1
|
Zhang Z, Wei Z, Gao L. Impact of lifestyle on diabetic nephropathy in aged 18-64 years: A population-based cross-sectional analysis from NHANES 2007-2018. J Diabetes Investig 2025. [PMID: 40348607 DOI: 10.1111/jdi.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/23/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The relationship between lifestyle and overall health has garnered significant attention. This study aimed to evaluate the association between lifestyle factors and diabetic nephropathy (DN) in adults aged 18-64 years. MATERIALS AND METHODS We conducted a cross-sectional study involving 2,389 participants from the 2007-2018 National Health and Nutrition Examination Survey (NHANES). The use of the Healthy Eating Index (HEI), Metabolic Equivalent of Task (MET), sleeping duration, smoking, and drinking status as indicators to assess lifestyle. Logistic regression analyses and restricted cubic splines were used to analyze the results. A subgroup analysis was performed to identify any variations. RESULTS Logistic regression analysis indicates that poor HEI and sleep duration but not physical activity are associated with an increased risk of DN. Subgroup analyses revealed significant interactions between HEI and age, blood pressure, HbA1c, and lipid control; MET interacted with blood pressure and HbA1c control; Sleeping duration interacted with age, smoking, blood pressure, HbA1c, and lipid control; Smoking interacted with age, blood pressure control, and lipid control; Drinking interacted with blood pressure control. Moreover, restricted cubic splines indicated that with increasing HEI, the prevalence of DN tended to decrease. However, the associations between the other two lifestyle factors (MET level and sleeping duration) and DN were all U-shaped. CONCLUSIONS Lifestyle factors are closely associated with diabetic nephropathy. Both unhealthy eating habits and inadequate or excessive sleeping duration and physical activity contribute to an increased risk of diabetic nephropathy, whereas no statistically significant association is observed in smoking and drinking.
Collapse
Affiliation(s)
- Zhijun Zhang
- Department of Endocrinology and Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaoyinling Wei
- Department of Endocrinology and Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Gao
- Department of Endocrinology and Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Bai Z, Liang J, Nie Y, Wang S, Chang D. The mediating role of the TyG index in the relationship between circadian syndrome and cancer among middle-aged and elderly Chinese. BMC Cancer 2025; 25:431. [PMID: 40065285 PMCID: PMC11895363 DOI: 10.1186/s12885-025-13816-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Circadian Syndrome (CircS) is a significant marker of metabolic imbalance and has been linked to various chronic diseases. However, its relationship with cancer risk remains underexplored. This research aims to explore the relationship between CircS and cancer, while also assessing the possible mediating role of the triglyceride glucose (TyG) index. METHODS Baseline data from the 2011 China Health and Retirement Longitudinal Study (CHARLS) and follow-up data from 2015 were analyzed, including participants' sociodemographic characteristics, health behaviors, and metabolic indicators. Linear regression, mediation analysis, and logistic regression were employed to explore relationships between CircS, cancer risk, and the TyG index, with a dose-response analysis conducted on TyG index and cancer risk. RESULTS Among 7,864 middle-aged and elderly participants, CircS was significantly and positively associated with cancer risk (r = 0.17, P < 0.001). The TyG index showed a significant correlation with both CircS (r = 0.52, P < 0.001) and cancer (r = 0.15, P < 0.001). Mediation modeling indicated that the TyG index partially mediated the association between CircS and cancer, accounting for 23% of this relationship. Additionally, a significant nonlinear dose-response relationship was observed between the TyG index and cancer risk (Pnonlinear = 0.0024). CONCLUSION Circadian syndrome is associated with increased cancer risk, with the TyG index partially mediating this relationship.
Collapse
Affiliation(s)
- Zilong Bai
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Jiale Liang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Yuanhua Nie
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Shilong Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Dongmin Chang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China.
| |
Collapse
|
3
|
Jamadar A, Ward CJ, Remadevi V, Varghese MM, Pabla NS, Gumz ML, Rao R. Circadian Clock Disruption and Growth of Kidney Cysts in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2025; 36:378-392. [PMID: 39401086 PMCID: PMC11888963 DOI: 10.1681/asn.0000000528] [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: 05/24/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Key Points Lack of Bmal1 , a circadian clock protein in renal collecting ducts disrupted the clock and increased cyst growth and fibrosis in an autosomal dominant polycystic kidney disease mouse model. Bmal1 gene deletion increased cell proliferation by increasing lipogenesis in kidney cells. Thus, circadian clock disruption could be a risk factor for accelerated disease progression in patients with autosomal dominant polycystic kidney disease. Background Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 and PKD2 genes and often progresses to kidney failure. ADPKD progression is not uniform among patients, suggesting that factors secondary to the PKD1/2 gene mutation could regulate the rate of disease progression. Here, we tested the effect of circadian clock disruption on ADPKD progression. Circadian rhythms are regulated by cell-autonomous circadian clocks composed of clock proteins. BMAL1 is a core constituent of the circadian clock. Methods To disrupt the circadian clock, we deleted Bmal1 gene in the renal collecting ducts of the Pkd1 RC/RC (RC/RC) mouse model of ADPKD (RC/RC;Bmal1 f/f;Pkhd1 cre, called double knockout [DKO] mice) and in Pkd1 knockout mouse inner medullary collecting duct cells (Pkd1Bmal1 KO mouse renal inner medullary collecting duct cells). Only male mice were used. Results Human nephrectomy ADPKD kidneys showed altered clock gene expression when compared with normal control human kidneys. When compared with RC/RC kidneys, DKO kidneys showed significantly altered clock gene expression, increased cyst growth, cell proliferation, apoptosis, and fibrosis. DKO kidneys also showed increased lipogenesis and cholesterol synthesis–related gene expression and increased tissue triglyceride levels compared with RC/RC kidneys. Similarly, in vitro , Pkd1Bmal1 KO cells showed altered clock genes, increased lipogenesis and cholesterol synthesis–related genes, and reduced fatty acid oxidation–related gene expression compared with Pkd1KO cells. The Pkd1Bmal1 KO cells showed increased cell proliferation compared with Pkd1KO cells, which was rescued by pharmacological inhibition of lipogenesis. Conclusions Renal collecting duct–specific Bmal1 gene deletion disrupted the circadian clock and triggered accelerated ADPKD progression by altering lipid metabolism–related gene expression.
Collapse
Affiliation(s)
- Abeda Jamadar
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Christopher J. Ward
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Viji Remadevi
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Meekha M. Varghese
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| | - Navjot S. Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Michelle L. Gumz
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Physiology and Aging, Department of Medicine, University of Florida, Gainesville, Florida
| | - Reena Rao
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
He X, Wang Z, Cheng L, Wang H, Sun Y. An antagonistic role of clock genes and lima1 in kidney regeneration. Commun Biol 2025; 8:29. [PMID: 39789202 PMCID: PMC11718004 DOI: 10.1038/s42003-025-07455-8] [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: 04/27/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
The circadian clock genes are known important for kidney development, maturation and physiological functions. However, whether and how they play a role in renal regeneration remain elusive. Here, by using the single cell RNA-sequencing (scRNA-seq) technology, we investigated the dynamic gene expression profiles and cell states after acute kidney injury (AKI) by gentamicin treatment in zebrafish. The core clock genes such as per1/2 and nr1d1, which encode transcriptional repressors of the circadian system, are strongly induced in the proximal tubule epithelial cells (PTECs). By generating mutant zebrafish lines, we show that per1a and nr1d1 are required for proper renal regeneration, by facilitating the expression of renal progenitor cell (RPC) genes. In per1a and nr1d1 mutants, the expression of RPC genes and the number of RPCs were decreased, resulting in a marked delay in nephron regeneration. lima1a, which encodes a cytoskeleton binding protein that functions to negatively regulate epithelial to mesenchymal transition (EMT), is identified as the direct target of the clock proteins. Down-regulation of lima1a is associated with enhanced EMT, increased expression of cell migration- and RPC markers, and accelerated nephron regeneration. We propose that per1a and nr1d1 are important for the formation of nephrongenic RPCs by repressing lima1a. Our findings using zebrafish provide important insights into the roles of the clock genes in kidney repair.
Collapse
Affiliation(s)
- Xian He
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, 430072, Wuhan, China
- The Innovation of Seed Design, Chinese Academy of Sciences, 430072, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Ziming Wang
- Center for Circadian Clocks, and School of Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, China
| | - Linxi Cheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, 430072, Wuhan, China
- The Innovation of Seed Design, Chinese Academy of Sciences, 430072, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Han Wang
- Center for Circadian Clocks, and School of Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, China
| | - Yuhua Sun
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, 430072, Wuhan, China.
- The Innovation of Seed Design, Chinese Academy of Sciences, 430072, Wuhan, China.
- University of Chinese Academy of Sciences, 100049, Beijing, PR China.
- Hubei Hongshan Laboratory, 430070, Wuhan, China.
| |
Collapse
|
5
|
Yang S, Ye Z, Chen L, Zhou X, Li W, Cheng F. Circadian Clock Gene Bmal1: A Molecular Bridge from AKI to CKD. Biomolecules 2025; 15:77. [PMID: 39858471 PMCID: PMC11762869 DOI: 10.3390/biom15010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) represent two frequently observed clinical conditions. AKI is characterized by an abrupt decrease in glomerular filtration rate (GFR), generally associated with elevated serum creatinine (sCr), blood urea nitrogen (BUN), and electrolyte imbalances. This condition usually persists for approximately a week, causing a transient reduction in kidney function. If these abnormalities continue beyond 90 days, the condition is redefined as chronic kidney disease (CKD) or may advance to end-stage renal disease (ESRD). Recent research increasingly indicates that maladaptive repair mechanisms after AKI significantly contribute to the development of CKD. Thus, implementing early interventions to halt the progression from AKI to CKD has the potential to markedly improve patient outcomes. Although considerable research has been conducted, the exact mechanisms linking AKI to CKD are complex, and effective treatments remain limited. Kidney function is influenced by circadian rhythms, with the circadian gene Bmal1 being vital in managing these cycles. Recent research indicates that Bmal1 is significantly involved in the progression of both AKI and CKD. In this study, we conducted a retrospective analysis of Bmal1's role in AKI and CKD, reviewed recent research advancements, and investigated how Bmal1 influences the pathological mechanisms underlying the progression from AKI to CKD. Additionally, we highlighted gaps in the existing research and examined the potential of Bmal1 as a therapeutic target in kidney disease management. This work aims to provide meaningful insights for future studies on the role of the circadian gene Bmal1 in the transition from AKI to CKD, with the goal of identifying therapeutic approaches to mitigate kidney disease progression.
Collapse
Affiliation(s)
- Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Lijia Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| |
Collapse
|
6
|
Ali R, Zhen Y, Zanna X, Lin J, Zhang C, Ma J, Zhong Y, Husien HM, Saleh AA, Wang M. Impact of Circadian Clock PER2 Gene Overexpression on Rumen Epithelial Cell Dynamics and VFA Transport Protein Expression. Int J Mol Sci 2024; 25:12428. [PMID: 39596493 PMCID: PMC11594904 DOI: 10.3390/ijms252212428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The circadian gene PER2 is recognized for its regulatory effects on cell proliferation and lipid metabolism across various non-ruminant cells. This study investigates the influence of PER2 gene overexpression on goat rumen epithelial cells using a constructed pcDNA3.1-PER2 plasmid, assessing its impact on circadian gene expression, cell proliferation, and mRNA levels of short-chain fatty acid (SCFA) transporters, alongside genes related to lipid metabolism, cell proliferation, and apoptosis. Rumen epithelial cells were obtained every four hours from healthy dairy goats (n = 3; aged 1.5 years; average weight 45.34 ± 4.28 kg), cultured for 48 h in vitro, and segregated into control (pcDNA3.1) and overexpressed (pcDNA3.1-PER2) groups, each with four biological replicates. The study examined the potential connection between circadian rhythms and nutrient assimilation in ruminant, including cell proliferation, apoptosis, cell cycle dynamics, and antioxidant activity and the expression of circadian-related genes, VFA transporter genes and regulatory factors. The introduction of the pcDNA3.1-PER2 plasmid drastically elevated PER2 expression levels by 3471.48-fold compared to controls (p < 0.01), confirming effective overexpression. PER2 overexpression resulted in a significant increase in apoptosis rates (p < 0.05) and a notable reduction in cell proliferation at 24 and 48 h post-transfection (p < 0.05), illustrating an inhibitory effect on rumen epithelial cell growth. PER2 elevation significantly boosted the expression of CCND1, WEE1, p21, and p16 (p < 0.05) while diminishing CDK4 expression (p < 0.05). While the general expression of intracellular inflammation genes remained stable, TNF-α expression notably increased. Antioxidant marker levels (SOD, MDA, GSH-Px, CAT, and T-AOC) exhibited no significant change, suggesting no oxidative damage due to PER2 overexpression. Furthermore, PER2 overexpression significantly downregulated AE2, NHE1, MCT1, and MCT4 mRNA expressions while upregulating PAT1 and VH+ ATPase. These results suggest that PER2 overexpression impairs cell proliferation, enhances apoptosis, and modulates VFA transporter-related factors in the rumen epithelium. This study implies that the PER2 gene may regulate VFA absorption through modulation of VFA transporters in rumen epithelial cells, necessitating further research into its specific regulatory mechanisms.
Collapse
Affiliation(s)
- Rahmat Ali
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Yongkang Zhen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Xi Zanna
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Jiaqi Lin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Chong Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Jianjun Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Yuhong Zhong
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Hosameldeen Mohamed Husien
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| | - Ahmad A. Saleh
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China;
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (R.A.); (H.M.H.)
| |
Collapse
|
7
|
Jamadar A, Ward CJ, Remadevi V, Varghese MM, Pabla NS, Gumz ML, Rao R. Circadian clock disruption and growth of kidney cysts in autosomal dominant polycystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606676. [PMID: 39211074 PMCID: PMC11361200 DOI: 10.1101/2024.08.05.606676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 and PKD2 genes, and often progresses to kidney failure. ADPKD progression is not uniform among patients, suggesting that factors secondary to the PKD1/2 gene mutation could regulate the rate of disease progression. Here we tested the effect of circadian clock disruption on ADPKD progression. Circadian rhythms are regulated by cell-autonomous circadian clocks composed of clock proteins. BMAL1 is a core constituent of the circadian clock. Methods To disrupt the circadian clock, we deleted Bmal1 gene in the renal collecting ducts of the Pkd1 RC/RC (RC/RC) mouse model of ADPKD (RC/RC; Bmal1 f/f ; Pkhd1 cre , called DKO mice), and in Pkd1 knockout mouse inner medullary collecting duct cells ( Pkd1Bmal1 KO mIMCD3 cells). Only male mice were used. Results Human nephrectomy ADPKD kidneys and Pkd1 KO mIMCD3 cells showed reduced Bmal1 gene expression compared to normal controls. When compared to RC/RC kidneys, DKO kidneys showed significantly altered clock gene expression, increased cyst growth, cell proliferation, apoptosis and fibrosis. DKO kidneys also showed increased lipogenesis and cholesterol synthesis-related gene expression, and increased tissue triglyceride levels compared to RC/RC kidneys. Similarly, in vitro, Pkd1Bmal1 KO cells showed altered clock genes, increased lipogenesis and cholesterol synthesis-related genes, and reduced fatty-acid oxidation-related gene expression compared to Pkd1KO cells. The Pkd1Bmal1 KO cells showed increased cell proliferation compared to Pkd1KO cells, which was rescued by pharmacological inhibition of lipogenesis. Conclusion Renal collecting duct specific Bmal1 gene deletion disrupts the circadian clock and triggers accelerated ADPKD progression by altering lipid metabolism-related gene expression. Key points Lack of BMAL1, a circadian clock protein in renal collecting ducts disrupted the clock and increased cyst growth and fibrosis in an ADPKD mouse model.BMAL1 gene deletion increased cell proliferation by increasing lipogenesis in kidney cells.Thus, circadian clock disruption could be a risk factor for accelerated disease progression in patients with ADPKD.
Collapse
|
8
|
Yang Z, Chen L, Huang Y, Dong J, Yan Q, Li Y, Qiu J, Li H, Zhao D, Liu F, Tang D, Dai Y. Proteomic profiling of laser capture microdissection kidneys from diabetic nephropathy patients. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1243:124231. [PMID: 38996754 DOI: 10.1016/j.jchromb.2024.124231] [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: 01/11/2024] [Revised: 05/23/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Diabetic nephropathy (DN) remains the primary cause of end-stage renal disease (ESRD), warranting equal attention and separate analysis of glomerular, tubular, and interstitial lesions in its diagnosis and intervention. This study aims to identify the specific proteomics characteristics of DN, and assess changes in the biological processes associated with DN. 5 patients with DN and 5 healthy kidney transplant donor control individuals were selected for analysis. The proteomic characteristics of glomeruli, renal tubules, and renal interstitial tissue obtained through laser capture microscopy (LCM) were studied using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Significantly, the expression of multiple heat shock proteins (HSPs), tubulins, and heterogeneous nuclear ribonucleoproteins (hnRNPs) in glomeruli and tubules was significantly reduced. Differentially expressed proteins (DEPs) in the glomerulus showed significant enrichment in pathways related to cell junctions and cell movement, including the regulation of actin cytoskeleton and tight junction. DEPs in renal tubules were significantly enriched in glucose metabolism-related pathways, such as glucose metabolism, glycolysis/gluconeogenesis, and the citric acid cycle. Moreover, the glycolysis/gluconeogenesis pathway was a co-enrichment pathway in both DN glomeruli and tubules. Notably, ACTB emerged as the most crucial protein in the protein-protein interaction (PPI) analysis of DEPs in both glomeruli and renal tubules. In this study, we delve into the unique proteomic characteristics of each sub-region of renal tissue. This enhances our understanding of the potential pathophysiological changes in DN, particularly the potential involvement of glycolysis metabolic disorder, glomerular cytoskeleton and cell junctions. These insights are crucial for further research into the identification of disease biomarkers and the pathogenesis of DN.
Collapse
Affiliation(s)
- Zhiqian Yang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Liangmei Chen
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Yingxin Huang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; Department of Nephrology, Xiaolan People's Hospital of Zhongshan, 528400, China
| | - Jingjing Dong
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Qiang Yan
- Department of Organ Transplantation, 924 Hospital, Guilin 541002, China
| | - Ya Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Jing Qiu
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Haitao Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Da Zhao
- The First Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan 232001, Anhui, China
| | - Fanna Liu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China.
| | - Donge Tang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China.
| | - Yong Dai
- Comprehensive Health Industry Research Center, Taizhou Research Institute, Southern University of Science and Technology, Taizhou 317000, China; The First Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan 232001, Anhui, China.
| |
Collapse
|
9
|
Siew K, Nestler KA, Nelson C, D'Ambrosio V, Zhong C, Li Z, Grillo A, Wan ER, Patel V, Overbey E, Kim J, Yun S, Vaughan MB, Cheshire C, Cubitt L, Broni-Tabi J, Al-Jaber MY, Boyko V, Meydan C, Barker P, Arif S, Afsari F, Allen N, Al-Maadheed M, Altinok S, Bah N, Border S, Brown AL, Burling K, Cheng-Campbell M, Colón LM, Degoricija L, Figg N, Finch R, Foox J, Faridi P, French A, Gebre S, Gordon P, Houerbi N, Valipour Kahrood H, Kiffer FC, Klosinska AS, Kubik A, Lee HC, Li Y, Lucarelli N, Marullo AL, Matei I, McCann CM, Mimar S, Naglah A, Nicod J, O'Shaughnessy KM, Oliveira LCD, Oswalt L, Patras LI, Lai Polo SH, Rodríguez-Lopez M, Roufosse C, Sadeghi-Alavijeh O, Sanchez-Hodge R, Paul AS, Schittenhelm RB, Schweickart A, Scott RT, Choy Lim Kam Sian TC, da Silveira WA, Slawinski H, Snell D, Sosa J, Saravia-Butler AM, Tabetah M, Tanuwidjaya E, Walker-Samuel S, Yang X, Yasmin, Zhang H, Godovac-Zimmermann J, Sarder P, Sanders LM, Costes SV, Campbell RAA, Karouia F, Mohamed-Alis V, Rodriques S, Lynham S, Steele JR, Baranzini S, Fazelinia H, Dai Z, Uruno A, Shiba D, Yamamoto M, A C Almeida E, Blaber E, Schisler JC, Eisch AJ, Muratani M, Zwart SR, et alSiew K, Nestler KA, Nelson C, D'Ambrosio V, Zhong C, Li Z, Grillo A, Wan ER, Patel V, Overbey E, Kim J, Yun S, Vaughan MB, Cheshire C, Cubitt L, Broni-Tabi J, Al-Jaber MY, Boyko V, Meydan C, Barker P, Arif S, Afsari F, Allen N, Al-Maadheed M, Altinok S, Bah N, Border S, Brown AL, Burling K, Cheng-Campbell M, Colón LM, Degoricija L, Figg N, Finch R, Foox J, Faridi P, French A, Gebre S, Gordon P, Houerbi N, Valipour Kahrood H, Kiffer FC, Klosinska AS, Kubik A, Lee HC, Li Y, Lucarelli N, Marullo AL, Matei I, McCann CM, Mimar S, Naglah A, Nicod J, O'Shaughnessy KM, Oliveira LCD, Oswalt L, Patras LI, Lai Polo SH, Rodríguez-Lopez M, Roufosse C, Sadeghi-Alavijeh O, Sanchez-Hodge R, Paul AS, Schittenhelm RB, Schweickart A, Scott RT, Choy Lim Kam Sian TC, da Silveira WA, Slawinski H, Snell D, Sosa J, Saravia-Butler AM, Tabetah M, Tanuwidjaya E, Walker-Samuel S, Yang X, Yasmin, Zhang H, Godovac-Zimmermann J, Sarder P, Sanders LM, Costes SV, Campbell RAA, Karouia F, Mohamed-Alis V, Rodriques S, Lynham S, Steele JR, Baranzini S, Fazelinia H, Dai Z, Uruno A, Shiba D, Yamamoto M, A C Almeida E, Blaber E, Schisler JC, Eisch AJ, Muratani M, Zwart SR, Smith SM, Galazka JM, Mason CE, Beheshti A, Walsh SB. Cosmic kidney disease: an integrated pan-omic, physiological and morphological study into spaceflight-induced renal dysfunction. Nat Commun 2024; 15:4923. [PMID: 38862484 PMCID: PMC11167060 DOI: 10.1038/s41467-024-49212-1] [Show More Authors] [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: 01/20/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Missions into Deep Space are planned this decade. Yet the health consequences of exposure to microgravity and galactic cosmic radiation (GCR) over years-long missions on indispensable visceral organs such as the kidney are largely unexplored. We performed biomolecular (epigenomic, transcriptomic, proteomic, epiproteomic, metabolomic, metagenomic), clinical chemistry (electrolytes, endocrinology, biochemistry) and morphometry (histology, 3D imaging, miRNA-ISH, tissue weights) analyses using samples and datasets available from 11 spaceflight-exposed mouse and 5 human, 1 simulated microgravity rat and 4 simulated GCR-exposed mouse missions. We found that spaceflight induces: 1) renal transporter dephosphorylation which may indicate astronauts' increased risk of nephrolithiasis is in part a primary renal phenomenon rather than solely a secondary consequence of bone loss; 2) remodelling of the nephron that results in expansion of distal convoluted tubule size but loss of overall tubule density; 3) renal damage and dysfunction when exposed to a Mars roundtrip dose-equivalent of simulated GCR.
Collapse
Affiliation(s)
- Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK.
| | - Kevin A Nestler
- The Institute for Biomedical Sciences (IBS), The George Washington University, Washington, DC, USA
| | - Charlotte Nelson
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Viola D'Ambrosio
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
- Department of Experimental and Translational Medicine, Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| | - Chutong Zhong
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Zhongwang Li
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
- Centre for Advanced Biomedical Imaging, University College London, London, UK
- Centre for Computational Medicine, University College London, London, UK
| | - Alessandra Grillo
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Elizabeth R Wan
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Vaksha Patel
- Department of Renal Medicine, University College London, London, UK
| | - Eliah Overbey
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - JangKeun Kim
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - Sanghee Yun
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael B Vaughan
- School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Tissue Engineering and Biomaterials Group, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Chris Cheshire
- Bioinformatics and Computational Biology Laboratory, The Francis Crick Institute, London, UK
| | - Laura Cubitt
- Applied Biotechnology Laboratory, The Francis Crick Institute, London, UK
| | - Jessica Broni-Tabi
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | | | - Valery Boyko
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Cem Meydan
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - Peter Barker
- MRC MDU Mouse Biochemistry Laboratory, University of Cambridge, Cambridge, UK
| | - Shehbeel Arif
- Center for Data Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fatemeh Afsari
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Noah Allen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mohammed Al-Maadheed
- Anti-Doping Laboratory Qatar, Doha, Qatar
- Centre of Metabolism and Inflammation, University College London, London, UK
| | - Selin Altinok
- School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nourdine Bah
- Applied Biotechnology Laboratory, The Francis Crick Institute, London, UK
| | - Samuel Border
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Amanda L Brown
- Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keith Burling
- MRC MDU Mouse Biochemistry Laboratory, University of Cambridge, Cambridge, UK
| | - Margareth Cheng-Campbell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - Lorianna M Colón
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Lovorka Degoricija
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Nichola Figg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rebecca Finch
- School of Health, Science and Wellbeing, Staffordshire University, Stoke-on-Trent, UK
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - Pouya Faridi
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Alison French
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Samrawit Gebre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Peter Gordon
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Nadia Houerbi
- Physiology, Biophysics & Systems Biology, Weill Cornell Medical College, New York, NY, USA
| | - Hossein Valipour Kahrood
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Monash Bioinformatics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Frederico C Kiffer
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aleksandra S Klosinska
- Division of Experimental Medicine & Immunotherapeutics (EMIT), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Angela Kubik
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Han-Chung Lee
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Nicholas Lucarelli
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Anthony L Marullo
- School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Irina Matei
- Cornell Center for Immunology, Cornell University, Ithaca, NY, USA
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Colleen M McCann
- Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sayat Mimar
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Ahmed Naglah
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Jérôme Nicod
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine & Immunotherapeutics (EMIT), Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Leah Oswalt
- Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - San-Huei Lai Polo
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | | | - Candice Roufosse
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | | | | | - Anindya S Paul
- Department of Medicine-Nephrology & Intelligent Critical Care Center, University of Florida, Gainesville, FL, USA
| | - Ralf Bernd Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Annalise Schweickart
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Terry Chin Choy Lim Kam Sian
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Willian A da Silveira
- School of Health, Science and Wellbeing, Staffordshire University, Stoke-on-Trent, UK
- International Space University, 67400, Illkirch-Graffenstaden, France
| | - Hubert Slawinski
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Daniel Snell
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Julio Sosa
- University Health Network, Toronto, ON, Canada
| | | | - Marshall Tabetah
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA
| | - Erwin Tanuwidjaya
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, University College London, London, UK
- Centre for Computational Medicine, University College London, London, UK
| | | | - Yasmin
- Division of Experimental Medicine & Immunotherapeutics (EMIT), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Haijian Zhang
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | - Pinaki Sarder
- Department of Medicine-Quantitative Health Section, University of Florida, Gainesville, FL, USA
- Departments of Biomedical Engineering and Electrical and Computer Engineering, University of Florida, Gainesville, FL, USA
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Robert A A Campbell
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Fathi Karouia
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Space Research Within Reach, San Francisco, CA, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vidya Mohamed-Alis
- Anti-Doping Laboratory Qatar, Doha, Qatar
- Centre of Metabolism and Inflammation, University College London, London, UK
| | - Samuel Rodriques
- Applied Biotechnology Laboratory, The Francis Crick Institute, London, UK
| | | | - Joel Ricky Steele
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sergio Baranzini
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Hossein Fazelinia
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Zhongquan Dai
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Dai Shiba
- Mouse Epigenetics Project, ISS/Kibo experiment, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Ibaraki, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Ibaraki, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Eduardo A C Almeida
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Elizabeth Blaber
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
- Stanley Center for Psychiatric Research, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jonathan C Schisler
- Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Masafumi Muratani
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sara R Zwart
- Department of Preventative Medicine and Community Health, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medical College, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Broad Institute, Cambridge, MA, USA
- Space Biosciences Division, Universities Space Research Association (USRA), Washington, DC, USA
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK.
| |
Collapse
|
10
|
Wang L, Tian H, Wang H, Mao X, Luo J, He Q, Wen P, Cao H, Fang L, Zhou Y, Yang J, Jiang L. Disrupting circadian control of autophagy induces podocyte injury and proteinuria. Kidney Int 2024; 105:1020-1034. [PMID: 38387504 DOI: 10.1016/j.kint.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
The circadian clock influences a wide range of biological process and controls numerous aspects of physiology to adapt to the daily environmental changes caused by Earth's rotation. The kidney clock plays an important role in maintaining tubular function, but its effect on podocytes remains unclear. Here, we found that podocytes expressed CLOCK proteins, and that 2666 glomerular gene transcripts (13.4%), including autophagy related genes, had 24-hour circadian rhythms. Deletion of Clock in podocytes resulted in 1666 gene transcripts with the loss of circadian rhythm including autophagy genes. Podocyte-specific Clock knockout mice at age three and eight months showed deficient autophagy, loss of podocytes and increased albuminuria. Chromatin immunoprecipitation (ChIP) sequence analysis indicated autophagy related genes were targets of CLOCK in podocytes. ChIP-PCR further confirmed Clock binding to the promoter regions of Becn1 and Atg12, two autophagy related genes. Furthermore, the association of CLOCK regulated autophagy with chronic sleep fragmentation and diabetic kidney disease was analyzed. Chronic sleep fragmentation resulted in the loss of glomerular Clock rhythm, inhibition of podocyte autophagy, and proteinuria. Rhythmic oscillations of Clock also disappeared in high glucose treated podocytes and in glomeruli from diabetic mice. Finally, circadian differences in podocyte autophagy were also abolished in diabetic mice. Deletion Clock in podocytes aggravated podocyte injury and proteinuria in diabetic mice. Thus, our findings demonstrate that clock-dependent regulation of autophagy may be essential for podocyte survival. Hence. loss of circadian controlled autophagy may play an important role in podocyte injury and proteinuria.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Han Tian
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyan Wang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Mao
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Luo
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingyun He
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Wen
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongdi Cao
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Fang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Yang Zhou
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Junwei Yang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lei Jiang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Shen J, Ying L, Wu J, Fang Y, Zhou W, Qi C, Gu L, Mou S, Yan Y, Tian M, Ni Z, Che X. Integrative ATAC-seq and RNA-seq analysis associated with diabetic nephropathy and identification of novel targets for treatment by dapagliflozin. Cell Biochem Funct 2024; 42:e3943. [PMID: 38379015 DOI: 10.1002/cbf.3943] [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: 07/22/2023] [Revised: 12/01/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024]
Abstract
Dapagliflozin (DAPA) are clinically effective in improving diabetic nephropathy (DN). However, whether and how chromatin accessibility changed by DN responds to DAPA treatment is unclear. Therefore, we performed ATAC-seq, RNA-seq, and weighted gene correlation network analysis to identify the chromatin accessibility, the messenger RNA (mRNA) expression, and the correlation between clinical phenotypes and mRNA expression using kidney from three mouse groups: db/m mice (Controls), db/db mice (case group), and those treated with DAPA (treatment group). RNA-Seq and ATAC-seq conjoint analysis revealed many overlapping pathways and networks suggesting that the transcriptional changes of DN and DAPA intervention largely occured dependently on chromatin remodeling. Specifically, the results showed that some key signal transduction pathways, such as immune dysfunction, glucolipid metabolism, oxidative stress and xenobiotic and endobiotic metabolism, were repeatedly enriched in the analysis of the RNA-seq data alone, as well as combined analysis with ATAC-seq data. Furthermore, we identified some candidate genes (UDP glucuronosyltransferase 1 family, Dock2, Tbc1d10c, etc.) and transcriptional regulators (KLF6 and GFI1) that might be associated with DN and DAPA restoration. These reversed genes and regulators confirmed that pathways related to immune response and metabolism pathways were critically involved in DN progression.
Collapse
Affiliation(s)
- Jianxiao Shen
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Ying
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajia Wu
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Fang
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyan Zhou
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaojun Qi
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Mou
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuru Yan
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Tian
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiajing Che
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Juffre A, Gumz ML. Recent advances in understanding the kidney circadian clock mechanism. Am J Physiol Renal Physiol 2024; 326:F382-F393. [PMID: 38174377 PMCID: PMC11207534 DOI: 10.1152/ajprenal.00214.2023] [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: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Circadian rhythms are endogenous biological oscillations that regulate various physiological processes in organisms, including kidney function. The kidney plays a vital role in maintaining homeostasis by regulating water and electrolyte balance, blood pressure, and excretion of metabolic waste products, all of which display circadian rhythmicity. For this reason, studying the circadian regulation of the kidney is important, and the time of day is a biological and experimental variable that must be considered. Over the past decade, considerable progress has been made in understanding the molecular mechanisms underlying circadian regulation within the kidney. In this review, the current knowledge regarding circadian rhythms in the kidney is explored, focusing on the molecular clock machinery, circadian control of renal functions, and the impact of disrupted circadian rhythms on kidney health. In addition, parameters that should be considered and future directions are outlined in this review.
Collapse
Affiliation(s)
- Alexandria Juffre
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
13
|
Zhu X, Ding L, Zhang X, Wang H, Chen N. Association between physical frailty, circadian syndrome and cardiovascular disease among middle-aged and older adults: a longitudinal study. BMC Geriatr 2024; 24:199. [PMID: 38413861 PMCID: PMC10900721 DOI: 10.1186/s12877-024-04787-8] [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: 06/07/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Physical frailty (PF) and circadian syndrome (CircS) are proposed as novel risks for cardiovascular disease (CVD), but little attention is paid to their combined impact on CVD. This study aimed to investigate the association of PF, CircS and CVD in middle-aged and older adults. METHODS The sample comprised 8512 participants aged at least 45 years from the China Health and Retirement Longitudinal Study (CHARLS) 2011. PF was examined by the physical frailty phenotype scale. CircS was assessed by the components of the International Diabetes Federation (IDF) MetS plus short sleep duration and depression. The cut-off for CircS was set as ≥ 4. CVD was defined as the presence of physician-diagnosed heart disease and/or stroke. A total of 6176 participants without CVD recruited from CHARLS 2011 and were followed up in 2018. RESULTS The prevalence of CVD in total populations, neither CircS or PF, PF alone, CircS alone and both CircS and PF were 13.0%, 7.4%, 15.5%, 17.4%, and 30.2%, respectively. CircS was more likely to be PF [OR (95%CI): 2.070 (1.732 ∼ 2.472)] than those without CircS. Both CircS alone [OR (95% CI): 1.954 (1.663 ∼ 2.296)], and coexisting CircS and PF [3.508 (2.739 ∼ 4.494)] were associated with CVD. Longitudinal analysis showed that individuals with both CircS and PF (HR: 1.716, 95%CI: 1.314 ∼ 2.240) and CircS alone [1.520 (1.331 ∼ 1.737)] were more likely to have new onset CVD than neither CircS or PF peers. CONCLUSION PF and CircS together are associated with higher CVD risk, which provided new evidence for a strong relation that warrants attention to assessing PF and CircS and in community to promote healthy aging.
Collapse
Affiliation(s)
- Xinhong Zhu
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China.
- Hubei Shizhen Laboratory, Wuhan, China.
| | - Linlin Ding
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaona Zhang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| | - Heqing Wang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| | - Ningbo Chen
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
14
|
Lee DY, Jung I, Park SY, Yu JH, Seo JA, Kim KJ, Kim NH, Yoo HJ, Kim SG, Choi KM, Baik SH, Kim NH. Attention to Innate Circadian Rhythm and the Impact of Its Disruption on Diabetes. Diabetes Metab J 2024; 48:37-52. [PMID: 38173377 PMCID: PMC10850272 DOI: 10.4093/dmj.2023.0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
Novel strategies are required to reduce the risk of developing diabetes and/or clinical outcomes and complications of diabetes. In this regard, the role of the circadian system may be a potential candidate for the prevention of diabetes. We reviewed evidence from animal, clinical, and epidemiological studies linking the circadian system to various aspects of the pathophysiology and clinical outcomes of diabetes. The circadian clock governs genetic, metabolic, hormonal, and behavioral signals in anticipation of cyclic 24-hour events through interactions between a "central clock" in the suprachiasmatic nucleus and "peripheral clocks" in the whole body. Currently, circadian rhythmicity in humans can be subjectively or objectively assessed by measuring melatonin and glucocorticoid levels, core body temperature, peripheral blood, oral mucosa, hair follicles, rest-activity cycles, sleep diaries, and circadian chronotypes. In this review, we summarized various circadian misalignments, such as altered light-dark, sleep-wake, rest-activity, fasting-feeding, shift work, evening chronotype, and social jetlag, as well as mutations in clock genes that could contribute to the development of diabetes and poor glycemic status in patients with diabetes. Targeting critical components of the circadian system could deliver potential candidates for the treatment and prevention of type 2 diabetes mellitus in the future.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - So Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyeong Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- BK21 FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Korea
| |
Collapse
|
15
|
Wang Y, Liu T, Cai Y, Liu W, Guo J. SIRT6's function in controlling the metabolism of lipids and glucose in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1244705. [PMID: 37876546 PMCID: PMC10591331 DOI: 10.3389/fendo.2023.1244705] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes mellitus (DM) and the main cause of excess mortality in patients with type 2 DM. The pathogenesis and progression of DN are closely associated with disorders of glucose and lipid metabolism. As a member of the sirtuin family, SIRT6 has deacetylation, defatty-acylation, and adenosine diphosphate-ribosylation enzyme activities as well as anti-aging and anticancer activities. SIRT6 plays an important role in glucose and lipid metabolism and signaling, especially in DN. SIRT6 improves glucose and lipid metabolism by controlling glycolysis and gluconeogenesis, affecting insulin secretion and transmission and regulating lipid decomposition, transport, and synthesis. Targeting SIRT6 may provide a new therapeutic strategy for DN by improving glucose and lipid metabolism. This review elaborates on the important role of SIRT6 in glucose and lipid metabolism, discusses the potential of SIRT6 as a therapeutic target to improve glucose and lipid metabolism and alleviate DN occurrence and progression of DN, and describes the prospects for future research.
Collapse
Affiliation(s)
- Ying Wang
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuzi Cai
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Peng Z, Liang Y, Liu X, Shao J, Hu N, Zhang X. New insights into the mechanisms of diabetic kidney disease: Role of circadian rhythm and Bmal1. Biomed Pharmacother 2023; 166:115422. [PMID: 37660646 DOI: 10.1016/j.biopha.2023.115422] [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: 06/24/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
It is common for diabetic kidney disease (DKD) to be complicated by abnormal blood glucose, blood lipids, and blood pressure rhythms. Thus, it is essential to examine diagnostic and treatment plans from the perspective of circadian disruption. This brief review discusses the clinical relevance of circadian rhythms in DKD and how the core clock gene encoding brain and muscle arnt-like protein 1 (BMAL1) functions owing to the importance of circadian rhythm disruption processes, including the excretion of urinary protein and irregular blood pressure, which occur in DKD. Exploring Bmal1 and its potential mechanisms and signaling pathways in DKD following contact with Sirt1 and NF-κB is novel and important. Finally, potential pharmacological and behavioral intervention strategies for DKD circadian rhythm disturbance are outlined. This review aids in unveiling novel, potential molecular targets for DKD based on circadian rhythms.
Collapse
Affiliation(s)
- Zhimei Peng
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China; Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Yanting Liang
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Xueying Liu
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Jie Shao
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Nan Hu
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Xinzhou Zhang
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China; Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| |
Collapse
|
17
|
Kunke M, Knöfler H, Dahlke E, Zanon Rodriguez L, Böttner M, Larionov A, Saudenova M, Ohrenschall GM, Westermann M, Porubsky S, Bernardes JP, Häsler R, Magnin JL, Koepsell H, Jouret F, Theilig F. Targeted deletion of von-Hippel-Lindau in the proximal tubule conditions the kidney against early diabetic kidney disease. Cell Death Dis 2023; 14:562. [PMID: 37626062 PMCID: PMC10457389 DOI: 10.1038/s41419-023-06074-7] [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: 04/19/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023]
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease. Glomerular hyperfiltration and albuminuria subject the proximal tubule (PT) to a subsequent elevation of workload, growth, and hypoxia. Hypoxia plays an ambiguous role in the development and progression of DKD and shall be clarified in our study. PT-von-Hippel-Lindau (Vhl)-deleted mouse model in combination with streptozotocin (STZ)-induced type I diabetes mellitus (DM) was phenotyped. In contrary to PT-Vhl-deleted STZ-induced type 1 DM mice, proteinuria and glomerular hyperfiltration occurred in diabetic control mice the latter due to higher nitric oxide synthase 1 and sodium and glucose transporter expression. PT Vhl deletion and DKD share common alterations in gene expression profiles, including glomerular and tubular morphology, and tubular transport and metabolism. Compared to diabetic control mice, the most significantly altered in PT Vhl-deleted STZ-induced type 1 DM mice were Ldc-1, regulating cellular oxygen consumption rate, and Zbtb16, inhibiting autophagy. Alignment of altered genes in heat maps uncovered that Vhl deletion prior to STZ-induced DM preconditioned the kidney against DKD. HIF-1α stabilization leading to histone modification and chromatin remodeling resets most genes altered upon DKD towards the control level. These data demonstrate that PT HIF-1α stabilization is a hallmark of early DKD and that targeting hypoxia prior to the onset of type 1 DM normalizes renal cell homeostasis and prevents DKD development.
Collapse
Affiliation(s)
- Madlen Kunke
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | - Hannah Knöfler
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | - Eileen Dahlke
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | | | - Martina Böttner
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | - Alexey Larionov
- Institute of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | | | | | | | | | - Joana P Bernardes
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Robert Häsler
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Hermann Koepsell
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Liège, Belgium
- Division of Nephrology, CHU of Liège, University of Liège (CHU ULiège), Liège, Belgium
| | - Franziska Theilig
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany.
- Institute of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
18
|
Chen W, Zheng D, Chen H, Ye T, Liu Z, Qi J, Shen H, Ruan H, Cui W, Deng L. Circadian Clock Regulation via Biomaterials for Nucleus Pulposus. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301037. [PMID: 37060338 DOI: 10.1002/adma.202301037] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/11/2023] [Indexed: 05/30/2023]
Abstract
Circadian clock disorder during tissue degeneration has been considered the potential pathogenesis for various chronic diseases, such as intervertebral disc degeneration (IVDD). In this study, circadian clock-regulating biomaterials (ClockMPs) that can effectively activate the intrinsic circadian clock of nucleus pulposus cells (NPCs) in IVDD and improve the physiological function of NPCs for disc regeneration are fabricated via air-microfluidic technique and the chemical cross-linking between polyvinyl alcohol and modified-phenylboronic acid. In vitro experiments verified that ClockMPs can scavenge reactive oxygen species to maintain a stable microenvironment for the circadian clock by promoting the binding of BMAL1 and CLOCK proteins. ClockMPs can regulate the expression of core circadian clock genes by activating the PI3K-AKT pathway in NPCs to remodel the intrinsic circadian clock and promote extracellular matrix synthesis. Furthermore, in vivo experiments of IVDD treated with ClockMPs proved that ClockMPs can promote disc regeneration by regulating the circadian clock of NPCs. In conclusion, ClockMPs provided a novel and promising strategy for circadian clock regulation during tissue regeneration.
Collapse
Affiliation(s)
- Wei Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Dandan Zheng
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Hao Chen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Zhihong Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Jin Qi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Hongxing Shen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Huitong Ruan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
19
|
Luo Y, Zhang L, Zhao T. Identification and analysis of cellular senescence-associated signatures in diabetic kidney disease by integrated bioinformatics analysis and machine learning. Front Endocrinol (Lausanne) 2023; 14:1193228. [PMID: 37396184 PMCID: PMC10313062 DOI: 10.3389/fendo.2023.1193228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/04/2023] [Indexed: 07/04/2023] Open
Abstract
Background Diabetic kidney disease (DKD) is a common complication of diabetes that is clinically characterized by progressive albuminuria due to glomerular destruction. The etiology of DKD is multifactorial, and numerous studies have demonstrated that cellular senescence plays a significant role in its pathogenesis, but the specific mechanism requires further investigation. Methods This study utilized 5 datasets comprising 144 renal samples from the Gene Expression Omnibus (GEO) database. We obtained cellular senescence-related pathways from the Molecular Signatures Database and evaluated the activity of senescence pathways in DKD patients using the Gene Set Enrichment Analysis (GSEA) algorithm. Furthermore, we identified module genes related to cellular senescence pathways through Weighted Gene Co-Expression Network Analysis (WGCNA) algorithm and used machine learning algorithms to screen for hub genes related to senescence. Subsequently, we constructed a cellular senescence-related signature (SRS) risk score based on hub genes using the Least Absolute Shrinkage and Selection Operator (LASSO), and verified mRNA levels of hub genes by RT-PCR in vivo. Finally, we validated the relationship between the SRS risk score and kidney function, as well as their association with mitochondrial function and immune infiltration. Results The activity of cellular senescence-related pathways was found to be elevated among DKD patients. Based on 5 hub genes (LIMA1, ZFP36, FOS, IGFBP6, CKB), a cellular senescence-related signature (SRS) was constructed and validated as a risk factor for renal function decline in DKD patients. Notably, patients with high SRS risk scores exhibited extensive inhibition of mitochondrial pathways and upregulation of immune cell infiltration. Conclusion Collectively, our findings demonstrated that cellular senescence is involved in the process of DKD, providing a novel strategy for treating DKD.
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingxiao Zhang
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tongfeng Zhao
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Chronobiology and Nanotechnology for Personalized Cancer Therapy. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|