated measures to prevent perioperative acute kidney injury (AKI) in elderly patients. RECENT FINDINGS Given a reduced renal reserve and the burden of comorbidities, the senescent kidney is susceptible to develop perioperative AKI and is less able to recover when injury occurs. Current evidence suggests that preoperative statin therapy, tight glycemic control or urine alkalinization with bicarbonate do not protect the kidneys from harm. The theoretical kidney protective effect of preoperative aspirin therapy or renal vasodilatation with atrial natriuretic peptide or fenoldopam is only supported by low-quality evidence that needs further evaluation. Although questions regarding the amount and timing of fluid resuscitation during surgery are seeking answers in ongoing multicenter studies, the harmful effect of hydroxyethyl starches (HES) and hyperchloremic solutions is now surrounded by strong evidence. SUMMARY The future increase in elderly patients being exposed to surgery calls for improved perioperative management to prevent collaterally increased AKI. Although pharmacological therapies aiming to protect the kidneys from harm are under evaluation, hemodynamic optimization and avoidance of nephrotoxic drugs, including HES and hyperchloremic solutions, are critical for the elderly perioperative patient.
Collapse
|
30
|
Nagai T, Nitta K, Kanasaki M, Koya D, Kanasaki K. The biological significance of angiotensin-converting enzyme inhibition to combat kidney fibrosis. Clin Exp Nephrol 2014; 19:65-74. [PMID: 24975544 DOI: 10.1007/s10157-014-1000-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/08/2014] [Indexed: 12/23/2022]
Abstract
Both angiotensin-converting enzyme inhibitor (ACE-I) and angiotensin II receptor blocker have been recognized as renin-angiotensin system (RAS) inhibitors. These two RAS inhibitors are rarely recognized as drugs with distinct pharmacological effects in the clinic or most clinical trials. Some preclinical basic research and clinical trials indicate that ACE-I might display superior organ-protective effects, especially anti-fibrotic effects. Such anti-fibrotic effects of ACE-I could be associated with an endogenous anti-fibrotic peptide, N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP). In this review, we focused on the anti-fibrotic effects of RAS inhibition and the endogenous anti-fibrotic peptide AcSDKP.
Collapse
Affiliation(s)
- Takako Nagai
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | | | | | | | | |
Collapse
|
31
|
Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, Srivastava SP, Koya D. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 2014; 63:2120-31. [PMID: 24574044 DOI: 10.2337/db13-1029] [Citation(s) in RCA: 281] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Kidney fibrosis is the final common pathway of all progressive chronic kidney diseases, of which diabetic nephropathy is the leading cause. Endothelial-to-mesenchymal transition (EndMT) has emerged as one of the most important origins of matrix-producing fibroblasts. Dipeptidyl peptidase-4 (DPP-4) inhibitors have been introduced into the market as antidiabetes drugs. Here, we found that the DPP-4 inhibitor linagliptin ameliorated kidney fibrosis in diabetic mice without altering the blood glucose levels associated with the inhibition of EndMT and the restoration of microRNA 29s. Streptozotocin-induced diabetic CD-1 mice exhibited kidney fibrosis and strong immunoreactivity for DPP-4 by 24 weeks after the onset of diabetes. At 20 weeks after the onset of diabetes, mice were treated with linagliptin for 4 weeks. Linagliptin-treated diabetic mice exhibited a suppression of DPP-4 activity/protein expression and an amelioration of kidney fibrosis associated with the inhibition of EndMT. The therapeutic effects of linagliptin on diabetic kidneys were associated with the suppression of profibrotic programs, as assessed by mRNA microarray analysis. We found that the induction of DPP-4 observed in diabetic kidneys may be associated with suppressed levels of microRNA 29s in diabetic mice; linagliptin restored microRNA 29s and suppressed DPP-4 protein levels. Using cultured endothelial cells, we found that linagliptin inhibited TGF-β2-induced EndMT, and such anti-EndMT effects of linagliptin were mediated through microRNA 29 induction. These results indicate the possible novel pleiotropic action of linagliptin to restore normal kidney function in diabetic patients with renal impairment.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Sen Shi
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Megumi Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Jianhua He
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Takako Nagai
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | | | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
32
|
Radosavljevic N, Nikolic D, Lazovic M, Jeremic A. Hip fractures in a geriatric population - rehabilitation based on patients needs. Aging Dis 2014; 5:177-82. [PMID: 24900940 DOI: 10.14336/ad.2014.0500177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/10/2013] [Accepted: 10/14/2013] [Indexed: 01/05/2023] Open
Abstract
With an increased life expectancy in humans and thus an increase in the number of the elderly population, the frequency of hip fractures will rise as well. Aside from a higher incidence, hip fractures in a geriatric population is a significant problem due to the possible onset of severe and in some cases dramatic complications and consequences. The primary purpose of treatment and rehabilitation in the elderly after a hip fracture is to improve an individual's quality of life. It is important to underline that principles and methods of functional restoration after hip fracture should consider careful planning of a rehabilitation program individually for every patient and its implementation with respect to decisions made by the rehabilitation team.
Collapse
Affiliation(s)
| | - Dejan Nikolic
- Physical Medicine and Rehabilitation Department, University Children's Hospital, Belgrade, Serbia
| | | | | |
Collapse
|
33
|
Kanasaki K, Nagai T, Nitta K, Kitada M, Koya D. N-acetyl-seryl-aspartyl-lysyl-proline: a valuable endogenous anti-fibrotic peptide for combating kidney fibrosis in diabetes. Front Pharmacol 2014; 5:70. [PMID: 24782774 PMCID: PMC3995071 DOI: 10.3389/fphar.2014.00070] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/25/2014] [Indexed: 12/11/2022] Open
Abstract
Fibroproliferative diseases are responsible for 45% of deaths in the developed world. Curing organ fibrosis is essential for fibroproliferative diseases. Diabetic nephropathy is a common fibroproliferative disease of the kidney and is associated with multiorgan dysfunction. However, therapy to combat diabetic nephropathy has not yet been established. In this review, we discuss the novel therapeutic possibilities for kidney fibrosis in diabetes focusing on the endogenous anti-fibrotic peptide, N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), which is the substrate for angiotensin-converting enzyme and exhibits meaningful anti-fibrotic effects in various experimental models of fibrotic disease.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University Uchinada, Ishikawa, Japan
| | - Takako Nagai
- Department of Diabetology and Endocrinology, Kanazawa Medical University Uchinada, Ishikawa, Japan
| | - Kyoko Nitta
- Department of Diabetology and Endocrinology, Kanazawa Medical University Uchinada, Ishikawa, Japan
| | - Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University Uchinada, Ishikawa, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University Uchinada, Ishikawa, Japan
| |
Collapse
|
34
|
Abstract
Aging is associated with structural and functional changes in the kidney. Structural changes include glomerulosclerosis, thickening of the basement membrane, increase in mesangial matrix, tubulointerstitial fibrosis and arteriosclerosis. Glomerular filtration rate is maintained until the fourth decade of life, after which it declines. Parallel reductions in renal blood flow occur with redistribution of blood flow from the cortex to the medulla. Other functional changes include an increase in glomerular basement permeability and decreased ability to dilute or concentrate urine.
Collapse
Affiliation(s)
- Zeina Karam
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
35
|
Suzuki Y, Tsuneyama N, Fukui N, Sugai T, Watanabe J, Ono S, Saito M, Inoue Y, Someya T. Effect of risperidone metabolism and P-glycoprotein gene polymorphism on QT interval in patients with schizophrenia. THE PHARMACOGENOMICS JOURNAL 2014; 14:452-6. [PMID: 24589909 DOI: 10.1038/tpj.2014.6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 11/09/2022]
Abstract
Risperidone (RIS) is a frequently used efficacious psychotropic drug. However, it prolongs the QTc interval and may cause fatal arrhythmia. Little is known on the determinants of this RIS side effect. RIS is metabolized by CYP2D6, and is subject to drug efflux by P-glycoprotein (P-gp) encoded by the ATP-binding cassette subfamily B member 1 (ABCB1) gene. P-gp removes both RIS and its metabolite 9-OH-RIS from cardiac tissue. To investigate the effect of RIS metabolism and ABCB1 gene polymorphisms on QTc, steady-state plasma RIS and 9-OH-RIS levels, and QTc were measured. CYP2D6, ABCB1 C3435T and G2677T/A genotypes were determined in 66 schizophrenia patients on RIS. QTc was significantly longer in patients with ABCB1 3435CT+3435 TT than in those with 3435CC (P=0.006). ABCB1 G2677T/A genotype did not affect QTc. Multiple regression analysis showed that C/T or T/T genotypes at the ABCB1 C3435T locus, lower weight, and older age prolonged QTc. In summary, the T allele of the ABCB1 C3435T genotype should be considered in future diagnostic development efforts for RIS-associated QT.
Collapse
Affiliation(s)
- Y Suzuki
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - N Tsuneyama
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - N Fukui
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - T Sugai
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - J Watanabe
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - S Ono
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - M Saito
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Y Inoue
- MP-Technopharma Corporation Technology Department, Fukuoka, Japan
| | - T Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
36
|
Nitta K, Okada K, Yanai M, Takahashi S. Aging and chronic kidney disease. Kidney Blood Press Res 2014; 38:109-20. [PMID: 24642796 DOI: 10.1159/000355760] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2014] [Indexed: 11/19/2022] Open
Abstract
A recent report has dealt with geriatric nephrology, including epidemiology and pathophysiology of chronic kidney disease (CKD), attempting to get nephrologists to pay more attention to elderly CKD patients. The aims of this article are to summarize the morphological and functional properties of the aging kidney, and to better understand nephrology care for elderly CKD patients. The kidneys are affected by the aging process, which results in numerous effects on the renal system. In addition, the elderly population is hetereogenous - some have a decline in GFR explained by diseases that complicate aging such as arteriosclerosis with hypertension, whereas in the most of healthy adults the decline in GFR is much more modest and not inevitable. The values for normal estimated glomerular filtration rate (eGFR) in aging population have important implications for the diagnosis of CKD in the elderly. However, the MDRD equation underestimates mean eGFR by 25% and the CKD-EPI equation underestimates mean GFR by 16%. This bias may lead to misclassifying healthy older persons as having CKD. It is also still unknown whether and how age influences the predictive role of other risk factors for end-stage renal disease (ESRD) and death in referred as well as unreferred patients. The risk of ESRD was reported to be higher than the risk of death without ESRD for ages <60 years, and independent of eGFR. Proteinuria significantly increased the risk of ESRD with advancing age. In older patients on nephrology care, the risk of ESRD prevailed over mortality even when eGFR was not severely impaired. Proteinuria increases the risk of ESRD, while the predictive role of other modifiable risk factors was unchanged compared with younger patients. The decision to initiate renal replacement therapy in the elderly is complicated by more challenges than in younger patients. Calorie restriction and Klotho deficiency may be a candidate therapeutic target for attenuating kidney aging. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Kosaku Nitta
- International Kidney Evaluation Association Japan (IKEAJ), Tokyo, Japan
| | | | | | | |
Collapse
|
37
|
Stenvinkel P, Larsson TE. Chronic kidney disease: a clinical model of premature aging. Am J Kidney Dis 2013; 62:339-51. [PMID: 23357108 DOI: 10.1053/j.ajkd.2012.11.051] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/19/2012] [Indexed: 12/21/2022]
Abstract
Premature aging is a process associated with a progressive accumulation of deleterious changes over time, an impairment of physiologic functions, and an increase in the risk of disease and death. Regardless of genetic background, aging can be accelerated by the lifestyle choices and environmental conditions to which our genes are exposed. Chronic kidney disease is a common condition that promotes cellular senescence and premature aging through toxic alterations in the internal milieu. This occurs through several mechanisms, including DNA and mitochondria damage, increased reactive oxygen species generation, persistent inflammation, stem cell exhaustion, phosphate toxicity, decreased klotho expression, and telomere attrition. Because recent evidence suggests that both increased local signaling of growth factors (through the nutrient-sensing mammalian target of rapamycin) and decreased klotho expression are important modulators of aging, interventions that target these should be tested in this prematurely aged population.
Collapse
Affiliation(s)
- Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|