51
|
Chiao YA, Zhang H, Sweetwyne M, Whitson J, Ting YS, Basisty N, Pino LK, Quarles E, Nguyen NH, Campbell MD, Zhang T, Gaffrey MJ, Merrihew G, Wang L, Yue Y, Duan D, Granzier HL, Szeto HH, Qian WJ, Marcinek D, MacCoss MJ, Rabinovitch P. Late-life restoration of mitochondrial function reverses cardiac dysfunction in old mice. eLife 2020; 9:e55513. [PMID: 32648542 PMCID: PMC7377906 DOI: 10.7554/elife.55513] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/07/2020] [Indexed: 12/26/2022] Open
Abstract
Diastolic dysfunction is a prominent feature of cardiac aging in both mice and humans. We show here that 8-week treatment of old mice with the mitochondrial targeted peptide SS-31 (elamipretide) can substantially reverse this deficit. SS-31 normalized the increase in proton leak and reduced mitochondrial ROS in cardiomyocytes from old mice, accompanied by reduced protein oxidation and a shift towards a more reduced protein thiol redox state in old hearts. Improved diastolic function was concordant with increased phosphorylation of cMyBP-C Ser282 but was independent of titin isoform shift. Late-life viral expression of mitochondrial-targeted catalase (mCAT) produced similar functional benefits in old mice and SS-31 did not improve cardiac function of old mCAT mice, implicating normalizing mitochondrial oxidative stress as an overlapping mechanism. These results demonstrate that pre-existing cardiac aging phenotypes can be reversed by targeting mitochondrial dysfunction and implicate mitochondrial energetics and redox signaling as therapeutic targets for cardiac aging.
Collapse
Affiliation(s)
- Ying Ann Chiao
- Department of Pathology, University of WashingtonSeattleUnited States
- Aging and Metabolism Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
| | - Huiliang Zhang
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Mariya Sweetwyne
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Jeremy Whitson
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Ying Sonia Ting
- Department of Genome Science, University of WashingtonSeattleUnited States
| | | | - Lindsay K Pino
- Department of Genome Science, University of WashingtonSeattleUnited States
| | - Ellen Quarles
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Ngoc-Han Nguyen
- Department of Pathology, University of WashingtonSeattleUnited States
| | | | - Tong Zhang
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandUnited States
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandUnited States
| | - Gennifer Merrihew
- Department of Genome Science, University of WashingtonSeattleUnited States
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of WashingtonSeattleUnited States
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of MissouriColumbiaUnited States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of MissouriColumbiaUnited States
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
| | | | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandUnited States
| | - David Marcinek
- Department of Radiology, University of WashingtonSeattleUnited States
| | - Michael J MacCoss
- Department of Genome Science, University of WashingtonSeattleUnited States
| | - Peter Rabinovitch
- Department of Pathology, University of WashingtonSeattleUnited States
| |
Collapse
|
52
|
Chu Y, Lan RS, Huang R, Feng H, Kumar R, Dayal S, Chan K, Dai D. Glutathione peroxidase-1 overexpression reduces oxidative stress, and improves pathology and proteome remodeling in the kidneys of old mice. Aging Cell 2020; 19:e13154. [PMID: 32400101 PMCID: PMC7294784 DOI: 10.1111/acel.13154] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/29/2020] [Indexed: 12/15/2022] Open
Abstract
This study investigated the direct roles of hydrogen peroxide (H2 O2 ) in kidney aging using transgenic mice overexpressing glutathione peroxidase-1 (GPX1 TG). We demonstrated that kidneys in old mice recapitulated kidneys in elderly humans and were characterized by glomerulosclerosis, tubular atrophy, interstitial fibrosis, and loss of cortical mass. Scavenging H2 O2 by GPX1 TG significantly reduced mitochondrial and total cellular reactive oxygen species (ROS) and mitigated oxidative damage, thus improving these pathologies. The potential mechanisms by which ROS are increased in the aged kidney include a decreased abundance of an anti-aging hormone, Klotho, in kidney tissue, and decreased expression of nuclear respiratory factor 2 (Nrf2), a master regulator of the stress response. Decreased Klotho or Nrf2 was not improved in the kidneys of old GPX1 TG mice, even though mitochondrial morphology was better preserved. Using laser capture microdissection followed by label-free shotgun proteomics analysis, we show that the glomerular proteome in old mice was characterized by decreased abundance of cytoskeletal proteins (critical for maintaining normal glomerular function) and heat shock proteins, leading to increased accumulation of apolipoprotein E and inflammatory molecules. Targeted proteomic analysis of kidney tubules from old mice showed decreased abundance of fatty acid oxidation enzymes and antioxidant proteins, as well as increased abundance of glycolytic enzymes and molecular chaperones. GPX1 TG partially attenuated the remodeling of glomerular and tubule proteomes in aged kidneys. In summary, mitochondria from GPX1 TG mice are protected and kidney aging is ameliorated via its antioxidant activities, independent and downstream of Nrf2 or Klotho signaling.
Collapse
Affiliation(s)
- Yi Chu
- Department of PathologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Renny S. Lan
- Proteomics CoreDepartment of Biochemistry and Molecular BiologyUniversity of Arkansas Medical Sciences College of MedicineLittle RockArkansas
| | - Rui Huang
- Department of Statistics and Actuarial ScienceCollege of Liberal Arts and SciencesUniversity of IowaIowa CityIowa
| | - Hao Feng
- Department of PathologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Rahul Kumar
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Sanjana Dayal
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Kung‐Sik Chan
- Department of Statistics and Actuarial ScienceCollege of Liberal Arts and SciencesUniversity of IowaIowa CityIowa
| | - Dao‐Fu Dai
- Department of PathologyCarver College of MedicineUniversity of IowaIowa CityIowa
| |
Collapse
|
53
|
Hamatani H, Eng DG, Hiromura K, Pippin JW, Shankland SJ. CD44 impacts glomerular parietal epithelial cell changes in the aged mouse kidney. Physiol Rep 2020; 8:e14487. [PMID: 32597007 PMCID: PMC7322268 DOI: 10.14814/phy2.14487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
CD44 contributes to the activation of glomerular parietal epithelial cells (PECs). Although CD44 expression is higher in PECs of healthy aged mice, the biological role of CD44 in PECs in this context remains unclear. Accordingly, young (4 months) and aged (24 months) CD44-/- mice were compared to age-matched CD44+/+ mice, both aged in a nonstressed environment. Parietal epithelial cell densities were similar in both young and aged CD44+/+ and CD44-/- mice. Phosphorylated ERK 1/2 (pERK) was higher in aged CD44+/+ mice. Vimentin and α-SMA, markers of changes to the epithelial cell phenotype, were present in PECs in aged CD44+/+ mice, but absent in aged CD44-/- mice in both outer cortical (OC) and juxtamedullary (JM) glomeruli. Because age-related glomerular hypertrophy was lower in CD44-/- mice, mTOR activation was assessed by phospho-S6 ribosomal protein (pS6RP) staining. Parietal epithelial cells and glomerular tuft staining for pS6RP was lower in aged CD44-/- mice compared to aged CD44+/+ mice. Podocyte density was higher in aged CD44-/- mice in both OC and JM glomeruli. These changes were accompanied by segmental and global glomerulosclerosis in aged CD44+/+ mice, but absent in aged CD44-/- mice. These results show that the increase in CD44 in PECs in aged kidneys contributes to several changes to the glomerulus during healthy aging in mice, and may involve ERK and mTOR activation.
Collapse
Affiliation(s)
- Hiroko Hamatani
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Diana G. Eng
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Jeffrey W. Pippin
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Stuart J. Shankland
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
54
|
Sugiyama M, Wada Y, Kanazawa N, Tachibana S, Suzuki T, Matsumoto K, Iyoda M, Honda H, Shibata T. A cross-sectional analysis of clinicopathologic similarities and differences between Henoch-Schönlein purpura nephritis and IgA nephropathy. PLoS One 2020; 15:e0232194. [PMID: 32324811 PMCID: PMC7179927 DOI: 10.1371/journal.pone.0232194] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction Recent studies noted that Henoch-Schönlein purpura nephritis (HSPN) and IgA nephropathy (IgAN) share the feature of galactose-deficient IgA1 (Gd-IgA1)-oriented pathogenesis, although there are distinct clinical differences. We aimed to clarify the clinicopathologic differences between these 2 diseases. Methods We cross-sectionally analyzed adult patients with HSPN (n = 24) or IgAN (n = 56) who underwent renal biopsy (RB) between 2008 and 2018 at Showa University Hospital. Serum Gd-IgA1 (s-Gd-IgA1) levels at the time of RB were compared among study groups using enzyme-linked immunosorbent assay (ELISA) with anti-human Gd-IgA1-specific monoclonal antibody (KM55). We also immunohistochemically stained paraffin-embedded sections for glomerular Gd-IgA1 (g-Gd-IgA1)-deposition using KM55. Serum inflammatory cytokines were measured using ELISA. Results Glomerular endothelial injury with subendothelial IgA deposition was significant in patients with HSPN. Serum IL-8, MCP-1, TNF-α, and IL-6 levels were significantly higher in patients with HSPN than IgAN. Levels of s-Gd-IgA1 were comparable among patients with HSPN and IgAN, and a similar degree of g-Gd-IgA1-deposition was detected in both diseases. Furthermore, g-Gd-IgA1-deposition was evident in patients with histopathologically advanced HSPN or IgAN. In HSPN, significant positive correlations between s-Gd-IgA1 levels and crescent formation or IL-6 elevation were confirmed, and g-Gd-IgA1 intensity showed a significant positive correlation with MCP-1 and a tendency to positively correlate with IL-8. Meanwhile, patients with IgAN showed no correlation between inflammatory cytokines and both-Gd-IgA1. Moreover, most g-Gd-IgA1-positive areas were not double stained with CD31 in HSPN. Conclusions Although assessing both-Gd-IgA1 alone was insufficient to distinguish between HSPN and IgAN, patients with HSPN showed considerable glomerular capillaritis with subendothelial IgA deposition and significant elevation of serum inflammatory cytokines. Furthermore, such glomerular subendothelial IgA deposition might not contain Gd-IgA1, and factors associated with Gd-IgA1 were inconsistent among these 2 diseases. Thus, developmental mechanisms for IgAN might not apply to HSPN completely, and these 2 diseases still have different aspects.
Collapse
Affiliation(s)
- Motonori Sugiyama
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yukihiro Wada
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Shibata
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
55
|
Goligorsky MS. Chronic Kidney Disease: A Vicarious Relation to Premature Cell Senescence. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1164-1171. [PMID: 32194054 DOI: 10.1016/j.ajpath.2020.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/25/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease (CKD), commonly fostering nonrenal complications, themselves more life threatening than renal pathology, remains enigmatic. Despite more than a century of intense research, therapeutic options to halt or reverse renal disease are rather limited. Recently, similarity between manifestations of progressive CKD and aging kidney has attracted investigative attention that revealed senescent cells and secreting proinflammatory and profibrotic mediators in all renal compartments, even at young age, in patients with kidney maladies. The overlapping features of these categories have been noticed previously and are briefly summarized herein. I propose two hypothetical scenarios for interactive association of kidney diseases and cell senescence, both culminating in progressive deterioration of renal function. Persistence of senescent cells is considered as a critical contributor to this association; and the mechanisms explaining persistence, such as activation of cell cycle regulators, anti-apoptotic stimuli, metabolic aberrations, and their interactions, are discussed. The mutual encroachment of underlying kidney disease and cell senescence bring about the conclusion that both entities merge along the natural history of the disease. This putative interpretation of vicarious relation between cell senescence and CKD may expand the arsenal of pharmacotherapy to include the judicious use of senotherapeutics in the management of renal disease.
Collapse
Affiliation(s)
- Michael S Goligorsky
- Renal Research Institute, and the Departments of Medicine, Pharmacology, and Physiology, New York Medical College at Touro University, Valhalla, New York.
| |
Collapse
|
56
|
Domondon M, Polina I, Nikiforova AB, Sultanova RF, Kruger C, Vasileva VY, Fomin MV, Beeson GC, Nieminen AL, Smythe N, Maldonado EN, Stadler K, Ilatovskaya DV. Renal Glomerular Mitochondria Function in Salt-Sensitive Hypertension. Front Physiol 2020; 10:1588. [PMID: 32116733 PMCID: PMC7010849 DOI: 10.3389/fphys.2019.01588] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Salt-sensitive (SS) hypertension is accompanied with an early onset of proteinuria, which results from the loss of glomerular podocytes. Here, we hypothesized that glomerular damage in the SS hypertension occurs in part due to mitochondria dysfunction, and we used a unique model of freshly isolated glomeruli to test this hypothesis. In order to mimic SS hypertension, we used Dahl SS rats, an established animal model. Animals were fed a 0.4% NaCl (normal salt, NS) diet or challenged with a high salt (HS) 4% NaCl diet for 21 days to induce an increase in blood pressure (BP). Similar to previous studies, we found that HS diet caused renal hypertrophy, increased BP, glomerulosclerosis, and renal lesions such as fibrosis and protein casts. We did not observe changes in mitochondrial biogenesis in the renal cortex or isolated glomeruli fractions. However, Seahorse assay performed on freshly isolated glomeruli revealed that basal mitochondrial respiration, maximal respiration, and spare respiratory capacity were lower in the HS compared to the NS group. Using confocal imaging and staining for mitochondrial H2O2 using mitoPY1, we detected an intensified response to an acute H2O2 application in the podocytes of the glomeruli isolated from the HS diet fed group. TEM analysis showed that glomerular mitochondria from the HS diet fed group have structural abnormalities (swelling, enlargement, less defined cristae). Therefore, we report that glomerular mitochondria in SS hypertension are functionally and structurally defective, and this impairment could eventually lead to loss of podocytes and proteinuria. Thus, the glomerular–mitochondria axis can be targeted in novel treatment strategies for hypertensive glomerulosclerosis.
Collapse
Affiliation(s)
- Mark Domondon
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States
| | - Iuliia Polina
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States
| | - Anna B Nikiforova
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States.,Institute of Theoretical and Experimental Biophysics, Pushchino, Russia
| | - Regina F Sultanova
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States.,Saint-Petersburg State Chemical Pharmaceutical University, Saint Petersburg, Russia
| | - Claudia Kruger
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Valeriia Y Vasileva
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States.,Institute of Cytology Russian Academy of Science, Saint Petersburg, Russia
| | - Mikhail V Fomin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States
| | - Gyda C Beeson
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Anna-Liisa Nieminen
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Nancy Smythe
- Department of Pathology, Medical University of South Carolina, Charleston, SC, United States
| | - Eduardo N Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Daria V Ilatovskaya
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
57
|
Fujii Y, Matsumura H, Yamazaki S, Shirasu A, Nakakura H, Ogihara T, Ashida A. Efficacy of a mitochondrion-targeting agent for reducing the level of urinary protein in rats with puromycin aminonucleoside-induced minimal-change nephrotic syndrome. PLoS One 2020; 15:e0227414. [PMID: 31905213 PMCID: PMC6944386 DOI: 10.1371/journal.pone.0227414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/18/2019] [Indexed: 01/22/2023] Open
Abstract
Background Oxidative stress is a major factor responsible for minimal-change nephrotic syndrome (MCNS), which occurs most commonly in children. However, the influence of oxidative stress localized to mitochondria remains unclear. We examined the effect of a mitochondrion-targeting antioxidant, MitoTEMPO, in rats with puromycin aminonucleoside (PAN)-induced MCNS to clarify the degree to which mitochondrial oxidative stress affects MCNS. Materials and methods Thirty Wistar rats were divided into three groups: normal saline group (n = 7), PAN group (n = 12), and PAN + MitoTEMPO group (n = 11). Rats in the PAN and PAN + MitoTEMPO groups received PAN on day 1, and those in the PAN + MitoTEMPO group received MitoTEMPO on days 0 to 9. Whole-day urine samples were collected on days 3 and 9, and samples of glomeruli and blood were taken for measurement of lipid peroxidation products. We also estimated the mitochondrial damage score in podocytes in all 3 groups using electron microscopy. Results Urinary protein excretion on day 9 and the levels of lipid peroxidation products in urine, glomeruli, and blood were significantly lower in the PAN + MitoTEMPO group than in the PAN group (p = 0.0019, p = 0.011, p = 0.039, p = 0.030). The mitochondrial damage score in podocytes was significantly lower in the PAN + MitoTEMPO group than in the PAN group (p <0.0001). Conclusions This mitochondrion-targeting agent was shown to reduce oxidative stress and mitochondrial damage in a MCNS model. A radical scavenger targeting mitochondria could be a promising drug for treatment of MCNS.
Collapse
Affiliation(s)
- Yuko Fujii
- Department of Pediatrics, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Hideki Matsumura
- Department of Pediatrics, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Satoshi Yamazaki
- Department of Pediatrics, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Akihiko Shirasu
- Department of Pediatrics, Hirakata City Hospital, Hirakata, Osaka, Japan
| | - Hyogo Nakakura
- Department of Hemodialysis and Apheresis, Arisawa General Hospital, Hirakata, Osaka, Japan
| | - Tohru Ogihara
- Department of Pediatrics, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Akira Ashida
- Department of Pediatrics, Osaka Medical College, Takatsuki, Osaka, Japan
- * E-mail:
| |
Collapse
|
58
|
Jeena M, Kim S, Jin S, Ryu JH. Recent Progress in Mitochondria-Targeted Drug and Drug-Free Agents for Cancer Therapy. Cancers (Basel) 2019; 12:cancers12010004. [PMID: 31861339 PMCID: PMC7016936 DOI: 10.3390/cancers12010004] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023] Open
Abstract
The mitochondrion is a dynamic eukaryotic organelle that controls lethal and vital functions of the cell. Being a critical center of metabolic activities and involved in many diseases, mitochondria have been attracting attention as a potential target for therapeutics, especially for cancer treatment. Structural and functional differences between healthy and cancerous mitochondria, such as membrane potential, respiratory rate, energy production pathway, and gene mutations, could be employed for the design of selective targeting systems for cancer mitochondria. A number of mitochondria-targeting compounds, including mitochondria-directed conventional drugs, mitochondrial proteins/metabolism-inhibiting agents, and mitochondria-targeted photosensitizers, have been discussed. Recently, certain drug-free approaches have been introduced as an alternative to induce selective cancer mitochondria dysfunction, such as intramitochondrial aggregation, self-assembly, and biomineralization. In this review, we discuss the recent progress in mitochondria-targeted cancer therapy from the conventional approach of drug/cytotoxic agent conjugates to advanced drug-free approaches.
Collapse
|
59
|
The Role of Cardiolipin and Mitochondrial Damage in Kidney Transplant. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3836186. [PMID: 31885786 PMCID: PMC6899302 DOI: 10.1155/2019/3836186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is highly incident and prevalent in the world. The death of patients with CKD is primarily due to cardiovascular disease. Renal transplantation (RT) emerges as the best management alternative for patients with CKD. However, the incidence of acute renal graft dysfunction is 11.8% of the related living donor and 17.4% of the cadaveric donor. Anticardiolipin antibodies (ACAs) or antiphospholipid antibodies (APAs) are important risk factors for acute renal graft dysfunction. The determination of ACA or APA to candidates for RT could serve as prognostic markers of early graft failure and would indicate which patients could benefit from anticoagulant therapy. Cardiolipin is a fundamental molecule that plays an important role in the adequate conformation of the mitochondrial cristae and the correct assembly of the mitochondrial respiratory supercomplexes and other proteins essential for proper mitochondrial function. Cardiolipin undergoes a nonrandom oxidation process by having pronounced specificity unrelated to the polyunsaturation pattern of its acyl groups. Accumulation of hydroxyl derivatives and cardiolipin hydroperoxides has been observed in the affected tissues, and recent studies showed that oxidation of cardiolipin is carried out by a cardiolipin-specific peroxidase activity of cardiolipin-bound cytochrome c. Cardiolipin could be responsible for the proapoptotic production of death signals. Cardiolipin modulates the production of energy and participates in inflammation, mitophagy, and cellular apoptosis. The determination of cardiolipin or its antibodies is an attractive therapeutic, diagnostic target in RT and kidney diseases.
Collapse
|
60
|
Wyss JC, Kumar R, Mikulic J, Schneider M, Mary JL, Aebi JD, Juillerat-Jeanneret L, Golshayan D. Differential Effects of the Mitochondria-Active Tetrapeptide SS-31 (D-Arg-dimethylTyr-Lys-Phe-NH 2) and Its Peptidase-Targeted Prodrugs in Experimental Acute Kidney Injury. Front Pharmacol 2019; 10:1209. [PMID: 31780923 PMCID: PMC6857474 DOI: 10.3389/fphar.2019.01209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
The mitochondria-active tetrapeptide SS-31 can control oxidative tissue damage in kidney diseases. To investigate other potential beneficial nephroprotective effects of SS-31, in vivo murine models of acute tubular injury and glomerular damage were developed. Reduction of acute kidney injury was demonstrated in mice treated with SS-31. The expression of mRNAs involved in acute inflammatory and oxidative stress responses in the diseased kidneys confirmed that SS-31 could regulate these pathways in our in vivo models. Furthermore, ex vivo histoenzymography of mouse kidneys showed that aminopeptidase A (APA), the enzyme involved in the processing of angiotensin (Ang) II to Ang III, was induced in the diseased kidneys, and its activity was inhibited by SS-31. As the renin–angiotensin system (RAS) is a main regulator of kidney functions, the modulation of Ang receptors (ATR) and APA by SS-31 was further investigated using mRNAs extracted from diseased kidneys. Following acute tubular and/or glomerular damage, the expression of the AT1R mRNA was upregulated, which could be selectively downregulated upon SS-31 administration to the animals. At the same time, SS-31 was able to increase the expression of the AT2R, which may contribute to limit renal damage. Consequently, SS-31-based prodrugs were developed as substrates and/or inhibitors for APA and were screened using cells expressing high levels of APA, showing its selective regulation by α-Glu-SS-31. Thus, a link between SS-31 and the RAS opens new therapeutic implications for SS-31 in kidney diseases.
Collapse
Affiliation(s)
- Jean-Christophe Wyss
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Rajesh Kumar
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Josip Mikulic
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Manfred Schneider
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jean-Luc Mary
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Johannes D Aebi
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,University Institute of Pathology, CHUV and UNIL, Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
61
|
Kim SR, Jiang K, Ogrodnik M, Chen X, Zhu XY, Lohmeier H, Ahmed L, Tang H, Tchkonia T, Hickson LJ, Kirkland JL, Lerman LO. Increased renal cellular senescence in murine high-fat diet: effect of the senolytic drug quercetin. Transl Res 2019; 213:112-123. [PMID: 31356770 PMCID: PMC6783353 DOI: 10.1016/j.trsl.2019.07.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022]
Abstract
Obesity and dyslipidemia can be associated with cellular senescence, and may impair kidney function. However, whether senescence contributes to renal dysfunction in these conditions remains unclear. Quercetin is an abundant dietary flavonoid that selectively clears inhibiting PI3K/AKT and p53/p21/serpines and inducing apoptosis. We hypothesized that high-fat-diet-induced obesity causes renal senescence, which would be mitigated by quercetin. C57BL/6J mice fed either standard chow or high-fat diets (HFDs) were treated with quercetin (50 mg/kg) or vehicle 5-days biweekly via oral gavage for 10 weeks. Subsequently, renal function was studied in vivo using magnetic resonance imaging, and renal senescence and histology were evaluated ex vivo. Mice fed with a HFD developed obesity and hypercholesterolemia, whereas renal size remained unchanged. Murine obesity impaired renal function and cortical oxygenation, and induced glomerulomegaly. Renal markers of senescence (eg, expression of p16, p19, and p53) and its secretory phenotype were upregulated in the obese hypercholesterolemic compared to lean mice in renal tubular cells, but attenuated in quercetin-treated murine kidneys, as was renal fibrosis. Quercetin treatment also increased renal cortical oxygenation and decreased plasma creatinine levels in obese mice, whereas body weight and cholesterol levels were unaltered. Therefore, murine obesity and dyslipidemia induce renal tissue senescence and impairs kidney function, which is alleviated by chronic senolytic treatment. These findings implicate senescence in loss of kidney function in murine dyslipidemia and obesity, and support further studies of senolytic therapy in obesity.
Collapse
Affiliation(s)
- Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mikolaj Ogrodnik
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Xiaojun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hannah Lohmeier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Leena Ahmed
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
62
|
Sex differences in transcriptomic profiles in aged kidney cells of renin lineage. Aging (Albany NY) 2019; 10:606-621. [PMID: 29676999 PMCID: PMC5940125 DOI: 10.18632/aging.101416] [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: 03/01/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
Renin expressing cells in the kidney’s juxta-glomeruluar compartment likely also serve as progenitors for adult glomerular cells in disease. Although these cells of renin lineage (CoRL) decrease in number with advancing kidney age, accompanied by less responsiveness to typical stimuli such as ACE-inhibition, mechanisms and the impact of sex as a biological variable with age are not known. Accordingly, labeled CoRL were sorted from individual young (2m) and aged (27m) male and female Ren1cCre|ZsGreen reporter mice, and their transcriptomic profiles analyzed by RNA seq. When both aged female and male mice were combined, there were 48 differentially expressed genes (DEG) compared to young mice. However, when compared to their young sex-matched mice, aged female and male mice had 159 and 503 DEGs respectively. In addition to marked differences in individual genes between aged female and male mice, gene ontology analysis showed major pathway differences by sex. The majority of DEGs in one sex did not significantly change or changed in the opposite direction in the other sex. These results show that in CoRL of advanced age, individual genes and gene ontologies change, but differ between female and male mice, highlighting sex related differences the aging process.
Collapse
|
63
|
Liu D, Jin F, Shu G, Xu X, Qi J, Kang X, Yu H, Lu K, Jiang S, Han F, You J, Du Y, Ji J. Enhanced efficiency of mitochondria-targeted peptide SS-31 for acute kidney injury by pH-responsive and AKI-kidney targeted nanopolyplexes. Biomaterials 2019; 211:57-67. [DOI: 10.1016/j.biomaterials.2019.04.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/28/2022]
|
64
|
Mitochondrial dysfunction in diabetic kidney disease. Clin Chim Acta 2019; 496:108-116. [PMID: 31276635 DOI: 10.1016/j.cca.2019.07.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/26/2022]
Abstract
Although diabetic kidney disease (DKD) is the most common cause of end-stage kidney disease worldwide, the pathogenic mechanisms are poorly understood. There is increasing evidence that mitochondrial dysfunction contributes to the development and progression of DKD. Because the kidney is the organ with the second highest oxygen consumption in our body, it is distinctly sensitive to mitochondrial dysfunction. Mitochondrial dysfunction contributes to the progression of chronic kidney disease irrespective of underlying cause. More importantly, high plasma glucose directly damages renal tubular cells, resulting in a wide range of metabolic and cellular dysfunction. Overproduction of reactive oxygen species (ROS), activation of apoptotic pathway, and defective mitophagy are interlinked mechanisms that play pivotal roles in the progression of DKD. Although renal tubular cells have the highest mitochondrial content, podocytes, mesangial cells, and glomerular endothelial cells may all be affected by diabetes-induced mitochondrial injury. Urinary mitochondrial DNA (mtDNA) is readily detectable and may serve as a marker of mitochondrial damage in DKD. Unfortunately, pharmacologic modulation of mitochondrial dysfunction for the treatment of DKD is still in its infancy. Nonetheless, understanding the pathobiology of mitochondrial dysfunction in DKD would facilitate the development of novel therapeutic strategies.
Collapse
|
65
|
Popov LD. Mitochondrial peptides—appropriate options for therapeutic exploitation. Cell Tissue Res 2019; 377:161-165. [DOI: 10.1007/s00441-019-03049-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
66
|
Zhang LH, Zhu XY, Eirin A, Nargesi AA, Woollard JR, Santelli A, Sun IO, Textor SC, Lerman LO. Early podocyte injury and elevated levels of urinary podocyte-derived extracellular vesicles in swine with metabolic syndrome: role of podocyte mitochondria. Am J Physiol Renal Physiol 2019; 317:F12-F22. [PMID: 31042059 DOI: 10.1152/ajprenal.00399.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome (MetS) is associated with nutrient surplus and kidney hyperfiltration, accelerating chronic renal failure. The potential involvement of podocyte damage in early MetS remains unclear. Mitochondrial dysfunction is an important determinant of renal damage, but whether it contributes to MetS-related podocyte injury remains unknown. Domestic pigs were studied after 16 wk of diet-induced MetS, MetS treated with the mitochondria-targeted peptide elamipretide (ELAM; 0.1 mg·kg-1·day-1 sc) for the last month of diet, and lean controls (n = 6 pigs/group). Glomerular filtration rate (GFR) and renal blood flow (RBF) were measured using multidetector computed tomography, and podocyte and mitochondrial injury were measured by light and electron microscopy. Urinary levels of podocyte-derived extracellular vesicles (pEVs; nephrin positive/podocalyxin positive) were characterized by flow cytometry. Body weight, blood pressure, RBF, and GFR were elevated in MetS. Glomerular size and glomerular injury score were also elevated in MetS and decreased after ELAM treatment. Evidence of podocyte injury, impaired podocyte mitochondria, and foot process width were all increased in MetS but restored with ELAM. The urinary concentration of pEVs was elevated in MetS pigs and directly correlated with renal dysfunction, glomerular injury, and fibrosis and inversely correlated with glomerular nephrin expression. Additionally, pEV numbers were elevated in the urine of obese compared with lean human patients. Early MetS induces podocyte injury and mitochondrial damage, which can be blunted by mitoprotection. Urinary pEVs reflecting podocyte injury might represent early markers of MetS-related kidney disease and a novel therapeutic target.
Collapse
Affiliation(s)
- Li-Hong Zhang
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota.,Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | | | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Adrian Santelli
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - In O Sun
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
67
|
Kim SR, Eirin A, Zhang X, Lerman A, Lerman LO. Mitochondrial Protection Partly Mitigates Kidney Cellular Senescence in Swine Atherosclerotic Renal Artery Stenosis. Cell Physiol Biochem 2019; 52:617-632. [PMID: 30907989 DOI: 10.33594/000000044] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIMS Atherosclerotic renal artery stenosis (ARAS) may cause kidney injury and mitochondrial dysfunction, which is linked to cellular senescence. Elamipretide, a mitochondria-targeted peptide, improves renal function in ARAS, but whether it alleviates senescence is unknown. We hypothesized that elamipretide would reduce senescence stenotic kidney (STK) in ARAS. METHODS Domestic pigs were randomized to control and unilateral ARAS untreated or treated with subcutaneous elamipretide (5d/wk) for 4 weeks starting after 6 weeks of ARAS or sham (n=6 each). After completion of treatment, STK renal blood flow (RBF) and glomerular filtration rate (GFR) were assessed in-vivo using multi-detector computed-tomography. Renal fibrosis and oxidative stress were analyzed in trichrome- and dihydroethidium-stained slides, respectively. Mitochondrial markers involved in the electrontransport chain (COX4, ATP/ADP ratio), biogenesis (PGC1α, PPARα), dynamics (MFN2, DRP1), and mitophagy (parkin, p62) were measured in the kidney using ELISA, western-blot, and immunohistochemistry. Cellular senescence (senescence-associated β-galactosidase and heterochromatin foci, phosphorylated-H2AX, and p16/21/53) and senescence-associated secretory phenotype (SASP; PAI-1, MCP-1, TGFβ, and TNFα) markers were studied by microscopy, quantitative reverse transcription-polymerase chain reaction, and western-blot. RESULTS Blood pressure was elevated whereas STK-RBF and GFR were decreased in ARAS pigs, and tissue scarring was increased. ARAS induced STK cellular senescence and accumulated dysfunctional mitochondria, which were associated with cardiolipin loss, upregulated mitochondrial biogenesis, and defective mitophagy. Elamipretide normalized STK-RBF and GFR, alleviated fibrosis and oxidative stress, and restored mitochondrial cardiolipin, biogenesis, and mitophagy in ARAS, but did not change SASP markers, and attenuated only senescenceassociated β-galactosidase activity and p53 gene expression. CONCLUSION Mitochondrial protection improved renal function and fibrosis in the ARAS STK, but only partly mitigated cellular senescence. This finding suggests that mitochondrial dysfunction may not be a major determinant of cellular senescence in the early stage of ARAS.
Collapse
Affiliation(s)
- Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Xin Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Amir Lerman
- Cardiovascular Diseases, Mayo Clinic, Rochester, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA.,Cardiovascular Diseases, Mayo Clinic, Rochester, USA,
| |
Collapse
|
68
|
Quadri MM, Fatima SS, Che RC, Zhang AH. Mitochondria and Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:501-524. [PMID: 31399982 DOI: 10.1007/978-981-13-8871-2_25] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondria are important organelles in eukaryotic cells and perform a variety of biosynthetic and metabolic functions. Many human diseases are closely related to mitochondrial dysfunction. Kidney is an organ with high-energy requirements, which is distributed with a large number of mitochondria. Mitochondrial dysfunction plays a crucial role in the pathogenesis of kidney disease, and studies have shown that mitochondrial dysfunction is involved in the physiological process of renal fibrosis. This review introduced the biogenesis and pathophysiology of mitochondria, illustrated the involvement of mitochondrial dysfunction in renal fibrosis based on various kinds of cells, and finally summarized the latest mitochondria-targeted therapies.
Collapse
Affiliation(s)
| | - Syeda-Safoorah Fatima
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ruo-Chen Che
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
69
|
Szeto HH, Liu S. Cardiolipin-targeted peptides rejuvenate mitochondrial function, remodel mitochondria, and promote tissue regeneration during aging. Arch Biochem Biophys 2018; 660:137-148. [DOI: 10.1016/j.abb.2018.10.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/13/2018] [Accepted: 10/18/2018] [Indexed: 12/21/2022]
|
70
|
Rovira-Llopis S, Apostolova N, Bañuls C, Muntané J, Rocha M, Victor VM. Mitochondria, the NLRP3 Inflammasome, and Sirtuins in Type 2 Diabetes: New Therapeutic Targets. Antioxid Redox Signal 2018; 29:749-791. [PMID: 29256638 DOI: 10.1089/ars.2017.7313] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Type 2 diabetes mellitus and hyperglycemia can lead to the development of comorbidities such as atherosclerosis and microvascular/macrovascular complications. Both type 2 diabetes and its complications are related to mitochondrial dysfunction and oxidative stress. Type 2 diabetes is also a chronic inflammatory condition that leads to inflammasome activation and the release of proinflammatory mediators, including interleukins (ILs) IL-1β and IL-18. Moreover, sirtuins are energetic sensors that respond to metabolic load, which highlights their relevance in metabolic diseases, such as type 2 diabetes. Recent Advances: Over the past decade, great progress has been made in clarifying the signaling events regulated by mitochondria, inflammasomes, and sirtuins. Nod-like receptor family pyrin domain containing 3 (NLRP3) is the best characterized inflammasome, and the generation of oxidant species seems to be critical for its activation. NLRP3 inflammasome activation and altered sirtuin levels have been observed in type 2 diabetes. Critical Issue: Despite increasing evidence of the relationship between the NLRP3 inflammasome, mitochondrial dysfunction, and oxidative stress and of their participation in type 2 diabetes physiopathology, therapeutic strategies to combat type 2 diabetes that target NLRP3 inflammasome and sirtuins are yet to be consolidated. FUTURE DIRECTIONS In this review article, we attempt to provide an overview of the existing literature concerning the crosstalk between mitochondrial impairment and the inflammasome, with particular attention to cellular and mitochondrial redox metabolism and the potential role of the NLRP3 inflammasome and sirtuins in the pathogenesis of type 2 diabetes. In addition, we discuss potential targets for therapeutic intervention based on these molecular interactions. Antioxid. Redox Signal. 29, 749-791.
Collapse
Affiliation(s)
- Susana Rovira-Llopis
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Nadezda Apostolova
- 2 Department of Pharmacology, University of Valencia , Valencia, Spain .,4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Celia Bañuls
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jordi Muntané
- 3 Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville , Seville, Spain .,4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Milagros Rocha
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain .,4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Victor M Victor
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain .,4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain .,5 Department of Physiology, University of Valencia , Valencia, Spain
| |
Collapse
|
71
|
Sabbah HN, Gupta RC, Singh-Gupta V, Zhang K, Lanfear DE. Abnormalities of Mitochondrial Dynamics in the Failing Heart: Normalization Following Long-Term Therapy with Elamipretide. Cardiovasc Drugs Ther 2018; 32:319-328. [PMID: 29951944 PMCID: PMC6133191 DOI: 10.1007/s10557-018-6805-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Abnormalities of MITO dynamics occur in HF and have been implicated in disease progression. This study describes the broad range abnormalities of mitochondrial (MITO) dynamics in Heart Failure with reduced ejection fraction (HF) and evaluates the effects of long-term therapy with elamipretide (ELAM), a MITO-targeting peptide, on these abnormalities. METHODS Studies were performed in left ventricular tissue from dogs and humans with HF, and were compared with tissue from healthy dogs and healthy donor human hearts. Dogs with HF were randomized to 3 months therapy with ELAM or vehicle. The following were evaluated in dog and human hearts: (1) regulators of MITO biogenesis, including endothelial nitric oxide synthase (eNOS), cyclic guanosine monophosphate (cGMP), and peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α, a transcription factor that drives MITO biogenesis); (2) regulators of MITO fission and fusion, including fission-1, dynamin-related protein-1, mitofusion-2, dominant optic atrophy-1, and mitofilin; and (3) determinants of cardiolipin (CL) synthesis and remodeling, including CL synthase-1, tafazzin-1, and acyl-CoA:lysocardiolipin acyltransferase-1. RESULTS The study showed decreased levels of eNOS, cGMP, and PGC-1α in HF (dog and human). Increased levels of fission-associated proteins, decreased levels of fusion-associated proteins, decreased mitofilin, and abnormalities of CL synthesis and remodeling were also observed. In all instances, these maladaptations were normalized following long-term therapy with ELAM. CONCLUSIONS Critical abnormalities of MITO dynamics occur in HF and are normalized following long-term therapy with ELAM. The findings provide support for the continued development of ELAM for the treatment of HF.
Collapse
Affiliation(s)
- Hani N Sabbah
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Ramesh C Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Vinita Singh-Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Kefei Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - David E Lanfear
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| |
Collapse
|
72
|
Mitochondrial bioenergetics, redox state, dynamics and turnover alterations in renal mass reduction models of chronic kidney diseases and their possible implications in the progression of this illness. Pharmacol Res 2018; 135:1-11. [PMID: 30030169 DOI: 10.1016/j.phrs.2018.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/13/2023]
Abstract
Nowadays, chronic kidney disease (CKD) is considered a worldwide public health problem. CKD is a term used to describe a set of pathologies that structurally and functionally affect the kidney, it is mostly characterized by the progressive loss of kidney function. Current therapeutic approaches are insufficient to avoid the development of this disease, which highlights the necessity of developing new strategies to reverse or at least delay CKD progression. Kidney is highly dependent on mitochondrial homeostasis and function, consequently, the idea that mitochondrial pathologies could play a pivotal role in the genesis and development of kidney diseases has risen. Although many research groups have recently published studies of mitochondrial function in acute kidney disease models, the existing information about CKD is still limited, especially in renal mass reduction (RMR) models. This paper focuses on reviewing current experimental information about the bioenergetics, dynamics (fission and fusion processes), turnover (mitophagy and biogenesis) and redox mitochondrial alterations in RMR, to discuss and integrate the mitochondrial changes triggered by nephron loss, as well as its relationship with loss of kidney function in CKD, in these models. Understanding these mechanisms would allow us to design new therapies that target these mitochondrial alterations.
Collapse
|
73
|
Saad A, Herrmann SMS, Eirin A, Ferguson CM, Glockner JF, Bjarnason H, McKusick MA, Misra S, Lerman LO, Textor SC. Phase 2a Clinical Trial of Mitochondrial Protection (Elamipretide) During Stent Revascularization in Patients With Atherosclerotic Renal Artery Stenosis. Circ Cardiovasc Interv 2018; 10:CIRCINTERVENTIONS.117.005487. [PMID: 28916603 DOI: 10.1161/circinterventions.117.005487] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/24/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Atherosclerotic renal artery stenosis reduces renal blood flow (RBF) and amplifies stenotic kidney hypoxia. Revascularization with percutaneous transluminal renal angioplasty (PTRA) and stenting often fails to recover renal function, possibly because of ischemia/reperfusion injury developing after PTRA. Elamipretide is a mitochondrial-targeted peptide that binds to cardiolipin and stabilizes mitochondrial function. We tested the hypothesis that elamipretide plus PTRA would improve renal function, oxygenation, and RBF in patients with atherosclerotic renal artery stenosis undergoing PTRA. METHODS AND RESULTS Inpatient studies were performed in patients with severe atherosclerotic renal artery stenosis scheduled for PTRA. Patients were treated before and during PTRA with elamipretide (0.05 mg/kg per hour intravenous infusion, n=6) or placebo (n=8). Stenotic kidney cortical/medullary perfusion and RBF were measured using contrast-enhanced multidetector CT, and renal oxygenation by 3-T blood oxygen level-dependent magnetic resonance imaging before and 3 months after PTRA. Age and basal glomerular filtration rate did not differ between groups. Blood oxygen level-dependent imaging demonstrated increased fractional hypoxia 24 hours after angiography and stenting in placebo (+47%) versus elamipretide (-6%). These were reverted to baseline 3 months later. Stenotic kidney RBF rose (202±29-262±115 mL/min; P=0.04) 3 months after PTRA in the elamipretide-treated group only. Over 3 months, systolic blood pressure decreased, and estimated glomerular filtration rate increased (P=0.003) more in the elamipretide group than in the placebo group (P=0.11). CONCLUSIONS Adjunctive elamipretide during PTRA was associated with attenuated postprocedural hypoxia, increased RBF, and improved kidney function in this pilot trial. These data support a role for targeted mitochondrial protection to minimize procedure-associated ischemic injury and to improve outcomes of revascularization for human atherosclerotic renal artery stenosis. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01755858.
Collapse
Affiliation(s)
- Ahmed Saad
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Sandra M S Herrmann
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Alfonso Eirin
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Christopher M Ferguson
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - James F Glockner
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Haraldur Bjarnason
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Michael A McKusick
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Sanjay Misra
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN
| | - Stephen C Textor
- From the Division of Nephrology and Hypertension (A.S., S.M.S.H., A.E., C.M.F., L.O.L., S.C.T.) and Department of Radiology (J.F.G., H.B., M.A.M., S.M.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
74
|
Gioscia-Ryan RA, Battson ML, Cuevas LM, Eng JS, Murphy MP, Seals DR. Mitochondria-targeted antioxidant therapy with MitoQ ameliorates aortic stiffening in old mice. J Appl Physiol (1985) 2018; 124:1194-1202. [PMID: 29074712 PMCID: PMC6008077 DOI: 10.1152/japplphysiol.00670.2017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
Aortic stiffening is a major independent risk factor for cardiovascular diseases, cognitive dysfunction, and other chronic disorders of aging. Mitochondria-derived reactive oxygen species are a key source of arterial oxidative stress, which may contribute to arterial stiffening by promoting adverse structural changes-including collagen overabundance and elastin degradation-and enhancing inflammation, but the potential for mitochondria-targeted therapeutic strategies to ameliorate aortic stiffening with primary aging is unknown. We assessed aortic stiffness [pulse-wave velocity (aPWV)], ex vivo aortic intrinsic mechanical properties [elastic modulus (EM) of collagen and elastin regions], and aortic protein expression in young (~6 mo) and old (~27 mo) male C57BL/6 mice consuming normal drinking water (YC and OC) or water containing mitochondria-targeted antioxidant MitoQ (250 µM; YMQ and OMQ) for 4 wk. Both baseline and postintervention aPWV values were higher in OC vs. YC (post: 482 ± 21 vs. 420 ± 5 cm/s, P < 0.05). MitoQ had no effect in young mice but decreased aPWV in old mice (OMQ, 426 ± 20, P < 0.05 vs. OC). MitoQ did not affect age-associated increases in aortic collagen-region EM, collagen expression, or proinflammatory cytokine expression, but partially attenuated age-associated decreases in elastin region EM and elastin expression. Our results demonstrate that MitoQ reverses in vivo aortic stiffness in old mice and suggest that mitochondria-targeted antioxidants may represent a novel, promising therapeutic strategy for decreasing aortic stiffness with primary aging and, possibly, age-related clinical disorders in humans. The destiffening effects of MitoQ treatment may be at least partially mediated by attenuation/reversal of age-related aortic elastin degradation. NEW & NOTEWORTHY We show that 4 wk of treatment with the mitochondria-specific antioxidant MitoQ in mice completely reverses the age-associated elevation in aortic stiffness, assessed as aortic pulse-wave velocity. The destiffening effects of MitoQ treatment may be at least partially mediated by attenuation of age-related aortic elastin degradation. Our results suggest that mitochondria-targeted therapeutic strategies may hold promise for decreasing arterial stiffening with aging in humans, possibly decreasing the risk of many chronic age-related clinical disorders.
Collapse
Affiliation(s)
- Rachel A Gioscia-Ryan
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Micah L Battson
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Lauren M Cuevas
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Jason S Eng
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge , Cambridge , United Kingdom
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| |
Collapse
|
75
|
Lee HJ, Feliers D, Barnes JL, Oh S, Choudhury GG, Diaz V, Galvan V, Strong R, Nelson J, Salmon A, Kevil CG, Kasinath BS. Hydrogen sulfide ameliorates aging-associated changes in the kidney. GeroScience 2018; 40:163-176. [PMID: 29717417 PMCID: PMC5964063 DOI: 10.1007/s11357-018-0018-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/09/2018] [Indexed: 01/05/2023] Open
Abstract
Aging is associated with replacement of normal kidney parenchyma by fibrosis. Because hydrogen sulfide (H2S) ameliorates kidney fibrosis in disease models, we examined its status in the aging kidney. In the first study, we examined kidney cortical H2S metabolism and signaling pathways related to synthesis of proteins including matrix proteins in young and old male C57BL/6 mice. In old mice, increase in renal cortical content of matrix protein involved in fibrosis was associated with decreased H2S generation and AMPK activity, and activation of insulin receptor (IR)/IRS-2-Akt-mTORC1-mRNA translation signaling axis that can lead to increase in protein synthesis. In the second study, we randomized 18-19 month-old male C57BL/6 mice to receive 30 μmol/L sodium hydrosulfide (NaHS) in drinking water vs. water alone (control) for 5 months. Administration of NaHS increased plasma free sulfide levels. NaHS inhibited the increase in kidney cortical content of matrix proteins involved in fibrosis and ameliorated glomerulosclerosis. NaHS restored AMPK activity and inhibited activation of IR/IRS-2-Akt-mTORC1-mRNA translation axis. NaHS inhibited age-related increase in kidney cortical content of p21, IL-1β, and IL-6, components of the senescence-associated secretory phenotype. NaHS abolished increase in urinary albumin excretion seen in control mice and reduced serum cystatin C levels suggesting improved glomerular clearance function. We conclude that aging-induced changes in the kidney are associated with H2S deficiency. Administration of H2S ameliorates aging-induced kidney changes probably by inhibiting signaling pathways leading to matrix protein synthesis.
Collapse
Affiliation(s)
- Hak Joo Lee
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA
| | - Denis Feliers
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA
| | - Jeffrey L. Barnes
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA ,South Texas Veterans Health Care System, San Antonio, TX USA
| | - Sae Oh
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA
| | - Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA ,South Texas Veterans Health Care System, San Antonio, TX USA
| | - Vivian Diaz
- Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA
| | - Veronica Galvan
- South Texas Veterans Health Care System, San Antonio, TX USA ,Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA
| | - Randy Strong
- South Texas Veterans Health Care System, San Antonio, TX USA ,Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA
| | - James Nelson
- Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA
| | - Adam Salmon
- South Texas Veterans Health Care System, San Antonio, TX USA ,Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA ,Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX USA
| | | | - Balakuntalam S. Kasinath
- Department of Medicine, University of Texas Health San Antonio, 7703, Floyd Curl Drive, MC7882, San Antonio, TX 78229 USA ,South Texas Veterans Health Care System, San Antonio, TX USA ,Barshop Institute for Longevity and Aging Studies, University of Texas Health, San Antonio, TX USA
| |
Collapse
|
76
|
Valentijn FA, Falke LL, Nguyen TQ, Goldschmeding R. Cellular senescence in the aging and diseased kidney. J Cell Commun Signal 2017; 12:69-82. [PMID: 29260442 PMCID: PMC5842195 DOI: 10.1007/s12079-017-0434-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022] Open
Abstract
The program of cellular senescence is involved in both the G1 and G2 phase of the cell cycle, limiting G1/S and G2/M progression respectively, and resulting in prolonged cell cycle arrest. Cellular senescence is involved in normal wound healing. However, multiple organs display increased senescent cell numbers both during natural aging and after injury, suggesting that senescent cells can have beneficial as well as detrimental effects in organismal aging and disease. Also in the kidney, senescent cells accumulate in various compartments with advancing age and renal disease. In experimental studies, forced apoptosis induction through the clearance of senescent cells leads to better preservation of kidney function during aging. Recent groundbreaking studies demonstrate that senescent cell depletion through INK-ATTAC transgene-mediated or cell-penetrating FOXO4-DRI peptide induced forced apoptosis, reduced age-associated damage and dysfunction in multiple organs, in particular the kidney, and increased performance and lifespan. Senescence is also involved in oncology and therapeutic depletion of senescent cells by senolytic drugs has been studied in experimental and human cancers. Although studies with senolytic drugs in models of kidney injury are lacking, their dose limiting side effects on other organs suggest that targeted delivery might be needed for successful application of senolytic drugs for treatment of kidney disease. In this review, we discuss (i) current understanding of the mechanisms and associated pathways of senescence, (ii) evidence of senescence occurrence and causality with organ injury, and (iii) therapeutic strategies for senescence depletion (senotherapy) including targeting, all in the context of renal aging and disease.
Collapse
Affiliation(s)
- F A Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - L L Falke
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
- Department of Internal Medicine, Diakonessenhuis, Utrecht, The Netherlands
| | - T Q Nguyen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands.
| |
Collapse
|
77
|
Compound effects of aging and experimental FSGS on glomerular epithelial cells. Aging (Albany NY) 2017; 9:524-546. [PMID: 28222042 PMCID: PMC5361679 DOI: 10.18632/aging.101176] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/09/2017] [Indexed: 12/27/2022]
Abstract
Advanced age portends a poorer prognosis in FSGS. To understand the impact of age on glomerular podocytes and parietal epithelial cells (PECs), experimental FSGS was induced in 3m-old mice (20-year old human age) and 27m-old mice (78-year old human age) by abruptly depleting podocytes with a cytopathic anti-podocyte antibody. Despite similar binding of the disease-inducing antibody, podocyte density was lower in aged FSGS mice compared to young FSGS mice. Activated PEC density was higher in aged versus young FSGS mice, as was the percentage of total activated PECs. Additionally, the percentage of glomeruli containing PECs with evidence of phosphorylated ERK and EMT was higher in aged FSGS mice. Extracellular matrix, measured by collagen IV and silver staining, was higher in aged FSGS mice along Bowman's capsule. However, collagen IV accumulation in the glomerular tufts alone and in glomeruli with both tuft and Bowman's capsule accumulation were similar in young FSGS and aged FSGS mice. Thus, the major difference in collagen IV staining in FSGS was along Bowman's capsule in aged mice. The significant differences in podocytes, PECs and extracellular matrixaccumulation between young mice and old mice with FSGS might explain the differences in outcomes in FSGS based on age.
Collapse
|
78
|
Abstract
AKI is associated with high morbidity and mortality, and it predisposes to the development and progression of CKD. Novel strategies that minimize AKI and halt the progression of CKD are urgently needed. Normal kidney function involves numerous different cell types, such as tubular epithelial cells, endothelial cells, and podocytes, working in concert. This delicate balance involves many energy-intensive processes. Fatty acids are the preferred energy substrates for the kidney, and defects in fatty acid oxidation and mitochondrial dysfunction are universally involved in diverse causes of AKI and CKD. This review provides an overview of ATP production and energy demands in the kidney and summarizes preclinical and clinical evidence of mitochondrial dysfunction in AKI and CKD. New therapeutic strategies targeting mitochondria protection and cellular bioenergetics are presented, with emphasis on those that have been evaluated in animal models of AKI and CKD. Targeting mitochondrial function and cellular bioenergetics upstream of cellular damage may offer advantages compared with targeting downstream inflammatory and fibrosis processes.
Collapse
Affiliation(s)
- Hazel H Szeto
- Mitochondrial Therapeutics Consulting, New York, New York
| |
Collapse
|