1
|
Martinez SA, Karel IZ, Silvaroli JA, Ahmed E, Kim JY, Stayton A, Patel PS, Afjal MA, Horton T, Bohmer M, Vanichapol T, Sander V, Andrade GM, Allison CV, Mondal M, Thorson VC, Partey A, Nimkar K, Williams V, Quimby J, Ganesan L, Madhavan SM, Davidson AJ, Peterson BR, Adebiyi A, Rao R, Sweet DH, Singh P, Bennett KM, Zepeda-Orozco D, Mallipattu SK, Eisenmann ED, Sparreboom A, Rovin BH, Bajwa A, Pabla NS. Resazurin dye is an in vivo sensor of kidney tubular function. Kidney Int 2025; 107:508-520. [PMID: 39733791 PMCID: PMC11845305 DOI: 10.1016/j.kint.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 11/01/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Glomerular filtration rate (GFR) is the main functional index of kidney health and disease. Currently, no methods are available to directly measure tubular mass and function. Here, we report a serendipitous finding that the in vitro cell viability dye resazurin can be used in mice as an exogenous sensor of tubular function. Intravenously injected resazurin exhibited significant plasma protein binding and was found to mainly undergo tubular secretion. Mechanistic studies showed that the blue-colored, weakly fluorescent resazurin is taken up by tubular cells through organic anion transporters, followed by conversion to a highly fluorescent, pink-colored resorufin by mitochondrial and cytosolic reductases, converted to an orange-colored β-d-glucuronide with subsequent efflux into the urine. Here we report a simple method in which the intravenous injection of resazurin is followed by the measurement of fluorescent metabolites in the urine, providing a sensitive readout of tubular function. Three mouse models of acute kidney injury (rhabdomyolysis, bilateral ischemia-reperfusion injury, and cisplatin nephrotoxicity) were tested and the resazurin-based method was able to sensitively detect the loss of tubular function much earlier than the increase in serum creatinine levels. Strikingly, in mice with unilateral ischemia-reperfusion injury and genetic mutation-linked kidney hypoplasia (oligosyndactylism, a genetic model for congenital kidney hypoplasia), the resazurin-based method was able to detect loss of tubular mass and function despite normal GFR levels. Collectively, our findings establish the preclinical utility of resazurin as a sensitive exogenous marker of tubular function and support future examination in larger animals for potential clinical translation.
Collapse
Affiliation(s)
- Shirely Acosta Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Isaac Z Karel
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Eman Ahmed
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Amanda Stayton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Prisha S Patel
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Mohammad Amir Afjal
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thomas Horton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Margaret Bohmer
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Gabriel Mayoral Andrade
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Corynne Vermillion Allison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Milon Mondal
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria C Thorson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alexandra Partey
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kartik Nimkar
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria Williams
- Division of Clinical Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jessica Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University Veterinary Medical Center, Columbus, Ohio, USA
| | - Latha Ganesan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sethu M Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Blake R Peterson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California, USA
| | - Kevin M Bennett
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Diana Zepeda-Orozco
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, USA; Renal Section, Northport VA Medical Center, Northport, New York, USA
| | - Eric D Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Brad H Rovin
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Amandeep Bajwa
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
2
|
Tran M, Jiao B, Du H, Zhou D, Yechoor V, Wang Y. TEAD1 Prevents Necroptosis and Inflammation in Cisplatin-Induced Acute Kidney Injury Through Maintaining Mitochondrial Function. Int J Biol Sci 2025; 21:565-578. [PMID: 39781453 PMCID: PMC11705647 DOI: 10.7150/ijbs.104335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
Cisplatin is widely used for the treatment of solid tumors and its antitumor effects are well established. However, a known complication of cisplatin administration is acute kidney injury (AKI). In this study, we examined the role of TEA domain family member 1 (TEAD1) in the pathogenesis of cisplatin-induced AKI. TEAD1 expression was upregulated in tubular epithelial cells of kidneys with cisplatin-induced AKI. TEAD1 floxed mice (TEAD1CON) mice treated with cisplatin developed tubular cell damage and impaired kidney function. In contrast, proximal tubule specific TEAD1 knockout (TEAD1PKO) mice treated with cisplatin had enhanced tubular cell damage and kidney dysfunction. Additionally, TEAD1PKO mice treated with cisplatin had augmented necroptotic cell death and inflammatory response compared to TEAD1CON mice with cisplatin. Knockdown of TEAD1 in mouse tubular epithelial cells showed increased intracellular ROS levels, reduced ATP production and impaired mitochondrial bioenergetics compared to control cells treated with cisplatin. Mechanistically, TEAD1 interacts with peroxisomal proliferator-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, to promote mitochondrial function. Taken together, our results indicate TEAD1 plays an important role in the pathogenesis of cisplatin-induced AKI through regulation of necroptosis and inflammation, which is associated with mitochondrial metabolism. Therefore, TEAD1 may represent a novel therapeutic target for cisplatin-induced AKI.
Collapse
Affiliation(s)
- Melanie Tran
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Baihai Jiao
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Hao Du
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Vijay Yechoor
- Department of Medicine, University of Pittsburg, Pittsburg, PA, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, USA
- Renal Section, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
3
|
Silvaroli JA, Bisunke B, Kim JY, Stayton A, Jayne LA, Martinez SA, Nguyen C, Patel PS, Vanichapol T, Verma V, Akhter J, Bolisetty S, Madhavan SM, Kuscu C, Coss CC, Zepeda-Orozco D, Parikh SV, Satoskar AA, Davidson AJ, Eason JD, Szeto HH, Pabla NS, Bajwa A. Genome-Wide CRISPR Screen Identifies Phospholipid Scramblase 3 as the Biological Target of Mitoprotective Drug SS-31. J Am Soc Nephrol 2024; 35:681-695. [PMID: 38530359 PMCID: PMC11164119 DOI: 10.1681/asn.0000000000000338] [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: 07/06/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
Key Points Szeto–Schiller-31–mediated mitoprotection is phospholipid scramblase 3–dependent. Phospholipid scramblase 3 is required for recovery after AKI. Background The synthetic tetrapeptide Szeto–Schiller (SS)-31 shows promise in alleviating mitochondrial dysfunction associated with common diseases. However, the precise pharmacological basis of its mitoprotective effects remains unknown. Methods To uncover the biological targets of SS-31, we performed a genome-scale clustered regularly interspaced short palindromic repeats screen in human kidney-2, a cell culture model where SS-31 mitigates cisplatin-associated cell death and mitochondrial dysfunction. The identified hit candidate gene was functionally validated using knockout cell lines, small interfering RNA-mediated downregulation, and tubular epithelial–specific conditional knockout mice. Biochemical interaction studies were also performed to examine the interaction of SS-31 with the identified target protein. Results Our primary screen and validation studies in hexokinase 2 and primary murine tubular epithelial cells showed that phospholipid scramblase 3 (PLSCR3), an understudied inner mitochondrial membrane protein, was essential for the protective effects of SS-31. For in vivo validation, we generated tubular epithelial–specific knockout mice and found that Plscr3 gene ablation did not influence kidney function under normal conditions or affect the severity of cisplatin and rhabdomyolysis-associated AKI. However, Plscr3 gene deletion completely abrogated the protective effects of SS-31 during cisplatin and rhabdomyolysis-associated AKI. Biochemical studies showed that SS-31 directly binds to a previously uncharacterized N -terminal domain and stimulates PLSCR3 scramblase activity. Finally, PLSCR3 protein expression was found to be increased in the kidneys of patients with AKI. Conclusions PLSCR3 was identified as the essential biological target that facilitated the mitoprotective effects of SS-31 in vitro and in vivo .
Collapse
Affiliation(s)
- Josie A. Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Bijay Bisunke
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amanda Stayton
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Laura A. Jayne
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Shirely A. Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Christopher Nguyen
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Prisha S. Patel
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Vivek Verma
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Juheb Akhter
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | | | - Sethu M. Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Cem Kuscu
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Diana Zepeda-Orozco
- Department of Pediatrics, The Ohio State University College of Medicine and Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Samir V. Parikh
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Anjali A. Satoskar
- Division of Renal and Transplant Pathology, Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - James D. Eason
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hazel H. Szeto
- Social Profit Network Research Lab, Menlo Park, California
| | - Navjot S. Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amandeep Bajwa
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Microbiology, Immunology, and Biochemistry; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
4
|
Lv D, Jiang H, Yang X, Li Y, Niu W, Zhang D. Advances in understanding of dendritic cell in the pathogenesis of acute kidney injury. Front Immunol 2024; 15:1294807. [PMID: 38433836 PMCID: PMC10904453 DOI: 10.3389/fimmu.2024.1294807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Acute kidney injury (AKI) is characterized by a rapid decline in renal function and is associated with a high morbidity and mortality rate. At present, the underlying mechanisms of AKI remain incompletely understood. Immune disorder is a prominent feature of AKI, and dendritic cells (DCs) play a pivotal role in orchestrating both innate and adaptive immune responses, including the induction of protective proinflammatory and tolerogenic immune reactions. Emerging evidence suggests that DCs play a critical role in the initiation and development of AKI. This paper aimed to conduct a comprehensive review and analysis of the role of DCs in the progression of AKI and elucidate the underlying molecular mechanism. The ultimate objective was to offer valuable insights and guidance for the treatment of AKI.
Collapse
Affiliation(s)
- Dongfang Lv
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huihui Jiang
- Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianzhen Yang
- Department of Urology, Afliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Li
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Engineering Laboratory of Urinary Organ and Functional Reconstruction of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Weipin Niu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Key Laboratory of Dominant Diseases of traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Denglu Zhang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Key Laboratory of Dominant Diseases of traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Ai S, Li Y, Zheng H, Wang Z, Liu W, Tao J, Li Y, Wang Y. Global research trends and hot spots on autophagy and kidney diseases: a bibliometric analysis from 2000 to 2022. Front Pharmacol 2023; 14:1275792. [PMID: 38099142 PMCID: PMC10719858 DOI: 10.3389/fphar.2023.1275792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Background: Autophagy is an essential cellular process involving the self-degradation and recycling of organelles, proteins, and cellular debris. Recent research has shown that autophagy plays a significant role in the occurrence and development of kidney diseases. However, there is a lack of bibliometric analysis regarding the relationship between autophagy and kidney diseases. Methods: A bibliometric analysis was conducted by searching for literature related to autophagy and kidney diseases in the Web of Science Core Collection (WoSCC) database from 2000 to 2022. Data processing was carried out using R package "Bibliometrix", VOSviewers, and CiteSpace. Results: A total of 4,579 articles related to autophagy and kidney diseases were collected from various countries. China and the United States were the main countries contributing to the publications. The number of publications in this field showed a year-on-year increasing trend, with open-access journals playing a major role in driving the literature output. Nanjing Medical University in China, Osaka University in Japan, and the University of Pittsburgh in the United States were the main research institutions. The journal "International journal of molecular sciences" had the highest number of publications, while "Autophagy" was the most influential journal in the field. These articles were authored by 18,583 individuals, with Dong, Zheng; Koya, Daisuke; and Kume, Shinji being the most prolific authors, and Dong, Zheng being the most frequently co-cited author. Research on autophagy mainly focused on diabetic kidney diseases, acute kidney injury, and chronic kidney disease. "Autophagy", "apoptosis", and "oxidative stress" were the primary research hotspots. Topics such as "diabetic kidney diseases", "sepsis", "ferroptosis", "nrf2", "hypertension" and "pi3k" may represent potential future development trends. Research on autophagy has gradually focused on metabolic-related kidney diseases such as diabetic nephropathy and hypertension. Additionally, PI3K, NRF2, and ferroptosis have been recent research directions in the field of autophagy mechanisms. Conclusion: This is the first comprehensive bibliometric study summarizing the relationship between autophagy and kidney diseases. The findings aid in identifying recent research frontiers and hot topics, providing valuable references for scholars investigating the role of autophagy in kidney diseases.
Collapse
Affiliation(s)
- Sinan Ai
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - JiaYin Tao
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaotan Li
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
6
|
虞 亘, 王 鑫, 骆 金, 苏 萧, 陶 怀, 闻 志, 关 翰. [Role of SPP1 in acute kidney injury induced by renal ischemia-reperfusion in rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1947-1954. [PMID: 38081614 PMCID: PMC10713472 DOI: 10.12122/j.issn.1673-4254.2023.11.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Indexed: 12/18/2023]
Abstract
OBJECTIVE To investigate the role of SPP1 gene in acute kidney injury induced by renal ischemia-reperfusion injury (IRI). METHODS Twelve Sprague-Dawley rats were randomly divided into sham group and IRI group (n=6) and subjected to sham operation and renal ischemia for 30 min induced by penal pedicle clamping using non-traumatic microvascular clamps, respectively.Serum creatinine and blood urea nitrogen levels were detected, and PAS staining was used for pathological examination of the kidneys in the two groups.The renal expressions of SPP1, α-SMA and caspase-3 were detected using immunohistochemistry and immunofluorescent staining.In cultured renal tubular epithelial cells (HK-2 cells), Western blotting was performed to detect the changes in expressions of SPP1, caspase-3, and Kim-1 proteins following hypoxiareoxygenation (H/R) and transfection with si-NC or si-SPP1;flow cytometry was employed to analyze apoptosis of the treated cells. RESULTS Renal IRI caused significant elevations of serum creatinine and blood urea nitrogen levels (P<0.05) and induced severe shedding and necrosis of the renal tubular epithelial cells in the rats, resulting also in significantly up-regulated renal expressions of SPP1, α-SMA and caspase-3(P<0.05).In HK-2 cells, H/R significantly increased the protein expression levels of SPP1, caspase-3, and Kim-1(P<0.05), and compared si-NC transfection, transfection with SPP1 obviously reduced caspase-3 and Kim-1 expressions and lowered apoptosis rate of the cells with H/R exposure (P<0.05). CONCLUSION SPP1 is up-regulated in the kidneys of rats with renal IRI, and down-regulation of SPP1 expression can inhibit H/R-induced apoptosis of renal tubular epithelial cells.
Collapse
Affiliation(s)
- 亘明 虞
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 鑫玮 王
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 金光 骆
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 萧 苏
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 怀祥 陶
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 志远 闻
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 翰 关
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| |
Collapse
|
7
|
Li JSY, Robertson H, Trinh K, Raghubar AM, Nguyen Q, Matigian N, Patrick E, Thomson AW, Mallett AJ, Rogers NM. Tolerogenic dendritic cells protect against acute kidney injury. Kidney Int 2023; 104:492-507. [PMID: 37244471 DOI: 10.1016/j.kint.2023.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 04/12/2023] [Accepted: 05/11/2023] [Indexed: 05/29/2023]
Abstract
Ischemia reperfusion injury is a common precipitant of acute kidney injury that occurs following disrupted perfusion to the kidney. This includes blood loss and hemodynamic shock, as well as during retrieval for deceased donor kidney transplantation. Acute kidney injury is associated with adverse long-term clinical outcomes and requires effective interventions that can modify the disease process. Immunomodulatory cell therapies such as tolerogenic dendritic cells remain a promising tool, and here we tested the hypothesis that adoptively transferred tolerogenic dendritic cells can limit kidney injury. The phenotypic and genomic signatures of bone marrow-derived syngeneic or allogeneic, Vitamin-D3/IL-10-conditioned tolerogenic dendritic cells were assessed. These cells were characterized by high PD-L1:CD86, elevated IL-10, restricted IL-12p70 secretion and a suppressed transcriptomic inflammatory profile. When infused systemically, these cells successfully abrogated kidney injury without modifying infiltrating inflammatory cell populations. They also provided protection against ischemia reperfusion injury in mice pre-treated with liposomal clodronate, suggesting the process was regulated by live, rather than reprocessed cells. Co-culture experiments and spatial transcriptomic analysis confirmed reduced kidney tubular epithelial cell injury. Thus, our data provide strong evidence that peri-operatively administered tolerogenic dendritic cells have the ability to protect against acute kidney injury and warrants further exploration as a therapeutic option. This technology may provide a clinical advantage for bench-to-bedside translation to affect patient outcomes.
Collapse
Affiliation(s)
- Jennifer S Y Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Sydney Medical School, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| | - Harry Robertson
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Katie Trinh
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Arti M Raghubar
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Matigian
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia; Queensland Cyber Infrastructure Foundation Bioinformatics, Brisbane, Queensland, Australia
| | - Ellis Patrick
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrew J Mallett
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia; Department of Renal Medicine, Townsville University Hospital, Townsville, Queensland, Australia; College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Sydney Medical School, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
8
|
Kim JY, Silvaroli JA, Martinez GV, Bisunke B, Luna Ramirez AV, Jayne LA, Feng MJHH, Girotra B, Acosta Martinez SM, Vermillion CR, Karel IZ, Ferrell N, Weisleder N, Chung S, Christman JW, Brooks CR, Madhavan SM, Hoyt KR, Cianciolo RE, Satoskar AA, Zepeda-Orozco D, Sullivan JC, Davidson AJ, Bajwa A, Pabla NS. Zinc finger protein 24-dependent transcription factor SOX9 up-regulation protects tubular epithelial cells during acute kidney injury. Kidney Int 2023; 103:1093-1104. [PMID: 36921719 PMCID: PMC10200760 DOI: 10.1016/j.kint.2023.02.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
Transcriptional profiling studies have identified several protective genes upregulated in tubular epithelial cells during acute kidney injury (AKI). Identifying upstream transcriptional regulators could lead to the development of therapeutic strategies augmenting the repair processes. SOX9 is a transcription factor controlling cell-fate during embryonic development and adult tissue homeostasis in multiple organs including the kidneys. SOX9 expression is low in adult kidneys; however, stress conditions can trigger its transcriptional upregulation in tubular epithelial cells. SOX9 plays a protective role during the early phase of AKI and facilitates repair during the recovery phase. To identify the upstream transcriptional regulators that drive SOX9 upregulation in tubular epithelial cells, we used an unbiased transcription factor screening approach. Preliminary screening and validation studies show that zinc finger protein 24 (ZFP24) governs SOX9 upregulation in tubular epithelial cells. ZFP24, a Cys2-His2 (C2H2) zinc finger protein, is essential for oligodendrocyte maturation and myelination; however, its role in the kidneys or in SOX9 regulation remains unknown. Here, we found that tubular epithelial ZFP24 gene ablation exacerbated ischemia, rhabdomyolysis, and cisplatin-associated AKI. Importantly, ZFP24 gene deletion resulted in suppression of SOX9 upregulation in injured tubular epithelial cells. Chromatin immunoprecipitation and promoter luciferase assays confirmed that ZFP24 bound to a specific site in both murine and human SOX9 promoters. Importantly, CRISPR/Cas9-mediated mutation in the ZFP24 binding site in the SOX9 promoter in vivo led to suppression of SOX9 upregulation during AKI. Thus, our findings identify ZFP24 as a critical stress-responsive transcription factor protecting tubular epithelial cells through SOX9 upregulation.
Collapse
Affiliation(s)
- Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Division of Nephrology and Hypertension, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bijay Bisunke
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Alanys V Luna Ramirez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Laura A Jayne
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Mei Ji He Ho Feng
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Bhavya Girotra
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Shirely M Acosta Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Corynne R Vermillion
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Isaac Z Karel
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Nicholas Ferrell
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sangwoon Chung
- Pulmonary, Sleep and Critical Care Medicine, Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - John W Christman
- Pulmonary, Sleep and Critical Care Medicine, Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - Craig R Brooks
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sethu M Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | - Anjali A Satoskar
- Division of Renal and Transplant Pathology, Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Division of Nephrology and Hypertension, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Amandeep Bajwa
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Navjot Singh Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
9
|
Han Z, Chen L, Peng H, Zheng H, Lin Y, Peng F, Fan Y, Xie X, Yang S, Wang Z, Yuan L, Wei X, Chen H. The role of thyroid hormone in the renal immune microenvironment. Int Immunopharmacol 2023; 119:110172. [PMID: 37086678 DOI: 10.1016/j.intimp.2023.110172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/24/2023]
Abstract
Thyroid hormones are essential for proper kidney growth and development. The kidney is not only the organ of thyroid hormone metabolism but also the target organ of thyroid hormone. Kidney disease is a common type of kidney damage, mainly including different types of acute kidney injury, chronic kidney disease, diabetic nephropathy, lupus nephritis, and renal cell carcinoma. The kidney is often damaged by an immune response directed against its antigens or a systemic immune response. A variety of immune cells in the innate and adaptive immune systems, including neutrophils, macrophages, dendritic cells, T lymphocytes, and B lymphocytes, is essential for maintaining immune homeostasis and preventing autoimmune kidney disease. Recent studies have found that thyroid hormone plays an indispensable role in the immune microenvironment of various kidney diseases. Thyroid hormones regulate the activity of neutrophils, and dendritic cells express triiodothyronine receptors. Compared to hypothyroidism, hyperthyroidism has a greater effect on neutrophils. Furthermore, in adaptive immune systems, thyroid hormone may activate T lymphocytes through several underlying mechanisms, such as mediating NF-κB, protein kinase C signalling pathways, and β-adrenergic receptors, leading to increased T lymphocyte activation. The present review discusses the effects of thyroid hormone metabolism regulation in the immune microenvironment on the function of various immune cells, especially neutrophils, macrophages, dendritic cells, T lymphocytes, and B lymphocytes. Although there are not enough data at this stage to conclude the clinical relevance of these findings, thyroid hormone metabolism may influence autoimmune kidney disease by regulating the renal immune microenvironment.
Collapse
Affiliation(s)
- Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liuyan Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyao Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongying Zheng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunhe Fan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuli Xie
- School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Simin Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhanzhan Wang
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China
| | - Lan Yuan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiuyan Wei
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | | |
Collapse
|
10
|
Liu M, Zeng M, Guo P, Zhang Y, Yang X, Jia J, Zhang Q, Zhang B, Cao B, Wang R, Zheng X, Feng W. Effects of three types of fresh Rehmannia glutinosa improve lipopolysaccharide-induced acute kidney injury in sepsis through the estrogen receptor pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:579-586. [PMID: 37051100 PMCID: PMC10083830 DOI: 10.22038/ijbms.2023.67322.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/14/2023] [Indexed: 04/14/2023]
Abstract
Objectives To explore the effects and mechanism of three types of fresh Rehmannia glutinosa, namely Beijing No. 3 (BJ3H), Huaizhong No. 1 (HZ1H), and Taisheng (TS) on lipopolysaccharide (LPS)-induced acute kidney injury in the sepsis (S-AKI) mice model through the estrogen receptor pathway. Materials and Methods BALB/c mice were randomly divided into control (CON), model (LPS), astragalus injection (ASI), BJ3H, HZ1H, TS water extract groups, the estrogen receptor antagonist ICI182,780 groups were added to each group. The antagonist groups received an intraperitoneal injection of ICI 0.5 hr before administration and an intraperitoneal injection of LPS 3 days after administration. The kidney pathology, function, inflammatory factors, immune cells, levels of reactive oxygen species (ROS), apoptosis, and the protein expression levels of TLR4/NF-κB/NLRP3 signaling pathway in the mice kidneys were detected. Results ASI, BJ3H, HZ1H, and TS improved LPS-induced renal pathology in S-AKI mice, reduced the kidney and serum levels of inflammatory factors, positive rates of macrophages and neutrophils, levels of ROS and apoptosis, and the relative expression levels of TLR4, MyD88, NF-κB p-p65/NF-κB p65, and NLRP3 proteins in the kidney. In addition, they increased the positive rate of dendritic cells (DCs) in the mice kidneys. The overall effect of HZ1H was superior to that of ASI, BJ3H, and TS. However, after adding ICI, the regulatory effects of drugs were inhibited. Conclusion The three types of fresh R. glutinosa may completely or partially affect the TLR4/NF-κB/NLRP3 signaling pathway through the estrogen receptor pathway to exert a protective effect on S-AKI.
Collapse
Affiliation(s)
- Meng Liu
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Mengnan Zeng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Pengli Guo
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Yuhan Zhang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xiaofeng Yang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jufang Jia
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Qinqin Zhang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Beibei Zhang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Bing Cao
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Ru Wang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xiaoke Zheng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Corresponding authors: Xiaoke Zheng. Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China. ; Weisheng Feng. Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Weisheng Feng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Corresponding authors: Xiaoke Zheng. Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China. ; Weisheng Feng. Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
11
|
Agrawal M, Rasiah PK, Bajwa A, Rajasingh J, Gangaraju R. Mesenchymal Stem Cell Induced Foxp3(+) Tregs Suppress Effector T Cells and Protect against Retinal Ischemic Injury. Cells 2021; 10:3006. [PMID: 34831229 PMCID: PMC8616393 DOI: 10.3390/cells10113006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 12/02/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are well known for immunomodulation; however, the mechanisms involved in their benefits in the ischemic retina are unknown. This study tested the hypothesis that MSC induces upregulation of transcription factor forkhead box protein P3 (Foxp3) in T cells to elicit immune modulation, and thus, protect against retinal damage. Induced MSCs (iMSCs) were generated by differentiating the induced pluripotent stem cells (iPSC) derived from urinary epithelial cells through a noninsertional reprogramming approach. In in-vitro cultures, iMSC transferred mitochondria to immune cells via F-actin nanotubes significantly increased oxygen consumption rate (OCR) for basal respiration and ATP production, suppressed effector T cells, and promoted differentiation of CD4+CD25+ T regulatory cells (Tregs) in coculture with mouse splenocytes. In in-vivo studies, iMSCs transplanted in ischemia-reperfusion (I/R) injured eye significantly increased Foxp3+ Tregs in the retina compared to that of saline-injected I/R eyes. Furthermore, iMSC injected I/R eyes significantly decreased retinal inflammation as evidenced by reduced gene expression of IL1β, VCAM1, LAMA5, and CCL2 and improved b-wave amplitudes compared to that of saline-injected I/R eyes. Our study demonstrates that iMSCs can transfer mitochondria to immune cells to suppress the effector T cell population. Additionally, our current data indicate that iMSC can enhance differentiation of T cells into Foxp3 Tregs in vitro and therapeutically improve the retina's immune function by upregulation of Tregs to decrease inflammation and reduce I/R injury-induced retinal degeneration in vivo.
Collapse
Affiliation(s)
- Mona Agrawal
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (M.A.); (P.K.R.)
| | - Pratheepa Kumari Rasiah
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (M.A.); (P.K.R.)
| | - Amandeep Bajwa
- James D. Eason Transplant Institute, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Johnson Rajasingh
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (M.A.); (P.K.R.)
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
12
|
Bai Y, Kim JY, Bisunke B, Jayne LA, Silvaroli JA, Balzer MS, Gandhi M, Huang KM, Sander V, Prosek J, Cianciolo RE, Baker SD, Sparreboom A, Jhaveri KD, Susztak K, Bajwa A, Pabla NS. Kidney toxicity of the BRAF-kinase inhibitor vemurafenib is driven by off-target ferrochelatase inhibition. Kidney Int 2021; 100:1214-1226. [PMID: 34534550 DOI: 10.1016/j.kint.2021.08.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 07/21/2021] [Accepted: 08/13/2021] [Indexed: 12/29/2022]
Abstract
A multitude of disease and therapy related factors drive the frequent development of kidney disorders in cancer patients. Along with chemotherapy, the newer targeted therapeutics can also cause kidney dysfunction through on and off-target mechanisms. Interestingly, among the small molecule inhibitors approved for the treatment of cancers that harbor BRAF-kinase activating mutations, vemurafenib can trigger tubular damage and acute kidney injury. BRAF is a proto-oncogene involved in cell growth. To investigate the underlying mechanisms, we developed cell culture and mouse models of vemurafenib kidney toxicity. At clinically relevant concentrations vemurafenib induces cell-death in transformed and primary mouse and human kidney tubular epithelial cells. In mice, two weeks of daily vemurafenib treatment causes moderate acute kidney injury with histopathological characteristics of kidney tubular epithelial cells injury. Importantly, kidney tubular epithelial cell-specific BRAF gene deletion did not influence kidney function under normal conditions or alter the severity of vemurafenib-associated kidney impairment. Instead, we found that inhibition of ferrochelatase, an enzyme involved in heme biosynthesis contributes to vemurafenib kidney toxicity. Ferrochelatase overexpression protected kidney tubular epithelial cells and conversely ferrochelatase knockdown increased the sensitivity to vemurafenib-induced kidney toxicity. Thus, our studies suggest that vemurafenib-associated kidney tubular epithelial cell dysfunction and kidney toxicity is BRAF-independent and caused, in part, by off-target ferrochelatase inhibition.
Collapse
Affiliation(s)
- Yuntao Bai
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Bijay Bisunke
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Laura A Jayne
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Michael S Balzer
- Department of Medicine and Genetics, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Megha Gandhi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jason Prosek
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Rachel E Cianciolo
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Northwell Health, Great Neck, New York, USA
| | - Katalin Susztak
- Department of Medicine and Genetics, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amandeep Bajwa
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Navjot Singh Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|