1
|
Yu F, Chen J, Zhang X, Ma Z, Wang J, Wu Q. Role of Neutrophil Extracellular Traps in Hypertension and Their Impact on Target Organs. J Clin Hypertens (Greenwich) 2024. [PMID: 39686847 DOI: 10.1111/jch.14942] [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: 06/13/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 12/18/2024]
Abstract
Hypertension is the predominant cause of cardiovascular diseases (CVDs) globally, and essential hypertension (EH) represents a significant public health challenge due to its multifactorial etiology involving complex interactions between genetic and environmental factors. However, the pathogenesis of EH is still unclear. Hypertension is a dysregulation in the renin-angiotensin-aldosterone system and sympathetic nervous system, both regulating saline homeostasis and cardiovascular function. However, current therapeutic interventions targeting these systems have limited efficacy in approximately 40% of cases, suggesting the involvement of alternative mechanisms. Inflammation is associated with the occurrence and progression of hypertension, but the underlying mechanism remains elusive, while chronic inflammation leads to tissue damage, fibrosis, and irreversible organ dysfunction. The development and maintenance of EH are caused by endothelial dysfunction, oxidative stress, and chronic inflammation. Neutrophils are involved in both acute and chronic inflammation since they represent the primary line of defense against inflammatory insults once recruited to the inflamed site where they remove harmful impurities. The process involving the formation of neutrophil extracellular traps (NETs) is called NETosis are involved in the pathogenesis and progression of CVDs, including coronary artery disease, acute myocardial infarction, peripheral arterial disease, heart failure, and atrial fibrillation. Recent investigations demonstrated that NETs facilitate the development of hypertension; however, the precise role of NETs in hypertension remains largely elusive. Therefore, this review aims to provide an overview of the current understanding regarding the involvement of NETosis in hypertension and explore the potential therapies targeting NETs for future interventions.
Collapse
Affiliation(s)
- Fei Yu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianshu Chen
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaowei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhengke Ma
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jingtao Wang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiang Wu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Liu M, Di YM, May B, Zhang AL, Zhang L, Chen J, Wang R, Liu X, Xue CC. Renal protective effects and mechanisms of Astragalus membranaceus for diabetic kidney disease in animal models: An updated systematic review and meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155646. [PMID: 38733903 DOI: 10.1016/j.phymed.2024.155646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Astragalus membranaceus (AM) shows potential therapeutic benefits for managing diabetic kidney disease (DKD), a leading cause of kidney failure with no cure. However, its comprehensive effects on renal outcomes and plausible mechanisms remain unclear. PURPOSE This systematic review and meta-analysis aimed to synthesize the effects and mechanisms of AM on renal outcomes in DKD animal models. METHODS Seven electronic databases were searched for animal studies until September 2023. Risk of bias was assessed based on SYRCLE's Risk of Bias tool. Standardized mean difference (SMD) or mean difference (MD) were estimated for the effects of AM on serum creatinine (SCr), blood urea nitrogen (BUN), albuminuria, histological changes, oxidative stress, inflammation, fibrosis and glucolipids. Effects were pooled using random-effects models. Heterogeneity was presented as I2. Subgroup analysis investigated treatment- and animal-related factors for renal outcomes. Publication bias was assessed using funnel plots and Egger's test. Sensitivity analysis was performed to assess the results' robustness. RevMan 5.3 and Stata MP 15 software were used for statistical analysis. RESULTS Forty studies involving 1543 animals were identified for analysis. AM treatment significantly decreased SCr (MD = -19.12 μmol/l, 95 % CI: -25.02 to -13.23), BUN (MD = -6.72 mmol/l, 95 % CI: -9.32 to -4.12), urinary albumin excretion rate (SMD = -2.74, 95 % CI: -3.57, -1.90), histological changes (SMD = -2.25, 95 % CI: -3.19 to -1.32). AM treatment significantly improved anti-oxidative stress expression (SMD = 1.69, 95 % CI: 0.97 to 2.41), and decreased inflammation biomarkers (SMD = -3.58, 95 % CI: -5.21 to -1.95). AM treatment also decreased fibrosis markers (i.e. TGF-β1, CTGF, collagen IV, Wnt4 and β-catenin) and increased anti-fibrosis marker BMP-7. Blood glucose, lipids and kidney size were also improved compared with the DM control group. CONCLUSION AM could improve renal outcomes and alleviate injury through multiple signaling pathways. This indicates AM may be an option to consider for the development of future DKD therapeutics.
Collapse
Affiliation(s)
- Meifang Liu
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, 3083, Australia; Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Yuan Ming Di
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, 3083, Australia
| | - Brian May
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, 3083, Australia
| | - Anthony Lin Zhang
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, 3083, Australia
| | - Lei Zhang
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Junhui Chen
- Second Clinical College of Guangzhou University of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ruobing Wang
- Second Clinical College of Guangzhou University of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xusheng Liu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Charlie Changli Xue
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, 3083, Australia.
| |
Collapse
|
3
|
Yang Y, Meng L, Hu X, Li X. Renal functional outcomes after nephrectomy in patients with localized renal cell carcinoma and diabetes mellitus: a systematic review and meta-analysis. Int Urol Nephrol 2024; 56:1859-1868. [PMID: 38300449 DOI: 10.1007/s11255-023-03885-7] [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: 07/08/2023] [Accepted: 10/31/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION AND OBJECTIVE Diabetes mellitus (DM), one of the most common comorbidities in patients with renal cell carcinoma (RCC), was proven to be an important prognostic factor of overall survival for these patients. Regarding the influence on renal function after nephrectomy, evidence is still scant. This systematic review and meta-analysis was conducted to provide a more reliable analysis of the association between DM and long-term renal functional outcomes after nephrectomy. METHODS The PubMed, Web of Science, Embase and Cochrane Library (CENTRAL) databases were searched for eligible studies from inception to January 2023. Hazard ratios (HRs) with 95% confidence intervals (CIs) were extracted to evaluate the association between DM and renal functional outcomes using a random effects model. Stata 17.0 software was used for statistical analysis. RESULTS The meta-analysis included thirteen studies consisting of 8562 RCC patients who underwent nephrectomy. Preoperative comorbidity of DM was significantly associated with poor renal functional outcomes (HR = 1.91, 95% CI 1.48-2.48, p < 0.0001), regardless of ethnicity, follow-up time, body mass index (BMI) and age. However, in the radical nephrectomy subgroup, DM was not significantly associated with renal function decline (HR = 1.91, 95% CI 0.93-3.90, p = 0.0781). CONCLUSIONS The aggregate evidence indicated that preexisting DM may be associated with poor renal functional outcomes in patients with RCC after nephrectomy, especially in patients receiving partial nephrectomy. Urologists should focus more on the glycemic management of these patients after nephrectomy. More high-quality studies are needed to explore the influence of DM on renal function outcomes in postoperative patients.
Collapse
Affiliation(s)
- Yujia Yang
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Linghao Meng
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Xu Hu
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Li
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Piko N, Bevc S, Hojs R, Ekart R. Finerenone: From the Mechanism of Action to Clinical Use in Kidney Disease. Pharmaceuticals (Basel) 2024; 17:418. [PMID: 38675379 PMCID: PMC11054947 DOI: 10.3390/ph17040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic kidney disease is a frequent microvascular complication of diabetes and is currently the leading cause of chronic kidney disease and end-stage kidney disease worldwide. Although the prevalence of other complications of diabetes is falling, the number of diabetic patients with end-stage kidney disease in need of kidney replacement therapy is rising. In addition, these patients have extremely high cardiovascular risk. It is more than evident that there is a high unmet treatment need in patients with diabetic kidney disease. Finerenone is a novel nonsteroidal mineralocorticoid receptor antagonist used for treating diabetic kidney disease. It has predominant anti-fibrotic and anti-inflammatory effects and exhibits several renal and cardiac protective effects. This review article summarizes the current knowledge and future prospects of finerenone in treating patients with kidney disease.
Collapse
Affiliation(s)
- Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia;
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
5
|
Ozbek DA, Koc SC, Özkan NE, Kablan SE, Yet I, Uner M, Ozlu N, Nemutlu E, Lay I, Ayhan AS, Yildirim T, Arici M, Yilmaz SR, Erdem Y, Altun B. A comparative urinary proteomic and metabolomic analysis between renal aa amyloidosis and membranous nephropathy with clinicopathologic correlations. J Proteomics 2024; 293:105064. [PMID: 38154551 DOI: 10.1016/j.jprot.2023.105064] [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: 07/27/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
Urinary omics has become a powerful tool for elucidating pathophysiology of glomerular diseases. However, no urinary omics analysis has been performed yet on renal AA amyloidosis. Here, we performed a comparative urine proteomic and metabolomic analysis between recently diagnosed renal AA amyloidosis (AA) and membranous nephropathy (MN) patients. Urine samples of 22 (8 AA, 8 MN and 6 healthy control) patients were analyzed with nLC-MS/MS and GC/MS for proteomic and metabolomic studies, respectively. Pathological specimens were scored for glomerulosclerosis and tubulointerstitial fibrosis grades. Functional enrichment analysis between AA and control groups showed enrichment in cell adhesion related sub-domains. Uromodulin (UMOD) was lower, whereas ribonuclease 1 (RNase1) and α-1-microglobulin/bikunin precursor (AMBP) were higher in AA compared to MN group. Correlations were demonstrated between UMOD-proteinuria (r = -0.48, p = 0.03) and AMBP-eGFR (r = -0.69, p = 0.003) variables. Metabolomic analysis showed myo-inositol and urate were higher in AA compared to MN group. A positive correlation was detected between RNase1 and urate independent of eGFR values (r = 0.63, p = 0.01). Enrichment in cell adhesion related domains suggested a possible increased urinary shear stress due to amyloid fibrils. UMOD, AMBP and myo-inositol were related with tubulointerstitial damage, whereas RNase1 and urate were believed to be related with systemic inflammation in AA amyloidosis. SIGNIFICANCE: Urinary omics studies have become a standard tool for biomarker studies. However, no urinary omics analysis has been performed yet on renal AA amyloidosis. Here, we performed a comparative urinary omics analysis between recently diagnosed renal AA amyloidosis (AA), membranous nephropathy (MN) patients and healthy controls. Pathological specimens were scored with glomerulosclerosis (G) and tubulointerstitial fibrosis (IF) grades to consolidate the results of the omics studies and correlation analyzes. Functional enrichment analysis showed enrichment in cell adhesion related sub-domains due to downregulation of cadherins; which could be related with increased urinary shear stress due to amyloid deposition and disruption of tissue micro-architecture. In comparative proteomic analyzes UMOD was lower, whereas RNase1 and AMBP were higher in AA compared to MN group. Whereas in metabolomic analyzes; myo-inositol, urate and maltose were higher in AA compared to MN group. Correlations were demonstrated between UMOD-proteinuria (r = -0.48, p = 0.03), AMBP-eGFR (r = -0.69, p = 0.003) and between RNase1-Urate independent of eGFR values (r = 0.63, p = 0.01). This study is the first comprehensive urinary omics analysis focusing on renal AA Amyloidosis to the best of our knowledge. Based on physiologic roles and clinicopathologic correlations of the molecules; UMOD, AMBP and myo-inositol were related with tubulointerstitial damage, whereas RNase1 and urate were believed to be increased with systemic inflammation and endothelial damage in AA amyloidosis.
Collapse
Affiliation(s)
- Deniz Aral Ozbek
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey.
| | - Sila Cankurtaran Koc
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Nazlı Ezgi Özkan
- Koc University Research Center for Translational Medicine, Istanbul, Turkey
| | - Sevilay Erdogan Kablan
- Hacettepe University Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, Turkey
| | - Idil Yet
- Hacettepe University Graduate School of Health Sciences, Department of Bioinformatics, Ankara, Turkey
| | - Meral Uner
- Hacettepe University Faculty of Medicine, Department of Pathology, Ankara, Turkey
| | - Nurhan Ozlu
- Koc University Research Center for Translational Medicine, Istanbul, Turkey
| | - Emirhan Nemutlu
- Hacettepe University Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, Turkey
| | - Incilay Lay
- Hacettepe University Faculty of Medicine, Department of Biochemistry, Ankara, Turkey
| | - Arzu Saglam Ayhan
- Hacettepe University Faculty of Medicine, Department of Pathology, Ankara, Turkey
| | - Tolga Yildirim
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Mustafa Arici
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Seref Rahmi Yilmaz
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Yunus Erdem
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| | - Bulent Altun
- Hacettepe University Faculty of Medicine, Department of Nephrology, Ankara, Turkey
| |
Collapse
|
6
|
Taber-Hight E, Gilmore A, Friedman AN. Anti-obesity pharmacotherapy in adults with chronic kidney disease. Kidney Int 2024; 105:269-280. [PMID: 37926421 DOI: 10.1016/j.kint.2023.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023]
Abstract
Obesity is a leading risk factor for the development and progression of kidney disease and a major barrier to optimal management of patients with chronic kidney disease. While in the past anti-obesity drugs offered only modest weight loss efficacy in exchange for various safety and tolerability risks, a wave of safer, more tolerable, and more effective treatment options is transforming the management of obesity. This review evaluates current and future pharmacologic anti-obesity therapy in adults through a kidney-oriented lens. It also explores the goals of anti-obesity treatment, describes the underlying putative mechanisms of action, and raises important scientific questions that deserve further exploration in people with chronic kidney disease.
Collapse
Affiliation(s)
- Elizabeth Taber-Hight
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ashley Gilmore
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Allon N Friedman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
7
|
Liu M, Di YM, Zhang AL, Chen J, Wang R, Huang J, Zhang L, Xue CC, Liu X. Renal-protective effects of Chinese medicinal herbs and compounds for diabetic kidney disease in animal models: protocol for systematic review and meta-analysis. Syst Rev 2024; 13:23. [PMID: 38217017 PMCID: PMC10785383 DOI: 10.1186/s13643-023-02446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/23/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a common and severe complication of diabetes that can lead to end-stage renal disease with no cure. The first-line drugs recommended by clinical guidelines fail to achieve satisfactory effects for people with DKD. A Chinese herbal medicine Tangshen Qushi Formula (TQF) shows preliminary efficacy and safety in preserving renal function for people with DKD, but the effects on comprehensive renal outcomes remain unclear. We will conduct a systematic review and meta-analysis to evaluate the effects of TQF herbs and their compounds identified from ultra-high performance liquid chromatography-MS/MS in diabetic animal models with renal outcomes. METHODS This protocol complies with the guideline Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols. We will include studies investigating the effects of TQF herbs and compounds on diabetic rats or mice with renal outcomes. Six electronic databases will be searched from their inception to February 2023. Quality assessment will be conducted using SYRCLE's risk of bias tool. Standardized or weighted mean differences will be estimated for renal outcomes (creatinine, urea, proteinuria, histological changes, oxidative stress, inflammation, and kidney fibrosis). Data will be pooled using random-effects models. Heterogeneity across studies will be expressed as I2. Sensitivity analyses will explore treatment effects in adjusted models and within subgroups. Funnel plots and Egger's test will be used to explore publication bias. DISCUSSION The results of this review will provide valuable insights into the potential effects of TQF in managing DKD. The limitation is that the included studies will be animal studies from specific databases, and the interpretation of the findings must be cautious. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023432895. Registered on 19 July 2023 ( https://www.crd.york.ac.uk/PROSPERO/#recordDetails ).
Collapse
Affiliation(s)
- Meifang Liu
- The China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Ming Di
- The China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Anthony Lin Zhang
- The China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Junhui Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruobing Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juan Huang
- Pharmaceutical Research Department for New Drug Development and Authentication of Chinese Medicines, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Charlie Changli Xue
- The China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia.
| | - Xusheng Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
8
|
Huang Z, Nie H, Liu G, Li P, Peng YH, Xiao J, Gu W, Li TS. Losartan alleviates renal fibrosis by inhibiting the biomechanical stress-induced epithelial-mesenchymal transition of renal epithelial cells. Arch Biochem Biophys 2023; 748:109770. [PMID: 37783367 DOI: 10.1016/j.abb.2023.109770] [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: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023]
Abstract
Angiotensin receptor blockers (ARBs) have been reported to be beneficial of renal fibrosis, but the molecular and cellular mechanisms are still unclear. In this study, we investigated the effectiveness and relevant mechanism of ARBs in alleviating renal fibrosis, especially by focusing on biomechanical stress-induced epithelial to mesenchymal transition (EMT) of renal epithelial cells. Unilateral ureteral obstruction (UUO) renal fibrosis model was established in mice by ligating the left ureter, and then randomly received losartan at a low dose (1 mg/kg) or a regular dose (3 mg/kg) for 2 weeks. Compared to the control, histological analysis showed that losartan treatment at either a low dose or a regular dose effectively attenuated renal fibrosis in the UUO model. To further understand the mechanism, we ex vivo loaded primary human renal epithelial cells to 50 mmHg hydrostatic pressure. Western blot and immunostaining analyses indicated that the loading to 50 mmHg hydrostatic pressure for 24 h significantly upregulated vimentin, β-catenin and α-SMA, but downregulated E-cadherin in renal epithelial cells, suggesting the EMT. The addition of 10 or 100 nM losartan in medium effectively attenuated the EMT of renal epithelial cells induced by 50 mmHg hydrostatic pressure loading. Our in vivo and ex vivo experimental data suggest that losartan treatment, even at a low dose can effectively alleviate renal fibrosis in mouse UUO model, at least partly by inhibiting the biomechanical stress-induced EMT of renal epithelial cells. A low dose of ARBs may repurpose for renal fibrosis treatment.
Collapse
Affiliation(s)
- Zisheng Huang
- Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Japan; Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Han Nie
- Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Japan; Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Geng Liu
- Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Japan; Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Peilin Li
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Yong-Hua Peng
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jie Xiao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Weili Gu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, China.
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Japan; Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
9
|
Naaman SC, Bakris GL. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. Diabetes Care 2023; 46:1574-1586. [PMID: 37625003 PMCID: PMC10547606 DOI: 10.2337/dci23-0030] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/16/2023] [Indexed: 08/27/2023]
Abstract
Management of diabetic kidney disease (DKD) has evolved in parallel with our growing understanding of the multiple interrelated pathophysiological mechanisms that involve hemodynamic, metabolic, and inflammatory pathways. These pathways and others play a vital role in the initiation and progression of DKD. Since its initial discovery, the blockade of the renin-angiotensin system has remained a cornerstone of DKD management, leaving a large component of residual risk to be dealt with. The advent of sodium-glucose cotransporter 2 inhibitors followed by nonsteroidal mineralocorticoid receptor antagonists and, to some extent, glucagon-like peptide 1 receptor agonists (GLP-1 RAs) has ushered in a resounding paradigm shift that supports a pillared approach in maximizing treatment to reduce outcomes. This pillared approach is like that derived from the approach to heart failure treatment. The approach mandates that all agents that have been shown in clinical trials to reduce cardiovascular outcomes and/or mortality to a greater extent than a single drug class alone should be used in combination. In this way, each drug class focuses on a specific aspect of the disease's pathophysiology. Thus, in heart failure, β-blockers, sacubitril/valsartan, a mineralocorticoid receptor antagonist, and a diuretic are used together. In this article, we review the evolution of the pillar concept of therapy as it applies to DKD and discuss how it should be used based on the outcome evidence. We also discuss the exciting possibility that GLP-1 RAs may be an additional pillar in the quest to further slow kidney disease progression in diabetes.
Collapse
Affiliation(s)
- Sandra C. Naaman
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL
| | - George L. Bakris
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
10
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
11
|
Chung GE, Han K, Lee KN, Bae JH, Yang SY, Choi SY, Yim JY, Heo NJ. Association between fatty liver index and risk of end-stage renal disease stratified by kidney function in patients with type 2 diabetes: A nationwide population-based study. DIABETES & METABOLISM 2023; 49:101454. [PMID: 37244418 DOI: 10.1016/j.diabet.2023.101454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVE The effects of nonalcoholic fatty liver disease on the risk of end-stage renal disease (ESRD) remain unclear. We investigated the association between the fatty liver index (FLI) and risk of ESRD in patients with type 2 diabetes. METHODS This population-based observational cohort study enrolled patients with diabetes who underwent health screening between 2009 and 2012 and utilized data from the Korean National Health Insurance Services. The FLI functioned as a surrogate marker for the presence of hepatic steatosis. Chronic kidney disease (CKD) was defined as an estimated glomerular filtration rate < 60 ml/min/1.73 m² calculated using the Modification of Diet in Renal Disease equation. We performed Cox proportional hazards regression. RESULTS Incident ESRD developed in 19,476 of 1,900,598 patients with type 2 diabetes during a median follow-up of 7.2 years. After adjusting for conventional risk factors, patients with high FLI scores had a higher risk for ESRD: FLI, 30-59 [hazard ratio (HR) = 1.124; 95% confidence interval (CI), 1.083-1.166]; FLI ≥ 60 [HR = 1.278; 95% CI, 1.217-1.343] compared with those with FLI < 30. The association between a high FLI score (≥ 60) and incident ESRD was more prominent in women than in men (male, FLI ≥60: HR, 1.106; 95% CI = 1.041-1.176 and female, FLI ≥ 60: HR, 1.835; 95% CI = 1.689-1.995). The association between a high FLI score (≥ 60) and the risk of ESRD differed according to baseline kidney function. High FLI scores increased the risk of ESRD (HR = 1.268; 95% CI, 1.198-1.342) in patients with CKD at baseline. CONCLUSION High FLI scores are associated with a greater risk of ESRD in patients with type 2 diabetes with CKD at baseline. Close monitoring and appropriate management of hepatic steatosis may aid in preventing the progression of kidney dysfunction in patients with type 2 diabetes and CKD.
Collapse
Affiliation(s)
- Goh Eun Chung
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Biostatistics, College of Medicine, The Soongsil University, Seoul, Republic of Korea
| | - Kyu-Na Lee
- Department of Biomedicine & Health Science, Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Ho Bae
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sun Young Yang
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Su-Yeon Choi
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeong Yoon Yim
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Nam Ju Heo
- Department of Internal Medicine, Healthcare Research Institute, Gangnam Healthcare Center, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Zhang T, Wang X, Zhang Y, Yang Y, Yang C, Wei H, Zhao Q. Establishment of a potent weighted risk model for determining the progression of diabetic kidney disease. J Transl Med 2023; 21:381. [PMID: 37308973 DOI: 10.1186/s12967-023-04245-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a severe complication of diabetes. Currently, no effective measures are available to reduce the risk of DKD progression. This study aimed to establish a weighted risk model to determine DKD progression and provide effective treatment strategies. METHODS This was a hospital-based, cross-sectional study. A total of 1104 patients with DKD were included in this study. The random forest method was used to develop weighted risk models to assess DKD progression. Receiver operating characteristic curves were used to validate the models and calculate the optimal cutoff values for important risk factors. RESULTS We developed potent weighted risk models to evaluate DKD progression. The top six risk factors for DKD progression to chronic kidney disease were hemoglobin, hemoglobin A1c (HbA1c), serum uric acid (SUA), plasma fibrinogen, serum albumin, and neutrophil percentage. The top six risk factors for determining DKD progression to dialysis were hemoglobin, HbA1c, neutrophil percentage, serum albumin, duration of diabetes, and plasma fibrinogen level. Furthermore, the optimal cutoff values of hemoglobin and HbA1c for determining DKD progression were 112 g/L and 7.2%, respectively. CONCLUSION We developed potent weighted risk models for DKD progression that can be employed to formulate precise therapeutic strategies. Monitoring and controlling combined risk factors and prioritizing interventions for key risk factors may help reduce the risk of DKD progression.
Collapse
Affiliation(s)
- Tianxiao Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaodan Wang
- Department of Geratology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yueying Zhang
- Department of Geratology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ying Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Congying Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Huiyi Wei
- School of Medicine, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Qingbin Zhao
- Department of Geratology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
- Department of Geratology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
13
|
Youth versus adult-onset type 2 diabetic kidney disease: Insights into currently known structural differences and the potential underlying mechanisms. Clin Sci (Lond) 2022; 136:1471-1483. [PMID: 36326718 PMCID: PMC10175439 DOI: 10.1042/cs20210627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/05/2022]
Abstract
Abstract
Type 2 diabetes (T2D) is a global health pandemic with significant humanitarian, economic, and societal implications, particularly for youth and young adults who are experiencing an exponential rise in incident disease. Youth-onset T2D has a more aggressive phenotype than adult-onset T2D, and this translates to important differences in rates of progression of diabetic kidney disease (DKD). We hypothesize that youth-onset DKD due to T2D may exhibit morphometric, metabolic, and molecular characteristics that are distinct from adult-onset T2D and develop secondary to inherent differences in renal energy expenditure and substrate metabolism, resulting in a central metabolic imbalance. Kidney structural changes that are evident at the onset of puberty also serve to exacerbate the organ’s baseline high rates of energy expenditure. Additionally, the physiologic state of insulin resistance seen during puberty increases the risk for kidney disease and is exacerbated by both concurrent diabetes and obesity. A metabolic mismatch in renal energetics may represent a novel target for pharmacologic intervention, both for prevention and treatment of DKD. Further investigation into the underlying molecular mechanisms resulting in DKD in youth-onset T2D using metabolomics and RNA sequencing of kidney tissue obtained at biopsy is necessary to expand our understanding of early DKD and potential targets for therapeutic intervention. Furthermore, large-scale clinical trials evaluating the duration of kidney protective effects of pharmacologic interventions that target a metabolic mismatch in kidney energy expenditure are needed to help mitigate the risk of DKD in youth-onset T2D.
Collapse
|
14
|
Cui QQ, Li XM, Xie Y. Study on the mechanism of warming yang and reducing turbidity decoction in the treatment of diabetic kidney disease based on network pharmacology. Medicine (Baltimore) 2022; 101:e30728. [PMID: 36181090 PMCID: PMC9524955 DOI: 10.1097/md.0000000000030728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
This study aimed to investigate the mechanism of warming yang and reducing turbidity decoction in the treatment of diabetic kidney disease (DKD) by network pharmacology. The active components and corresponding targets of warming yang and reducing turbidity decoction were screened through the Traditional Chinese Medicine Systems Pharmacology database, DKD-related targets were obtained from Genecard and Online Mendelian Inheritance in Man databases, and drug-disease common targets were screened through Venny online website. Then we used STRING and Cytoscape software to analyze and perform protein-protein interaction network, and used CytoNCA plug-in to perform topological analysis to screen out the core target. We used RStudio to performed gene ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. One hundred one active components in warming yang and reducing turbidity decoction participated in the regulation of the body's response to foreign bodies, lipopolysaccharides, metal ions, ketone bodies, hypoxia and oxidative stress by regulating 186 targets related to DKD, and played a role in the treatment of DKD by interfering with pathways such as interfered with lipids and atherosclerosis, PI3K-Akt, fluid shear stress and atherosclerosis, AGE-RAGE and cell senescence. It was implied that warming yang and reducing turbidity decoction had the features of multi components, multi targets and multi pathways in the treatment of DKD, which might create methods and directions for further verification of the molecular mechanism of warming yang and reducing turbidity decoction.
Collapse
Affiliation(s)
- Quan-Qing Cui
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Endocrinology, Gaozhou People’s Hospital, Gaozhou, Guangdong Province, China
| | - Xian-Min Li
- Department of Orthopedics, Gaozhou People’s Hospital, Gaozhou, Guangdong Province, China
| | - Ying Xie
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- *Correspondence: Ying Xie, Department of Endocrinology, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou 215008, Jiangsu Province, China (e-mail: )
| |
Collapse
|
15
|
The Influence of the Severity of Early Chronic Kidney Disease on Oxidative Stress in Patients with and without Type 2 Diabetes Mellitus. Int J Mol Sci 2022; 23:ijms231911196. [PMID: 36232497 PMCID: PMC9569590 DOI: 10.3390/ijms231911196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Early Chronic Kidney Disease (CKD) is a condition that tends to progress to End-Stage Kidney Disease (ESKD). Early diagnosis of kidney disease in the early stages can reduce complications. Alterations in renal function represent a complication of diabetes mellitus (DM). The mechanisms underlying the progression of CKD in diabetes could be associated with oxidative and inflammatory processes. This study aimed to evaluate the state of inflammation and oxidative stress (OS) on the progression of CKD in the early stages in patients with and without type 2 diabetes mellitus (T2DM). An analytical cross-sectional study was carried out in patients with CKD in early stages (1, 2, 3) with and without T2DM. The ELISA method determined the expression of pro-inflammatory cytokines IL-6 and TNF-α as well as lipoperoxides (LPO), nitric oxide (NO), and superoxide dismutase activity (SOD). Colorimetric methods determined glutathione peroxidase (GPx) and total antioxidant capacity (TAC). Patients with CKD and T2DM had significantly decreased antioxidant defenses for SOD (p < 0.01), GPx (p < 0.01), and TAC (p < 0.01) compared to patients without T2DM. Consequently, patients with T2DM had higher concentrations of oxidant markers, NO (p < 0.01), inflammation markers, IL-6 (p < 0.01), and TNF-α than patients without T2DM. CKD stages were not related to oxidative, antioxidant, and inflammatory marker outcomes in T2DM patients. Patients without T2DM presented an increase in SOD (p = 0.04) and a decrease in NO (p < 0.01) when the stage of CKD increased. In conclusion, patients with T2DM present higher levels of oxidative and inflammatory markers accompanied by a decrease in antioxidant defense. However, these oxidative status markers were associated with CKD stage progression in patients without T2DM. Thus, NO and SOD markers could help detect the early stages of CKD in patients who have not yet developed metabolic comorbidities such as T2DM.
Collapse
|
16
|
Sánchez-Jaramillo EA, Gasca-Lozano LE, Vera-Cruz JM, Hernández-Ortega LD, Gurrola-Díaz CM, Bastidas-Ramírez BE, Vargas-Guerrero B, Mena-Enríquez M, Martínez-Limón FDJ, Salazar-Montes AM. Nanoparticles Formulation Improves the Antifibrogenic Effect of Quercetin on an Adenine-Induced Model of Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23105392. [PMID: 35628203 PMCID: PMC9140764 DOI: 10.3390/ijms23105392] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Renal fibrosis is the final stage of chronic kidney injury characterized by glomerulosclerosis and tubulointerstitial fibrosis with parenchymal destruction. Quercetin belongs to the most studied flavonoids with antioxidant, anti-inflammatory, antifibrogenic, and antitumor activity. It modifies the TGF-β/Smad signaling pathway, decreasing profibrogenic expression molecules and inducing the expression of antioxidant, anti-inflammatory, and antifibrogenic molecules. However, quercetin exhibits poor water solubility and low absorption and bioavailability. This limitation was solved by developing a nanoparticles formulation that improves the solubility and bioavailability of several bioactive compounds. Therefore, we aimed to investigate the in vivo antifibrogenic effect of a quercetin nanoparticles formulation. Male C57BL/6 mice were induced into chronic renal failure with 50 mg/kg of adenine for four weeks. The animals were randomly grouped and treated with 25, 50, or 100 mg/kg of quercetin, either macroparticles or nanoparticles formulation. We performed biochemical, histological, and molecular analyses to evaluate and compare the effect of macroparticles versus nanoparticles formulation on kidney damage. Here, we demonstrated that smaller doses of nanoparticles exhibited the same beneficial effect as larger doses of macroparticles on preventing kidney damage. This finding translates into less quercetin consumption reaching the desired therapeutic effect.
Collapse
Affiliation(s)
- Esteban Andrés Sánchez-Jaramillo
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
| | - Luz Elena Gasca-Lozano
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
| | - José María Vera-Cruz
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico;
| | - Luis Daniel Hernández-Ortega
- Centro de Investigación Multidisciplinario en Salud, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico 555, Tonalá 45425, Jalisco, Mexico; (L.D.H.-O.); (M.M.-E.)
| | - Carmen Magdalena Gurrola-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
| | - Blanca Estela Bastidas-Ramírez
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
| | - Belinda Vargas-Guerrero
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
| | - Mayra Mena-Enríquez
- Centro de Investigación Multidisciplinario en Salud, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico 555, Tonalá 45425, Jalisco, Mexico; (L.D.H.-O.); (M.M.-E.)
| | | | - Adriana María Salazar-Montes
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro de Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico; (E.A.S.-J.); (L.E.G.-L.); (C.M.G.-D.); (B.E.B.-R.); (B.V.-G.)
- Correspondence:
| |
Collapse
|
17
|
Granata A, Pesce F, Iacoviello M, Anzaldi M, Amico F, Catalano M, Leonardi G, Gatta C, Costanza G, Corrao S, Gesualdo L. SGLT2 Inhibitors: A Broad Impact Therapeutic Option for the Nephrologist. FRONTIERS IN NEPHROLOGY 2022; 2:867075. [PMID: 37674992 PMCID: PMC10479658 DOI: 10.3389/fneph.2022.867075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/17/2022] [Indexed: 09/08/2023]
Abstract
Since their introduction as antidiabetic drugs, SGLT2 inhibitors (SGLT2i) have come a long way, proving to be beneficial on cardiovascular and renal outcomes independently of diabetes status. The benefits go far beyond glycemic control, and both the cardio- and nephroprotection are underpinned by diverse mechanisms. From the activation of tubule glomerular feedback and the consequent reduction in hyperfiltration to the improvement of hypoxia and oxidative stress in the renal cortex, SGLT2i have also been shown to inhibit hepcidin and limit podocyte damage. Likewise, they improve cardiac metabolism and bioenergetics, and reduce necrosis and cardiac fibrosis and the production of adipokines, cytokines, and epicardial adipose tissue mass. In terms of outcomes, the efficacy has been demonstrated on blood pressure control, BMI, albuminuria, stroke, heart disease, and mortality rate due to cardiovascular events. Patients with chronic kidney disease and proteinuria, with or without diabetes, treated with some SGLT2i have a reduced risk of progression. The analysis of subgroups of individuals with specific diseases such as IgA nephropathy has confirmed this solid effect on renal outcomes. Given these overarching activities on such a broad pathophysiological background and the favorable safety profile that goes with the use of SGLT2i, it is now certain that they are changing our approach to clinical interventions for important outcomes with an impressive impact.
Collapse
Affiliation(s)
- Antonio Granata
- Nephrology and Dialysis Unit, “Cannizzaro” Emergency Hospital, Catania, Italy
| | - Francesco Pesce
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Massimo Iacoviello
- Cardiology Unit, Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | | | - Francesco Amico
- Cardiology Unit, “Cannizzaro” Emergency Hospital, Catania, Italy
| | - Maria Catalano
- Cardiology Unit, “Cannizzaro” Emergency Hospital, Catania, Italy
| | - Giuseppe Leonardi
- Cardiology Unit, Azienda Ospedaliera Universitaria (A.O.U.) “Policlinico-San Marco”, Catania, Italy
| | - Carmela Gatta
- Internal Medicine Unit, Azienda Ospedaliera Universitaria (A.O.U.) “Policlinico-San Marco”, Catania, Italy
| | - Giusy Costanza
- Nephrology and Dialysis, “Vittorio Emanuele” Hospital, Gela, Italy
| | - Salvatore Corrao
- Department of Internal Medicine, “Azienda di Rilievo Nazionale ed Alta Specializzazione (ARNAS) Civico, Di Cristina e Benfratelli”, Palermo, Italy
| | - Loreto Gesualdo
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
18
|
Kim K, Jeong B, Lee YM, Son HE, Ryu JY, Park S, Jeong JC, Chin HJ, Kim S. Three-Dimensional Kidney-on-a-Chip Assessment of Contrast-Induced Kidney Injury: Osmolality and Viscosity. MICROMACHINES 2022; 13:mi13050688. [PMID: 35630155 PMCID: PMC9146534 DOI: 10.3390/mi13050688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 12/19/2022]
Abstract
Increased viscosity of concentrated contrast media (CM) in the renal tubules can perturb renal hemodynamics and have a detrimental effect on tubular epithelial cells. However, the effects of viscosity on contrast-induced nephropathy (CIN) remain poorly understood. Conventional in vitro culture studies do not reflect the rheological properties of CM. Therefore, we investigated the effects of CM viscosity on renal tubules using a kidney-on-a-chip and two different types of CM. Renal proximal tubule epithelial cells (RPTEC) were cultured in a three-dimensional microfluidic culture platform under bidirectional fluid shear stress. We treated the RPTEC with two types of CM: low- (LOCM, iopromide) and iso-osmolar contrast media (IOCM, iodixanol). Renal tubular cell injury induced by LOCM and IOCM was examined under different iodine concentrations (50–250 mgI/mL) and shear-stress conditions. LOCM showed a significant dose-dependent cytotoxic effect, which was significantly higher than that of IOCM under static and low-to-moderate shear stress conditions. However, high shear-stress resulted in reduced cell viability in IOCM; no difference between IOCM and LOCM was found under high shear-stress conditions. The cytotoxic effects were pronounced at a mean shear stress of 1 dyn/cm2 or higher. The high viscosity of IOCM slowed the fluid flow rate and augmented fluid shear-stress. We suggest an alternative in vitro model of CIN using the three-dimensional kidney-on-a-chip. Our results indicate a vital role of viscosity-induced nephrotoxicity under high shear-stress conditions, contrary to the findings of conventional in vitro studies.
Collapse
Affiliation(s)
- Kipyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University College of Medicine, Incheon 22332, Korea;
| | - Beomgyun Jeong
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon 34133, Korea;
| | - Yun-Mi Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
| | - Hyung-Eun Son
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
| | - Ji-Young Ryu
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
| | - Seokwoo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jong Cheol Jeong
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
| | - Ho Jun Chin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (Y.-M.L.); (H.-E.S.); (J.-Y.R.); (S.P.); (J.C.J.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-31-787-7051; Fax: +82-31-787-4052
| |
Collapse
|
19
|
Sugimoto H, Yumura W, Yamaguchi Y, Yamanaka N, Hashimoto S, Arai M, Matsuno S, Shimizu A, Arai T, Itabashi M, Takei T. Identification of histopathological and clinical spectrum of diabetic kidney disease based on an unsupervised hierarchical clustering analysis of elderly autopsy specimens. Nephrology (Carlton) 2022; 27:701-711. [DOI: 10.1111/nep.14044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Hikaru Sugimoto
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Wako Yumura
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Yasuko Yamaguchi
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
- Department of Analytic Human Pathology Nippon Medical School
| | - Noriko Yamanaka
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Seiji Hashimoto
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Masahiro Arai
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Shiho Matsuno
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Akira Shimizu
- Department of Analytic Human Pathology Nippon Medical School
| | - Tomio Arai
- Department of Pathology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Mitsuyo Itabashi
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| | - Takashi Takei
- Departments of Nephrology Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology
| |
Collapse
|
20
|
Miyano T, Suzuki A, Sakamoto N. Hyperosmotic stress induces epithelial-mesenchymal transition through rearrangements of focal adhesions in tubular epithelial cells. PLoS One 2021; 16:e0261345. [PMID: 34932568 PMCID: PMC8691603 DOI: 10.1371/journal.pone.0261345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/30/2021] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) of tubular epithelial cells is a hallmark of renal tubulointerstitial fibrosis and is associated with chronic renal injury as well as acute renal injury. As one of the incidences and risk factors for acute renal injury, increasing the osmolality in the proximal tubular fluid by administration of intravenous mannitol has been reported, but the detailed mechanisms remain unclear. Hyperosmotic conditions caused by mannitol in the tubular tissue may generate not only osmotic but also mechanical stresses, which are known to be able to induce EMT in epithelial cells, thereby contributing to renal injury. Herein, we investigate the effect of hyperosmolarity on EMT in tubular epithelial cells. Normal rat kidney (NRK)-52E cells were exposed to mannitol-induced hyperosmotic stress. Consequently, the hyperosmotic stress led to a reduced expression of the epithelial marker E-cadherin and an enhanced expression of the mesenchymal marker, α-smooth muscle actin (α-SMA), which indicates an initiation of EMT in NKR-52E cells. The hyperosmotic condition also induced time-dependent disassembly and rearrangements of focal adhesions (FAs) concomitant with changes in actin cytoskeleton. Moreover, prevention of FAs rearrangements by cotreatment with Y-27632, a Rho-associated protein kinase inhibitor, could abolish the effects of hyperosmotic mannitol treatment, thus attenuating the expression of α-SMA to the level in nontreated cells. These results suggest that hyperosmotic stress may induce EMT through FAs rearrangement in proximal tubular epithelial cells.
Collapse
Affiliation(s)
- Takashi Miyano
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Atsushi Suzuki
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
21
|
Hanouneh M, Echouffo Tcheugui JB, Jaar BG. Recent advances in diabetic kidney disease. BMC Med 2021; 19:180. [PMID: 34399760 PMCID: PMC8369797 DOI: 10.1186/s12916-021-02050-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 01/23/2023] Open
Affiliation(s)
- Mohamad Hanouneh
- Department of Medicine, Johns Hopkins University School of Medicine, 5601 Loch Raven Boulevard, Suite 3 North, Baltimore, MD, 21239, USA.,Nephrology Center of Maryland, Baltimore, MD, USA
| | - Justin B Echouffo Tcheugui
- Department of Medicine, Johns Hopkins University School of Medicine, 5601 Loch Raven Boulevard, Suite 3 North, Baltimore, MD, 21239, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, MD, USA
| | - Bernard G Jaar
- Department of Medicine, Johns Hopkins University School of Medicine, 5601 Loch Raven Boulevard, Suite 3 North, Baltimore, MD, 21239, USA. .,Nephrology Center of Maryland, Baltimore, MD, USA. .,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. .,Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, MD, USA.
| |
Collapse
|
22
|
Wang Y, Li N, Tian X, Lin L, Liang S, Zhao P, Dong Z, Wang Q, Li Q, Tang J, Luo Y. Evaluation of Renal Microperfusion in Diabetic Patients With Kidney Injury by Contrast-Enhanced Ultrasound. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2021; 40:1361-1368. [PMID: 33035374 DOI: 10.1002/jum.15516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/13/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES To conduct a quantitative analysis of renal microvascular perfusion in diabetic patients with kidney injury using contrast-enhanced ultrasound (CEUS). METHODS A total of 172 patients with type 2 diabetes mellitus and kidney injury were recruited from May 2017 to November 2019. After collection of clinical characteristics, a CEUS examination was performed after injection of the contrast agent SonoVue (Bracco SpA, Milan, Italy). Time-intensity curves and renal perfusion parameters were analyzed. Ultrasound-guided renal biopsy was performed. The patients were divided into a diabetic nephropathy (DN) group and a nondiabetic renal disease (NDRD) group according to renal pathologic results. The discrimination of perfusion parameters between the groups was analyzed statistically with SPSS version 19.0 software (IBM Corporation, Armonk, NY). Receiver operating characteristic curves were used to illustrate the diagnostic performance of indicators. RESULTS Ninety-eight patients, including 45 with DN (29 male; mean age ± SD, 57.76 ± 10.47 years) and 53 with NDRD (40 male; mean age, 48.7 ± 13.88 years) were included in this study. The peak enhancement (PE), wash-in the area under the curve (AUC), wash-in rate, wash-in perfusion index, wash-out AUC, wash-in and wash-out AUC, and wash-out rate were significantly different between the groups (P < .05). There were no differences in time-related parameters between the DN and NDRD groups (P > .05). The receiver operating characteristic curve analysis showed that the AUC for PE was 0.727, and PE lower than 7712.426 had diagnostic potential, with sensitivity of 81% and specificity of 40% in discriminating between NDRD and DN. CONCLUSIONS The quantification of CEUS parameters can discriminate DN in diabetic patients with kidney injury. The PE and AUC may be feasible parameters.
Collapse
Affiliation(s)
- Yiru Wang
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Nan Li
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaoqi Tian
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lin Lin
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuyuan Liang
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ping Zhao
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zheyi Dong
- Department of Nephrology, First Medical Center, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Diseases, Beijing, China
- State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, China
| | - Qian Wang
- Department of Nephrology, First Medical Center, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Diseases, Beijing, China
- State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, China
| | - Qiuyang Li
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Tang
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yukun Luo
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, China
- State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, China
| |
Collapse
|
23
|
A 3D Renal Proximal Tubule on Chip Model Phenocopies Lowe Syndrome and Dent II Disease Tubulopathy. Int J Mol Sci 2021; 22:ijms22105361. [PMID: 34069732 PMCID: PMC8161077 DOI: 10.3390/ijms22105361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/28/2022] Open
Abstract
Lowe syndrome and Dent II disease are X-linked monogenetic diseases characterised by a renal reabsorption defect in the proximal tubules and caused by mutations in the OCRL gene, which codes for an inositol-5-phosphatase. The life expectancy of patients suffering from Lowe syndrome is largely reduced because of the development of chronic kidney disease and related complications. There is a need for physiological human in vitro models for Lowe syndrome/Dent II disease to study the underpinning disease mechanisms and to identify and characterise potential drugs and drug targets. Here, we describe a proximal tubule organ on chip model combining a 3D tubule architecture with fluid flow shear stress that phenocopies hallmarks of Lowe syndrome/Dent II disease. We demonstrate the high suitability of our in vitro model for drug target validation. Furthermore, using this model, we demonstrate that proximal tubule cells lacking OCRL expression upregulate markers typical for epithelial–mesenchymal transition (EMT), including the transcription factor SNAI2/Slug, and show increased collagen expression and deposition, which potentially contributes to interstitial fibrosis and disease progression as observed in Lowe syndrome and Dent II disease.
Collapse
|
24
|
Shao BY, Zhang SF, Li HD, Meng XM, Chen HY. Epigenetics and Inflammation in Diabetic Nephropathy. Front Physiol 2021; 12:649587. [PMID: 34025445 PMCID: PMC8131683 DOI: 10.3389/fphys.2021.649587] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) leads to high morbidity and disability. Inflammation plays a critical role in the pathogenesis of DN, which involves renal cells and immune cells, the microenvironment, as well as extrinsic factors, such as hyperglycemia, chemokines, cytokines, and growth factors. Epigenetic modifications usually regulate gene expression via DNA methylation, histone modification, and non-coding RNAs without altering the DNA sequence. During the past years, numerous studies have been published to reveal the mechanisms of epigenetic modifications that regulate inflammation in DN. This review aimed to summarize the latest evidence on the interplay of epigenetics and inflammation in DN, and highlight the potential targets for treatment and diagnosis of DN.
Collapse
Affiliation(s)
- Bao-Yi Shao
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shao-Fei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Yong Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
25
|
Oliva-Damaso N, Mora-Gutiérrez JM, Bomback AS. Glomerular Diseases in Diabetic Patients: Implications for Diagnosis and Management. J Clin Med 2021; 10:1855. [PMID: 33923227 PMCID: PMC8123132 DOI: 10.3390/jcm10091855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
The prevalence of diabetes continues to rise worldwide. In addition to rising rates of diabetic kidney disease, we are also seeing a parallel rise in nondiabetic kidney disease among patients with diabetes. These nondiabetic lesions include focal segmental glomerulosclerosis, IgA nephropathy, membranous nephropathy, and other glomerular diseases. The management of diabetic kidney disease is rapidly evolving to include, beyond glycemic control and renin angiotensin inhibition, the use of sodium-glucose cotransporter 2 (SGLT2) inhibitors and mineralocorticoid receptor antagonists. These and other new treatment strategies should be applicable to managing glomerular disease in diabetic patients to reduce toxicities associated with immunosuppression and, in particular, corticosteroids. The prevalence of glomerular disease in diabetic patients is underappreciated. Diagnosis and appropriately treating these diseases remain an important avenue to modify kidney outcomes in diabetic patients.
Collapse
Affiliation(s)
- Nestor Oliva-Damaso
- Department of Medicine, Division of Nephrology, Hospital Costa del Sol, 29603 Marbella, Malaga, Spain;
| | - José María Mora-Gutiérrez
- Department of Medicine, Division of Nephrology, Clínica Universidad de Navarra, 31008 Pamplona, Navarra, Spain;
| | - Andrew S. Bomback
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
26
|
Wu L, Liu T, Gu Y. Microvillar dynamic in renal tubular epithelial cells mediated by insulin/PLCγ signal pathway. Biochem Biophys Res Commun 2020; 534:1020-1025. [PMID: 33131771 DOI: 10.1016/j.bbrc.2020.10.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/17/2020] [Indexed: 10/23/2022]
Abstract
Significant cellular morphology changes in renal tubules were observed in diabetes patients and animal models. However, the interaction between insulin and tubular epithelial cells microvillar structure remains obscure. To understand microvillar dynamics, we used Scanning Ion Conductance Microscope to visualize microvillar in the living cell. Here, we found two layers of microvilli on the tubular epithelial cell surface: short compact microvilli and netlike long microvilli. Insulin treatment could increase microvilli length and density. This process was mediated by the PI3K/PLCγ signaling pathway, other than the PI3K/Arp2/3 signal pathway. In conclusion, our findings present a novel insulin signaling transduction mechanism, which contributes to understanding renal tubular epithelial cell microvilli dynamic regulation.
Collapse
Affiliation(s)
- Lida Wu
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, 100871, Beijing, China; Translational and Regenerative Medicine Centre, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Tongri Liu
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, 100871, Beijing, China
| | - Yuchun Gu
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, 100871, Beijing, China; Translational and Regenerative Medicine Centre, Aston Medical School, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
27
|
Barry DM, McMillan EA, Kunar B, Lis R, Zhang T, Lu T, Daniel E, Yokoyama M, Gomez-Salinero JM, Sureshbabu A, Cleaver O, Di Lorenzo A, Choi ME, Xiang J, Redmond D, Rabbany SY, Muthukumar T, Rafii S. Molecular determinants of nephron vascular specialization in the kidney. Nat Commun 2019; 10:5705. [PMID: 31836710 PMCID: PMC6910926 DOI: 10.1038/s41467-019-12872-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 09/22/2019] [Indexed: 12/13/2022] Open
Abstract
Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders. The kidney is vascularized with highly specialized and zonated endothelial cells that are essential for its filtration function. Here, Barry et al. provide a single-cell RNA sequencing analysis of the kidney vasculature that highlights its transcriptional heterogeneity and uncovers pathways important for its development and function.
Collapse
Affiliation(s)
- David M Barry
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Elizabeth A McMillan
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Balvir Kunar
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Raphael Lis
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tyler Lu
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Edward Daniel
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Masataka Yokoyama
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Angara Sureshbabu
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Annarita Di Lorenzo
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - David Redmond
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sina Y Rabbany
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.,Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, 11549, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
28
|
Higgins CE, Tang J, Mian BM, Higgins SP, Gifford CC, Conti DJ, Meldrum KK, Samarakoon R, Higgins PJ. TGF-β1-p53 cooperativity regulates a profibrotic genomic program in the kidney: molecular mechanisms and clinical implications. FASEB J 2019; 33:10596-10606. [PMID: 31284746 PMCID: PMC6766640 DOI: 10.1096/fj.201900943r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease affects >15% of the U.S. population and >850 million individuals worldwide. Fibrosis is the common outcome of many chronic renal disorders and, although the etiology varies (i.e., diabetes, hypertension, ischemia, acute injury, and urologic obstructive disorders), persistently elevated renal TGF-β1 levels result in the relentless progression of fibrotic disease. TGF-β1 orchestrates the multifaceted program of renal fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery and redifferentiation, and subsequent tubulointerstitial fibrosis, eventually leading to chronic renal disease. Recent findings implicate p53 as a cofactor in the TGF-β1-induced signaling pathway and a transcriptional coregulator of several TGF-β1 profibrotic response genes by complexing with receptor-activated SMADs, which are homologous to the small worms (SMA) and Drosophilia mothers against decapentaplegic (MAD) gene families. The cooperative p53-TGF-β1 genomic cluster includes genes involved in cell growth control and extracellular matrix remodeling [e.g., plasminogen activator inhibitor-1 (PAI-1; serine protease inhibitor, clade E, member 1), connective tissue growth factor, and collagen I]. Although the molecular basis for this codependency is unclear, many TGF-β1-responsive genes possess p53 binding motifs. p53 up-regulation and increased p53 phosphorylation; moreover, they are evident in nephrotoxin- and ischemia/reperfusion-induced injury, diabetic nephropathy, ureteral obstructive disease, and kidney allograft rejection. Pharmacologic and genetic approaches that target p53 attenuate expression of the involved genes and mitigate the fibrotic response, confirming a key role for p53 in renal disorders. This review focuses on mechanisms whereby p53 functions as a transcriptional regulator within the TGF-β1 cluster with an emphasis on the potent fibrosis-promoting PAI-1 gene.-Higgins, C. E., Tang, J., Mian, B. M., Higgins, S. P., Gifford, C. C., Conti, D. J., Meldrum, K. K., Samarakoon, R., Higgins, P. J. TGF-β1-p53 cooperativity regulates a profibrotic genomic program in the kidney: molecular mechanisms and clinical implications.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, New York, USA
- Division of Urology, Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Kirstan K. Meldrum
- Division of Pediatric Urology, Central Michigan University, Mount Pleasant, Michigan, USA
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
- The Urological Institute of Northeastern New York, Albany, New York, USA
- Division of Urology, Department of Surgery, Albany Medical College, Albany, New York, USA
| |
Collapse
|
29
|
Aroor AR, Habibi J, Nistala R, Ramirez-Perez FI, Martinez-Lemus LA, Jaffe IZ, Sowers JR, Jia G, Whaley-Connell A. Diet-Induced Obesity Promotes Kidney Endothelial Stiffening and Fibrosis Dependent on the Endothelial Mineralocorticoid Receptor. Hypertension 2019; 73:849-858. [PMID: 30827147 PMCID: PMC6448566 DOI: 10.1161/hypertensionaha.118.12198] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/04/2019] [Indexed: 12/29/2022]
Abstract
Obesity is characterized by enhanced MR (mineralocorticoid receptor) activation, vascular stiffness, and associated cardiovascular and kidney disease. Consumption of a Western-style diet (WD), high in saturated fat and refined carbohydrates, by female mice, leads to obesity and vascular stiffening. Use of ECMR (endothelial cell-specific MR) knockout mice supports that ECMR activation is critical for development of vascular and cardiac fibrosis and stiffening. However, the role of ECMR activation in kidney inflammation and fibrosis remains unknown. We hypothesized that cell-specific deletion of ECMR would prevent WD-induced central aortic stiffness and protect the kidney from endothelial dysfunction and vascular stiffening. Four-week-old female ECMR KO and wild-type mice were fed either mouse chow or WD for 16 weeks. WD feeding increased body weight and fat mass, proteinuria, as well as vascular stiffness indices (pulse wave velocity and kidney artery stiffening) and impaired endothelial-dependent vasodilatation without blood pressure changes. The WD-induced kidney arterial stiffening was associated with attenuated eNOS (endothelial NO synthase) activation, increased oxidative stress, proinflammatory immune responses, alterations in extracellular matrix degradation pathways, and fibrosis. ECMR deletion prevented these abnormalities by improving eNOS activation and reducing macrophage proinflammatory M1 polarization, expression of TG2 (transglutaminase 2), and MMP (matrix metalloproteinase)-9. Our data support the concept that ECMR activation contributes to endothelial dysfunction, increased kidney artery fibrosis/stiffening, and impaired NOS (NO synthase) activation, processes associated with macrophage infiltration and polarization, inflammation, and oxidative stress, collectively resulting in tubulointerstitial fibrosis in females consuming a WD.
Collapse
Affiliation(s)
- Annayya R Aroor
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Javad Habibi
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Ravi Nistala
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Nephrology and Hypertension (R.N., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology (L.A.M.-L., J.R.S.)
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.)
| | - James R Sowers
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Department of Medical Pharmacology and Physiology (L.A.M.-L., J.R.S.)
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Guanghong Jia
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
| | - Adam Whaley-Connell
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Division of Nephrology and Hypertension (R.N., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| |
Collapse
|
30
|
Profibrotic epithelial phenotype: a central role for MRTF and TAZ. Sci Rep 2019; 9:4323. [PMID: 30867502 PMCID: PMC6416270 DOI: 10.1038/s41598-019-40764-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/19/2019] [Indexed: 01/05/2023] Open
Abstract
Epithelial injury is a key initiator of fibrosis but - in contrast to the previous paradigm - the epithelium in situ does not undergo wide-spread epithelial-mesenchymal/myofibroblast transition (EMT/EMyT). Instead, it assumes a Profibrotic Epithelial Phenotype (PEP) characterized by fibrogenic cytokine production. The transcriptional mechanisms underlying PEP are undefined. As we have shown that two RhoA/cytoskeleton-regulated transcriptional coactivators, Myocardin-related transcription factor (MRTF) and TAZ, are indispensable for EMyT, we asked if they might mediate PEP as well. Here we show that mechanical stress (cyclic stretch) increased the expression of transforming growth factor-β1 (TGFβ1), connective tissue growth factor (CTGF), platelet-derived growth factor and Indian Hedgehog mRNA in LLC-PK1 tubular cells. These responses were mitigated by siRNA-mediated silencing or pharmacological inhibition of MRTF (CCG-1423) or TAZ (verteporfin). RhoA inhibition exerted similar effects. Unilateral ureteral obstruction, a murine model of mechanically-triggered kidney fibrosis, induced tubular RhoA activation along with overexpression/nuclear accumulation of MRTF and TAZ, and increased transcription of the above-mentioned cytokines. Laser capture microdissection revealed TAZ, TGFβ1 and CTGF induction specifically in the tubular epithelium. CCG-1423 suppressed total renal and tubular expression of these proteins. Thus, MRTF regulates epithelial TAZ expression, and both MRTF and TAZ are critical mediators of PEP-related epithelial cytokine production.
Collapse
|
31
|
Current Opinion for Hypertension in Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:37-47. [DOI: 10.1007/978-981-13-8871-2_3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Alicic RZ, Rooney MT, Tuttle KR. Diabetic Kidney Disease: Challenges, Progress, and Possibilities. Clin J Am Soc Nephrol 2017; 12:2032-2045. [PMID: 28522654 PMCID: PMC5718284 DOI: 10.2215/cjn.11491116] [Citation(s) in RCA: 1566] [Impact Index Per Article: 223.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic kidney disease develops in approximately 40% of patients who are diabetic and is the leading cause of CKD worldwide. Although ESRD may be the most recognizable consequence of diabetic kidney disease, the majority of patients actually die from cardiovascular diseases and infections before needing kidney replacement therapy. The natural history of diabetic kidney disease includes glomerular hyperfiltration, progressive albuminuria, declining GFR, and ultimately, ESRD. Metabolic changes associated with diabetes lead to glomerular hypertrophy, glomerulosclerosis, and tubulointerstitial inflammation and fibrosis. Despite current therapies, there is large residual risk of diabetic kidney disease onset and progression. Therefore, widespread innovation is urgently needed to improve health outcomes for patients with diabetic kidney disease. Achieving this goal will require characterization of new biomarkers, designing clinical trials that evaluate clinically pertinent end points, and development of therapeutic agents targeting kidney-specific disease mechanisms (e.g., glomerular hyperfiltration, inflammation, and fibrosis). Additionally, greater attention to dissemination and implementation of best practices is needed in both clinical and community settings. INTRODUCTION
Collapse
Affiliation(s)
- Radica Z. Alicic
- Providence Health Care, Spokane, Washington
- University of Washington School of Medicine, Seattle, Washington
| | | | - Katherine R. Tuttle
- Providence Health Care, Spokane, Washington
- University of Washington School of Medicine, Seattle, Washington
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington
- Institute of Translational Health Sciences, Seattle, Washington; and
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
33
|
Kunnen SJ, Malas TB, Semeins CM, Bakker AD, Peters DJM. Comprehensive transcriptome analysis of fluid shear stress altered gene expression in renal epithelial cells. J Cell Physiol 2017; 233:3615-3628. [PMID: 29044509 PMCID: PMC5765508 DOI: 10.1002/jcp.26222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/05/2017] [Indexed: 12/21/2022]
Abstract
Renal epithelial cells are exposed to mechanical forces due to flow‐induced shear stress within the nephrons. Shear stress is altered in renal diseases caused by tubular dilation, obstruction, and hyperfiltration, which occur to compensate for lost nephrons. Fundamental in regulation of shear stress are primary cilia and other mechano‐sensors, and defects in cilia formation and function have profound effects on development and physiology of kidneys and other organs. We applied RNA sequencing to get a comprehensive overview of fluid‐shear regulated genes and pathways in renal epithelial cells. Functional enrichment‐analysis revealed TGF‐β, MAPK, and Wnt signaling as core signaling pathways up‐regulated by shear. Inhibitors of TGF‐β and MAPK/ERK signaling modulate a wide range of mechanosensitive genes, identifying these pathways as master regulators of shear‐induced gene expression. However, the main down‐regulated pathway, that is, JAK/STAT, is independent of TGF‐β and MAPK/ERK. Other up‐regulated cytokine pathways include FGF, HB‐EGF, PDGF, and CXC. Cellular responses to shear are modified at several levels, indicated by altered expression of genes involved in cell‐matrix, cytoskeleton, and glycocalyx remodeling, as well as glycolysis and cholesterol metabolism. Cilia ablation abolished shear induced expression of a subset of genes, but genes involved in TGF‐β, MAPK, and Wnt signaling were hardly affected, suggesting that other mechano‐sensors play a prominent role in the shear stress response of renal epithelial cells. Modulations in signaling due to variations in fluid shear stress are relevant for renal physiology and pathology, as suggested by elevated gene expression at pathological levels of shear stress compared to physiological shear.
Collapse
Affiliation(s)
- Steven J Kunnen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Tareq B Malas
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis M Semeins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
34
|
Fong D, Ullah MM, Lal JG, Abdelkader A, Ow CP, Hilliard LM, Ricardo SD, Kelly DJ, Evans RG. Renal cellular hypoxia in adenine-induced chronic kidney disease. Clin Exp Pharmacol Physiol 2016; 43:896-905. [DOI: 10.1111/1440-1681.12621] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/30/2016] [Accepted: 07/02/2016] [Indexed: 01/20/2023]
Affiliation(s)
- Debra Fong
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Md Mahbub Ullah
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Jaswini G. Lal
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Amany Abdelkader
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Connie P.C. Ow
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Lucinda M. Hilliard
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| | - Sharon D. Ricardo
- Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology; Monash University; Clayton Melbourne Vic. Australia
| | - Darren J. Kelly
- Department of Medicine; University of Melbourne; St Vincent's Hospital; Fitzroy Melbourne Vic. Australia
| | - Roger G. Evans
- Biomedicine Discovery Institute and Department of Physiology; Monash University; Clayton Melbourne Vic. Australia
| |
Collapse
|
35
|
Matsuki-Muramoto Y, Nozawa K, Uomori K, Sekigawa I, Takasaki Y. Bortezomib treatment prevents glomerulosclerosis associated with lupus nephritis in a murine model through suppressive effects on the immune and renin-angiotensin systems. Mod Rheumatol 2016; 27:77-86. [PMID: 27166507 DOI: 10.3109/14397595.2016.1170957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To clarify the mechanisms underlying lupus nephritis (LN) amelioration following bortezomib treatment. METHODS Bortezomib was administered subcutaneously every 3 days to NZB/W F1 mice, and the serum anti-double stranded (ds) deoxyribonucleic acid (DNA) antibody titers and proteinuria levels were measured. The renal samples and the splenocytes were examined histologically or used for real-time quantitative reverse transcription-polymerase chain reaction analysis after 18 weeks of treatment. Serum cytokine and anti-dsDNA antibody levels were measured using flow cytometry and enzyme-linked immunoassays every 3 weeks. Transforming growth factor (TGF)-β, angiotensin II type-1 receptor (AT1R), and type I collagen expression levels in the glomeruli were evaluated using immunohistochemistry. RESULTS Bortezomib reduced the serum anti-dsDNA antibody titers and the proteinuria levels. It prevented inflammatory cell infiltrations into and the deposition of immunoglobulin G within the glomeruli. Bortezomib reduced the interferon-γ, interleukin (IL)-4, and IL-10 levels in the serum and the ribonucleic acid expression levels for these cytokines within the splenocytes. Bortezomib prevented type I collagen synthesis by downregulating TGF-β and AT1R expression in the glomeruli. CONCLUSIONS Bortezomib exerts multiple immunosuppressive effects and thus ameliorates LN. Furthermore, bortezomib can prevent glomerulosclerosis formation in NZB/W F1 mice through suppressive effects on the renin-angiotensin system.
Collapse
Affiliation(s)
- Yuko Matsuki-Muramoto
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Kazuhisa Nozawa
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Kaori Uomori
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Iwao Sekigawa
- b Department of Internal Medicine and Rheumatology , Juntendo University Urayasu Hospital , Chiba , Japan , and.,c Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Yoshinari Takasaki
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| |
Collapse
|
36
|
Sureshbabu A, Muhsin SA, Choi ME. TGF-β signaling in the kidney: profibrotic and protective effects. Am J Physiol Renal Physiol 2016; 310:F596-F606. [PMID: 26739888 DOI: 10.1152/ajprenal.00365.2015] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is generally considered as a central mediator of fibrotic diseases. Indeed, much focus has been placed on inhibiting TGF-β and its downstream targets as ideal therapeutic strategies. However, pharmacological blockade of TGF-β has not yet translated into successful therapy for humans, which may be due to pleiotropic effects of TGF-β signaling. Equally, TGF-β signaling as a protective response in kidney injury has been relatively underexplored. An emerging body of evidence from experimental kidney disease models indicates multifunctionality of TGF-β capable of inducing profibrotic and protective effects. This review discusses recent advances highlighting the diverse roles of TGF-β in promoting not only renal fibrosis but also protective responses of TGF-β signaling. We review, in particular, growing evidence that supports protective effects of TGF-β by mechanisms which include inhibiting inflammation and induction of autophagy. Additional detailed studies are required to fully understand the diverse mechanisms of TGF-β actions in renal fibrosis and inflammation that will likely direct toward effective antifibrotic therapies.
Collapse
Affiliation(s)
- Angara Sureshbabu
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and
| | - Saif A Muhsin
- New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and .,New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| |
Collapse
|
37
|
Maggiorani D, Dissard R, Belloy M, Saulnier-Blache JS, Casemayou A, Ducasse L, Grès S, Bellière J, Caubet C, Bascands JL, Schanstra JP, Buffin-Meyer B. Shear Stress-Induced Alteration of Epithelial Organization in Human Renal Tubular Cells. PLoS One 2015; 10:e0131416. [PMID: 26146837 PMCID: PMC4493045 DOI: 10.1371/journal.pone.0131416] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022] Open
Abstract
Tubular epithelial cells in the kidney are continuously exposed to urinary fluid shear stress (FSS) generated by urine movement and recent in vitro studies suggest that changes of FSS could contribute to kidney injury. However it is unclear whether FSS alters the epithelial characteristics of the renal tubule. Here, we evaluated in vitro and in vivo the influence of FSS on epithelial characteristics of renal proximal tubular cells taking the organization of junctional complexes and the presence of the primary cilium as markers of epithelial phenotype. Human tubular cells (HK-2) were subjected to FSS (0.5 Pa) for 48h. Control cells were maintained under static conditions. Markers of tight junctions (Claudin-2, ZO-1), Par polarity complex (Pard6), adherens junctions (E-Cadherin, β-Catenin) and the primary cilium (α-acetylated Tubulin) were analysed by quantitative PCR, Western blot or immunocytochemistry. In response to FSS, Claudin-2 disappeared and ZO-1 displayed punctuated and discontinuous staining in the plasma membrane. Expression of Pard6 was also decreased. Moreover, E-Cadherin abundance was decreased, while its major repressors Snail1 and Snail2 were overexpressed, and β-Catenin staining was disrupted along the cell periphery. Finally, FSS subjected-cells exhibited disappeared primary cilium. Results were confirmed in vivo in a uninephrectomy (8 months) mouse model where increased FSS induced by adaptive hyperfiltration in remnant kidney was accompanied by both decreased epithelial gene expression including ZO-1, E-cadherin and β-Catenin and disappearance of tubular cilia. In conclusion, these results show that proximal tubular cells lose an important number of their epithelial characteristics after long term exposure to FSS both in vitro and in vivo. Thus, the changes in urinary FSS associated with nephropathies should be considered as potential insults for tubular cells leading to disorganization of the tubular epithelium.
Collapse
Affiliation(s)
- Damien Maggiorani
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Romain Dissard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Marcy Belloy
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Jean-Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Laure Ducasse
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Sandra Grès
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Julie Bellière
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Cécile Caubet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
| | - Bénédicte Buffin-Meyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France
- Université Toulouse III Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases - I2MC, Toulouse, France
- * E-mail:
| |
Collapse
|
38
|
Antoine MH, Debelle F, Piccirilli J, El Kaddouri F, Declèves AE, De Prez E, Husson C, Mies F, Bourgeade MF, Nortier JL. Human bone morphogenetic protein-7 does not counteract aristolochic acid-induced renal toxicity. J Appl Toxicol 2015; 35:1520-30. [PMID: 25663515 DOI: 10.1002/jat.3116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 11/08/2022]
Abstract
Aristolochic acids (AA) are nephrotoxic and profibrotic agents, leading to chronic kidney disease. As some controversial studies have reported a nephroprotective effect of exogenous recombinant human bone morphogenetic protein (rhBMP)-7 in several models of renal fibrosis, we investigated the putative effect of rhBMP-7 to prevent progressive tubulointerstitial damage after AA intoxication in vitro and in vivo. In vitro, the toxicity of AA on renal tubular cells was demonstrated by an increase in vimentin as well as a decrease in β-catenin expressions, reflecting a dedifferentiation process. Increased fibronectin and interleukin-6 levels were measured in the supernatants. Enhanced α-SMA mRNA levels associated to decreased E-cadherin mRNA levels were also measured. Incubation with rhBMP-7 only prevented the increase in vimentin and the decrease in β-catenin expressions. In vivo, in a rat model of AA nephropathy, severe tubulointerstitial lesions induced by AA after 10 and 35 days (collagen IV deposition and tubular atrophy), were not prevented by the rhBMP-7 treatment. Similarly, rhBMP-7 did not ameliorate the significant increase in urinary concentrations of transforming growth factor-β. In summary, our in vitro data demonstrated a poor beneficial effect of rhBMP-7 to reverse cell toxicity while, in vivo, there was no beneficial effect of rhBMP-7. Therefore, further investigations are needed to confirm the exact role of BMP-7 in progressive chronic kidney disease.
Collapse
Affiliation(s)
- Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Frédéric Debelle
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Julie Piccirilli
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Fadoua El Kaddouri
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Anne-Emilie Declèves
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels.,Laboratory of Molecular Physiology (URPhyM), University of Namur, Namur
| | - Eric De Prez
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Cécile Husson
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Frédérique Mies
- Laboratory of Physiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| |
Collapse
|
39
|
Bhise NS, Ribas J, Manoharan V, Zhang YS, Polini A, Massa S, Dokmeci MR, Khademhosseini A. Organ-on-a-chip platforms for studying drug delivery systems. J Control Release 2014; 190:82-93. [PMID: 24818770 DOI: 10.1016/j.jconrel.2014.05.004] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/24/2014] [Accepted: 05/02/2014] [Indexed: 01/03/2023]
Abstract
Novel microfluidic tools allow new ways to manufacture and test drug delivery systems. Organ-on-a-chip systems - microscale recapitulations of complex organ functions - promise to improve the drug development pipeline. This review highlights the importance of integrating microfluidic networks with 3D tissue engineered models to create organ-on-a-chip platforms, able to meet the demand of creating robust preclinical screening models. Specific examples are cited to demonstrate the use of these systems for studying the performance of drug delivery vectors and thereby reduce the discrepancies between their performance at preclinical and clinical trials. We also highlight the future directions that need to be pursued by the research community for these proof-of-concept studies to achieve the goal of accelerating clinical translation of drug delivery nanoparticles.
Collapse
Affiliation(s)
- Nupura S Bhise
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - João Ribas
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; Biocant - Biotechnology Innovation Center, 3060-197 Cantanhede, Portugal
| | - Vijayan Manoharan
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Yu Shrike Zhang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Alessandro Polini
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Solange Massa
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Mehmet R Dokmeci
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, USA; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|