1
|
Gao A, Lin Y, Chai Y, Han J, Wu L, Ye J. CXCL12/CXCR4 Axis Promotes the Chemotaxis and Phagocytosis of B Cells through the PI3K-AKT Signaling Pathway in an Early Vertebrate. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1676-1690. [PMID: 39441360 DOI: 10.4049/jimmunol.2300562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Chemokines play crucial roles in the regulation of immune cell migration and development. The CXCL12/CXCR4 axis has been extensively studied in mammals, but its regulatory mechanism in teleost fish remains unclear. In this study, we used Nile tilapia (Oreochromis niloticus) as a teleost model to investigate the mediation of the CXCL12/CXCR4 axis in IgM+ B cells. Our findings demonstrate that the CXCL12/CXCR4 axis exhibits chemotactic activity on IgM+ B cells and promotes the phagocytosis of IgM+ B cells. Blocking CXCR4 severely impairs the chemotaxis and phagocytosis of IgM+ B cells in vitro and reduces the percentages and numbers of IgM+ B cells that migrate to peripheral blood after pathogen infection in vivo. This reduction in migration leads to a decrease in the inflammatory response, an increase in tissue bacterial load, and a decrease in survival rate. We also discovered that the evolutionarily conserved PI3K-AKT signaling pathway and Girdin are involved in the immune response during Streptococcus agalactiae infection. Inhibitors of the PI3K-AKT signaling pathway prevent the chemotaxis and phagocytosis of IgM+ B cells, impair the expression and phosphorylation levels of related proteins in vitro, and prevent IgM+ B cells chemotaxis into the peripheral blood after pathogen infection in vivo. Furthermore, CXCR4 blocking significantly downregulates the expression of AKT and Girdin. Overall, our study reveals the regulatory mechanism of the CXCL12/CXCR4 axis on IgM+ B cells via the PI3K-AKT signaling pathway in tilapia, suggesting that the functions of the CXCL12/CXCR4 axis in B cells may be conserved between mammals and teleost fish.
Collapse
Affiliation(s)
- Along Gao
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yuhua Lin
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yiwen Chai
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jugan Han
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Liting Wu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jianmin Ye
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
2
|
Khbouz B, Musumeci L, Grahammer F, Jouret F. The Dual-specificity Phosphatase 3 (DUSP3): A Potential Target Against Renal Ischemia/Reperfusion Injury. Transplantation 2024; 108:2166-2173. [PMID: 39466786 DOI: 10.1097/tp.0000000000005009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Renal ischemia/reperfusion (I/R) injury is a common clinical challenge faced by clinicians in kidney transplantation. I/R is the leading cause of acute kidney injury, and it occurs when blood flow to the kidney is interrupted and subsequently restored. I/R impairs renal function in both short and long terms. Renal ischemic preconditioning refers to all maneuvers intended to prevent or attenuate ischemic damage. In this context, the present review focuses on the dual-specificity phosphatase 3 (DUSP3), also known as vaccinia H1-related phosphatase, an uncommon regulator of mitogen-activated protein kinase (MAPK) phosphorylation. DUSP3 has different biological functions: (1) it acts as a tumor modulator and (2) it is involved in the regulation of immune response, thrombosis, hemostasis, angiogenesis, and genomic stability. These functions occur either through MAPK-dependent or MAPK-independent mechanisms. DUSP3 genetic deletion dampens kidney damage and inflammation caused by I/R in mice, suggesting DUSP3 as a potential target for preventing renal I/R injury. Here, we discuss the putative role of DUSP3 in ischemic preconditioning and the potential mechanisms of such an attenuated inflammatory response via improved kidney perfusion and adequate innate immune response.
Collapse
Affiliation(s)
- Badr Khbouz
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Liège, Belgium
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine (Nephrology, Rheumatology, Endocrinology), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lucia Musumeci
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Liège, Belgium
- Department of Cardiovascular Surgery, CHU of Liège, Liège, Belgium
| | - Florian Grahammer
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine (Nephrology, Rheumatology, Endocrinology), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Liège, Belgium
- Division of Nephrology, CHU of Liège, University of Liège (CHU ULiège), Liège, Belgium
| |
Collapse
|
3
|
Asadi R, Shadpour P, Nakhaei A. Non-dialyzable uremic toxins and renal tubular cell damage in CKD patients: a systems biology approach. Eur J Med Res 2024; 29:412. [PMID: 39123228 PMCID: PMC11311939 DOI: 10.1186/s40001-024-01951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Chronic kidney disease presents global health challenges, with hemodialysis as a common treatment. However, non-dialyzable uremic toxins demand further investigation for new therapeutic approaches. Renal tubular cells require scrutiny due to their vulnerability to uremic toxins. METHODS In this study, a systems biology approach utilized transcriptomics data from healthy renal tubular cells exposed to healthy and post-dialysis uremic plasma. RESULTS Differential gene expression analysis identified 983 up-regulated genes, including 70 essential proteins in the protein-protein interaction network. Modularity-based clustering revealed six clusters of essential proteins associated with 11 pathological pathways activated in response to non-dialyzable uremic toxins. CONCLUSIONS Notably, WNT1/11, AGT, FGF4/17/22, LMX1B, GATA4, and CXCL12 emerged as promising targets for further exploration in renal tubular pathology related to non-dialyzable uremic toxins. Understanding the molecular players and pathways linked to renal tubular dysfunction opens avenues for novel therapeutic interventions and improved clinical management of chronic kidney disease and its complications.
Collapse
Affiliation(s)
- Roya Asadi
- Industrial Engineering Department, Faculty of Technical and Engineering, University of Science and Culture (USC), Tehran, Iran
| | - Pejman Shadpour
- Hospital Management Research Center (HMRC), Hasheminejad Kidney Center (HKC), Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Akram Nakhaei
- Computer Engineering Department, Mazandaran University of Science and Technology (MUST), Babol, Iran.
| |
Collapse
|
4
|
Bork T, Hernando-Erhard C, Liang W, Tian Z, Yamahara K, Huber TB. Cisplatin Nephrotoxicity Is Critically Mediated by the Availability of BECLIN1. Int J Mol Sci 2024; 25:2560. [PMID: 38473806 DOI: 10.3390/ijms25052560] [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: 01/18/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Cisplatin nephrotoxicity is a critical limitation of solid cancer treatment. Until now, the complex interplay of various pathophysiological mechanisms leading to proximal tubular cell apoptosis after cisplatin exposure has not been fully understood. In our study, we assessed the role of the autophagy-related protein BECLIN1 (ATG6) in cisplatin-induced acute renal injury (AKI)-a candidate protein involved in autophagy and with putative impact on apoptosis by harboring a B-cell lymphoma 2 (BCL2) interaction site of unknown significance. By using mice with heterozygous deletion of Becn1, we demonstrate that reduced intracellular content of BECLIN1 does not impact renal function or autophagy within 12 months. However, these mice were significantly sensitized towards cisplatin-induced AKI, and by using Becn1+/-;Sglt2-Cre;Tomato/EGFP mice with subsequent primary cell analysis, we confirmed that nephrotoxicity depends on proximal tubular BECLIN1 content. Mechanistically, BECLIN1 did not impact autophagy or primarily the apoptotic pathway. In fact, a lack of BECLIN1 sensitized mice towards cisplatin-induced ER stress. Accordingly, the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) blunted cisplatin-induced cell death in Becn1 heterozygosity. In conclusion, our data first highlight a novel role of BECLIN1 in protecting against cellular ER stress independent from autophagy. These novel findings open new therapeutic avenues to intervene in this important intracellular stress response pathway with a promising impact on future AKI management.
Collapse
Affiliation(s)
- Tillmann Bork
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Camila Hernando-Erhard
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Wei Liang
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhejia Tian
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany
| | - Kosuke Yamahara
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu 520-2192, Shiga, Japan
| | - Tobias B Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
5
|
Tang W, Wei Q. The metabolic pathway regulation in kidney injury and repair. Front Physiol 2024; 14:1344271. [PMID: 38283280 PMCID: PMC10811252 DOI: 10.3389/fphys.2023.1344271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Kidney injury and repair are accompanied by significant disruptions in metabolic pathways, leading to renal cell dysfunction and further contributing to the progression of renal pathology. This review outlines the complex involvement of various energy production pathways in glucose, lipid, amino acid, and ketone body metabolism within the kidney. We provide a comprehensive summary of the aberrant regulation of these metabolic pathways in kidney injury and repair. After acute kidney injury (AKI), there is notable mitochondrial damage and oxygen/nutrient deprivation, leading to reduced activity in glycolysis and mitochondrial bioenergetics. Additionally, disruptions occur in the pentose phosphate pathway (PPP), amino acid metabolism, and the supply of ketone bodies. The subsequent kidney repair phase is characterized by a metabolic shift toward glycolysis, along with decreased fatty acid β-oxidation and continued disturbances in amino acid metabolism. Furthermore, the impact of metabolism dysfunction on renal cell injury, regeneration, and the development of renal fibrosis is analyzed. Finally, we discuss the potential therapeutic strategies by targeting renal metabolic regulation to ameliorate kidney injury and fibrosis and promote kidney repair.
Collapse
Affiliation(s)
- Wenbin Tang
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
6
|
Liu Y, Liu N, He P, Cao S, Li H, Liu D. Arginine-methylated c-Myc affects mitochondrial mitophagy in mouse acute kidney injury via Slc25a24. J Cell Physiol 2024; 239:193-211. [PMID: 38164038 DOI: 10.1002/jcp.31160] [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: 07/12/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 01/03/2024]
Abstract
The transcription factor methylated c-Myc heterodimerizes with MAX to modulate gene expression, and plays an important role in energy metabolism in kidney injury but the exact mechanism remains unclear. Mitochondrial solute transporter Slc25a24 imports ATP into mitochondria and is central to energy metabolism. Gene Expression Omnibus data analysis reveals Slc25a24 and c-Myc are consistently upregulated in all the acute kidney injury (AKI) cells. Pearson correlation analysis also shows that Slc25a24 and c-Myc are strongly correlated (⍴ > 0.9). Mutant arginine methylated c-Myc (R299A and R346A) reduced its combination with MAX when compared with the wild type of c-Myc. On the other hand, the Slc25a24 levels were also correspondingly reduced, which induced the downregulation of ATP production. The results promoted reactive oxygen species (ROS) production and mitophagy generation. The study revealed that the c-Myc overexpression manifested the most pronounced mitochondrial DNA depletion. Additionally, the varied levels of mitochondrial proteins like TIM23, TOM20, and PINK1 in each group, particularly the elevated levels of PINK1 in AKI model groups and lower levels of TIM23 and TOM20 in the c-Myc overexpression group, suggest potential disruptions in mitochondrial dynamics and homeostasis, indicating enhanced mitophagy or mitochondrial loss. Therefore, arginine-methylated c-Myc affects mouse kidney injury by regulating mitochondrial ATP and ROS, and mitophagy via Slc25a24.
Collapse
Affiliation(s)
- Ying Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Naiquan Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping He
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shiyu Cao
- Grade 2018 Clinical Medicine, China Medical University, Shenyang, China
| | - Huabing Li
- Department of Nephrology, Tiemei General Hospital of Liaoning Province Health Industrial Group, Tieling, China
| | - Dajun Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Yang M, Lopez LN, Brewer M, Delgado R, Menshikh A, Clouthier K, Zhu Y, Vanichapol T, Yang H, Harris RC, Gewin L, Brooks CR, Davidson AJ, de Caestecker M. Inhibition of retinoic acid signaling in proximal tubular epithelial cells protects against acute kidney injury. JCI Insight 2023; 8:e173144. [PMID: 37698919 PMCID: PMC10619506 DOI: 10.1172/jci.insight.173144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Retinoic acid receptor (RAR) signaling is essential for mammalian kidney development but, in the adult kidney, is restricted to occasional collecting duct epithelial cells. We now show that there is widespread reactivation of RAR signaling in proximal tubular epithelial cells (PTECs) in human sepsis-associated acute kidney injury (AKI) and in mouse models of AKI. Genetic inhibition of RAR signaling in PTECs protected against experimental AKI but was unexpectedly associated with increased expression of the PTEC injury marker Kim1. However, the protective effects of inhibiting PTEC RAR signaling were associated with increased Kim1-dependent apoptotic cell clearance, or efferocytosis, and this was associated with dedifferentiation, proliferation, and metabolic reprogramming of PTECs. These data demonstrate the functional role that reactivation of RAR signaling plays in regulating PTEC differentiation and function in human and experimental AKI.
Collapse
Affiliation(s)
- Min Yang
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lauren N. Lopez
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maya Brewer
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rachel Delgado
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anna Menshikh
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelly Clouthier
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yuantee Zhu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thitinee Vanichapol
- Department of Molecular Medicine & Pathology, The University of Auckland, Auckland, New Zealand
| | - Haichun Yang
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Raymond C. Harris
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Leslie Gewin
- Washington University in St. Louis School of Medicine and the St. Louis Veterans Affairs Hospital, St. Louis, Missouri, USA
| | - Craig R. Brooks
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alan J. Davidson
- Department of Molecular Medicine & Pathology, The University of Auckland, Auckland, New Zealand
| | - Mark de Caestecker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
8
|
Yang M, Lopez LN, Brewer M, Delgado R, Menshikh A, Clouthier K, Zhu Y, Vanichapol T, Yang H, Harris R, Gewin L, Brooks C, Davidson A, de Caestecker MP. Inhibition of Retinoic Acid Signaling in Proximal Tubular Epithelial cells Protects against Acute Kidney Injury by Enhancing Kim-1-dependent Efferocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545113. [PMID: 37398101 PMCID: PMC10312711 DOI: 10.1101/2023.06.15.545113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Retinoic acid receptor (RAR) signaling is essential for mammalian kidney development, but in the adult kidney is restricted to occasional collecting duct epithelial cells. We now show there is widespread reactivation of RAR signaling in proximal tubular epithelial cells (PTECs) in human sepsis-associated acute kidney injury (AKI), and in mouse models of AKI. Genetic inhibition of RAR signaling in PTECs protects against experimental AKI but is associated with increased expression of the PTEC injury marker, Kim-1. However, Kim-1 is also expressed by de-differentiated, proliferating PTECs, and protects against injury by increasing apoptotic cell clearance, or efferocytosis. We show that the protective effect of inhibiting PTEC RAR signaling is mediated by increased Kim-1 dependent efferocytosis, and that this is associated with de-differentiation, proliferation, and metabolic reprogramming of PTECs. These data demonstrate a novel functional role that reactivation of RAR signaling plays in regulating PTEC differentiation and function in human and experimental AKI. Graphical abstract
Collapse
|
9
|
Tang Z, Wang K, Lo L, Chen J. Tg(Δ113p53:cmyc) Transgene Upregulates glut1 Expression to Promote Zebrafish Heart Regeneration. J Cardiovasc Dev Dis 2023; 10:246. [PMID: 37367411 DOI: 10.3390/jcdd10060246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
The heart switches its main metabolic substrate from glucose to fatty acids shortly after birth, which is one of reasons for the loss of heart regeneration capability in adult mammals. On the contrary, metabolic shifts from oxidative phosphorylation to glucose metabolism promote cardiomyocyte (CM) proliferation after heart injury. However, how glucose transportation in CMs is regulated during heart regeneration is still not fully understood. In this report, we found that the expression of Glut1 (slc2a1) was upregulated around the injury site of zebrafish heart, accompanied by an increase in glucose uptake at the injury area. Knockout of slc2a1a impaired zebrafish heart regeneration. Our previous study has demonstrated that the expression of Δ113p53 is activated after heart injury and Δ113p53+ CMs undergo proliferation to contribute to zebrafish heart regeneration. Next, we used the Δ113p53 promoter to generate the Tg(Δ113p53:cmyc) zebrafish transgenic line. Conditional overexpression of cmyc not only significantly promoted zebrafish CM proliferation and heart regeneration but also significantly enhanced glut1 expression at the injury site. Inhibiting Glut1 diminished the increase in CM proliferation in Tg(Δ113p53:cmyc) injured hearts of zebrafish. Therefore, our results suggest that the activation of cmyc promotes heart regeneration through upregulating the expression of glut1 to speed up glucose transportation.
Collapse
Affiliation(s)
- Zimu Tang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Kaiyuan Wang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Lijian Lo
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Chen
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Qiu S, Cai Y, Yao H, Lin C, Xie Y, Tang S, Zhang A. Small molecule metabolites: discovery of biomarkers and therapeutic targets. Signal Transduct Target Ther 2023; 8:132. [PMID: 36941259 PMCID: PMC10026263 DOI: 10.1038/s41392-023-01399-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/22/2023] Open
Abstract
Metabolic abnormalities lead to the dysfunction of metabolic pathways and metabolite accumulation or deficiency which is well-recognized hallmarks of diseases. Metabolite signatures that have close proximity to subject's phenotypic informative dimension, are useful for predicting diagnosis and prognosis of diseases as well as monitoring treatments. The lack of early biomarkers could lead to poor diagnosis and serious outcomes. Therefore, noninvasive diagnosis and monitoring methods with high specificity and selectivity are desperately needed. Small molecule metabolites-based metabolomics has become a specialized tool for metabolic biomarker and pathway analysis, for revealing possible mechanisms of human various diseases and deciphering therapeutic potentials. It could help identify functional biomarkers related to phenotypic variation and delineate biochemical pathways changes as early indicators of pathological dysfunction and damage prior to disease development. Recently, scientists have established a large number of metabolic profiles to reveal the underlying mechanisms and metabolic networks for therapeutic target exploration in biomedicine. This review summarized the metabolic analysis on the potential value of small-molecule candidate metabolites as biomarkers with clinical events, which may lead to better diagnosis, prognosis, drug screening and treatment. We also discuss challenges that need to be addressed to fuel the next wave of breakthroughs.
Collapse
Affiliation(s)
- Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), College of Chinese Medicine, Hainan Medical University, Xueyuan Road 3, Haikou, 571199, China
| | - Ying Cai
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Hong Yao
- First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Chunsheng Lin
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150001, China
| | - Yiqiang Xie
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), College of Chinese Medicine, Hainan Medical University, Xueyuan Road 3, Haikou, 571199, China.
| | - Songqi Tang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), College of Chinese Medicine, Hainan Medical University, Xueyuan Road 3, Haikou, 571199, China.
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), College of Chinese Medicine, Hainan Medical University, Xueyuan Road 3, Haikou, 571199, China.
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
11
|
Drummond BE, Ercanbrack WS, Wingert RA. Modeling Podocyte Ontogeny and Podocytopathies with the Zebrafish. J Dev Biol 2023; 11:9. [PMID: 36810461 PMCID: PMC9944608 DOI: 10.3390/jdb11010009] [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: 01/09/2023] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Podocytes are exquisitely fashioned kidney cells that serve an essential role in the process of blood filtration. Congenital malformation or damage to podocytes has dire consequences and initiates a cascade of pathological changes leading to renal disease states known as podocytopathies. In addition, animal models have been integral to discovering the molecular pathways that direct the development of podocytes. In this review, we explore how researchers have used the zebrafish to illuminate new insights about the processes of podocyte ontogeny, model podocytopathies, and create opportunities to discover future therapies.
Collapse
Affiliation(s)
| | | | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
12
|
Transgenic IDH2 R172K and IDH2 R140Q zebrafish models recapitulated features of human acute myeloid leukemia. Oncogene 2023; 42:1272-1281. [PMID: 36739363 PMCID: PMC10101851 DOI: 10.1038/s41388-023-02611-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/06/2023]
Abstract
Isocitrate dehydrogenase 2 (IDH2) mutations occur in more than 15% of cytogenetically normal acute myeloid leukemia (CN-AML) but comparative studies of their roles in leukemogenesis have been scarce. We generated zebrafish models of IDH2R172K and IDH2R140Q AML and reported their pathologic, functional and transcriptomic features and therapeutic responses to target therapies. Transgenic embryos co-expressing FLT3ITD and IDH2 mutations showed accentuation of myelopoiesis. As these embryos were raised to adulthood, full-blown leukemia ensued with multi-lineage dysplasia, increase in myeloblasts and marrow cellularity and splenomegaly. The leukemia cells were transplantable into primary and secondary recipients and resulted in more aggressive disease. Tg(Runx1:FLT3ITDIDH2R172K) but not Tg(Runx1:FLT3ITDIDH2R140Q) zebrafish showed an increase in T-cell development at embryonic and adult stages. Single-cell transcriptomic analysis revealed increased myeloid skewing, differentiation blockade and enrichment of leukemia-associated gene signatures in both zebrafish models. Tg(Runx1:FLT3ITDIDH2R172K) but not Tg(Runx1:FLT3ITDIDH2R140Q) zebrafish showed an increase in interferon signals at the adult stage. Leukemic phenotypes in both zebrafish could be ameliorated by quizartinib and enasidenib. In conclusion, the zebrafish models of IDH2 mutated AML recapitulated the morphologic, clinical, functional and transcriptomic characteristics of human diseases, and provided the prototype for developing zebrafish leukemia models of other genotypes that would become a platform for high throughput drug screening.
Collapse
|
13
|
Radiotherapy Advances in Renal Disease-Focus on Renal Ischemic Preconditioning. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010068. [PMID: 36671640 PMCID: PMC9855155 DOI: 10.3390/bioengineering10010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Ionizing irradiation is widely applied as a fundamental therapeutic treatment in several diseases. Acute kidney injury (AKI) represents a global public health problem with major morbidity and mortality. Renal ischemia/reperfusion (I/R) is the main cause of AKI. I/R injury occurs when blood flow to the kidney is transiently interrupted and then restored. Such an ischemic insult significantly impairs renal function in the short and long terms. Renal ischemic preconditioning (IPC) corresponds to the maneuvers intended to prevent or attenuate the ischemic damage. In murine models, irradiation-induced preconditioning (IP) renders the renal parenchyma resistant to subsequent damage by activating defense pathways involved in oxidative stress, angiogenesis, and inflammation. Before envisioning translational applications in patients, safe irradiation modalities, including timing, dosage, and fractionation, need to be defined.
Collapse
|
14
|
Zhao J, Liu H, Xia M, Chen Q, Wan L, Leng B, Tang C, Chen G, Liu Y, Zhang L, Liu H. Network Pharmacology and Experimental Validation to Explore That Celastrol Targeting PTEN is the Potential Mechanism of Tripterygium wilfordii (Lév.) Hutch Against IgA Nephropathy. Drug Des Devel Ther 2023; 17:887-900. [PMID: 36992900 PMCID: PMC10042171 DOI: 10.2147/dddt.s402503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Purpose Accumulating clinical evidence showed that Tripterygium hypoglaucum (Lév.) Hutch (THH) is effective against IgA nephropathy (IgAN), but the mechanism is still unclear. This study is to evaluate the renal protective effect and molecular mechanism of THH against IgAN via network pharmacology, molecular docking strategy and experimental validation. Methods Several databases were used for obtaining the active ingredients of THH, the corresponding targets, as well as the IgAN-related genes. The critical active ingredients, functional pathways, and potential for the combination of the hub genes and their corresponding active components were determined through bioinformatics analysis and molecular docking. The IgAN mouse model was treated with celastrol (1 mg/kg/d) for 21 days, and the aggregated IgA1-induced human mesangial cell (HMC) was treated with various concentrations of celastrol (25, 50 or 75 nM) for 48 h. The immunohistochemistry and Western blot techniques were applied to evaluate the protein expression of the predicted target. The cell counting kit 8 (CCK8) was used to detect HMC proliferation. Results A total of 17 active ingredients from THH were screened, covering 165 IgAN-related targets. The PPI network identified ten hub targets, including PTEN. The binding affinity between the celastrol and PTEN was the highest (-8.69 kJ/mol). The immunohistochemistry showed that celastrol promoted the expression of PTEN in the glomerulus of IgAN mice. Furthermore, the Western blot techniques showed that celastrol significantly elevated the expression of PTEN and inhibited PCNA and Cyclin D1 in vitro and in vivo. The CCK8 assay determined that celastrol decreased HMC proliferation in a concentration-dependent manner. Conclusion This study suggests that activating PTEN by celastrol may play a pivotal role in THH alleviating IgAN renal injury.
Collapse
Affiliation(s)
- Juanyong Zhao
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Haiyang Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Ming Xia
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qian Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Lili Wan
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Bin Leng
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Chengyuan Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Guochun Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yu Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Lei Zhang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Hong Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Correspondence: Hong Liu; Lei Zhang, Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China, Tel +86-13973116951; +86-18673174522, Email ;
| |
Collapse
|
15
|
DeVallance ER, Dustin CM, de Jesus DS, Ghouleh IA, Sembrat JC, Cifuentes-Pagano E, Pagano PJ. Specificity Protein 1-Mediated Promotion of CXCL12 Advances Endothelial Cell Metabolism and Proliferation in Pulmonary Hypertension. Antioxidants (Basel) 2022; 12:71. [PMID: 36670936 PMCID: PMC9854820 DOI: 10.3390/antiox12010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare yet devastating and incurable disease with few treatment options. The underlying mechanisms of PAH appear to involve substantial cellular proliferation and vascular remodeling, causing right ventricular overload and eventual heart failure. Recent evidence suggests a significant seminal role of the pulmonary endothelium in the initiation and promotion of PAH. Our previous work identified elevated reactive oxygen species (ROS)-producing enzyme NADPH oxidase 1 (NOX1) in human pulmonary artery endothelial cells (HPAECs) of PAH patients promoting endothelial cell proliferation in vitro. In this study, we interrogated chemokine CXCL12's (aka SDF-1) role in EC proliferation under the control of NOX1 and specificity protein 1 (Sp1). We report here that NOX1 can drive hypoxia-induced endothelial CXCL12 expression via the transcription factor Sp1 leading to HPAEC proliferation and migration. Indeed, NOX1 drove hypoxia-induced Sp1 activation, along with an increased capacity of Sp1 to bind cognate promoter regions in the CXCL12 promoter. Sp1 activation induced elevated expression of CXCL12 in hypoxic HPAECs, supporting downstream induction of expression at the CXCL12 promoter via NOX1 activity. Pathological levels of CXCL12 mimicking those reported in human PAH patient serum restored EC proliferation impeded by specific NOX1 inhibitor. The translational relevance of our findings is highlighted by elevated NOX1 activity, Sp1 activation, and CXCL12 expression in explanted lung samples from PAH patients compared to non-PAH controls. Analysis of phosphofructokinase, glucose-6-phosphate dehydrogenase, and glutaminase activity revealed that CXCL12 induces glutamine and glucose metabolism, which are foundational to EC cell proliferation. Indeed, in explanted human PAH lungs, demonstrably higher glutaminase activity was detected compared to healthy controls. Finally, infusion of recombinant CXCL12 into healthy mice amplified pulmonary arterial pressure, right ventricle remodeling, and elevated glucose and glutamine metabolism. Together these data suggest a central role for a novel NOX1-Sp1-CXCL12 pathway in mediating PAH phenotype in the lung endothelium.
Collapse
Affiliation(s)
- Evan R. DeVallance
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Christopher M. Dustin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel Simoes de Jesus
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Imad Al Ghouleh
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Cardiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John C. Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Eugenia Cifuentes-Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Patrick J. Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
16
|
Zhang Y, Zhang M, Xie Z, Ding Y, Huang J, Yao J, Lv Y, Zuo J. Research Progress and Direction of Novel Organelle-Migrasomes. Cancers (Basel) 2022; 15:cancers15010134. [PMID: 36612129 PMCID: PMC9817827 DOI: 10.3390/cancers15010134] [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: 11/07/2022] [Revised: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Migrasomes are organelles that are similar in structure to pomegranates, up to 3 μm in diameter, and contain small vesicles with a diameter of 50-100 nm. These membranous organelles grow at the intersections or tips of retracting fibers at the back of migrating cells. The process by which cells release migrasomes and their contents outside the cell is called migracytosis. The signal molecules are packaged in the migrasomes and released to the designated location by migrasomes to activate the surrounding cells. Finally, the migrasomes complete the entire process of information transmission. In this sense, migrasomes integrate time, space, and specific chemical information, which are essential for regulating physiological processes such as embryonic development and tumor invasion and migration. In this review, the current research progress of migrasomes, including the discovery of migrasomes and migracytosis, the structure of migrasomes, and the distribution and functions of migrasomes is discussed. The migratory marker protein TSPAN4 is highly expressed in various cancers and is associated with cancer invasion and migration. Therefore, there is still much research space for the pathogenesis of migratory bodies and cancer. This review also makes bold predictions and prospects for the research directions of the combination of migrasomes and clinical applications.
Collapse
Affiliation(s)
- Yu Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang 421001, China
| | - Minghui Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang 421001, China
| | - Zhuoyi Xie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang 421001, China
| | - Yubo Ding
- Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Jialu Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang 421001, China
| | - Jingwei Yao
- Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Yufan Lv
- Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Jianhong Zuo
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang 421001, China
- Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421002, China
- Clinical Laboratory, The Third Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421900, China
- Correspondence:
| |
Collapse
|
17
|
Wesselman HM, Nguyen TK, Chambers JM, Drummond BE, Wingert RA. Advances in Understanding the Genetic Mechanisms of Zebrafish Renal Multiciliated Cell Development. J Dev Biol 2022; 11:1. [PMID: 36648903 PMCID: PMC9844391 DOI: 10.3390/jdb11010001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cilia are microtubule-based organelles that project from the cell surface. In humans and other vertebrates, possession of a single cilium structure enables an assortment of cellular processes ranging from mechanosensation to fluid propulsion and locomotion. Interestingly, cells can possess a single cilium or many more, where so-called multiciliated cells (MCCs) possess apical membrane complexes with several dozen or even hundreds of motile cilia that beat in a coordinated fashion. Development of MCCs is, therefore, integral to control fluid flow and/or cellular movement in various physiological processes. As such, MCC dysfunction is associated with numerous pathological states. Understanding MCC ontogeny can be used to address congenital birth defects as well as acquired disease conditions. Today, researchers used both in vitro and in vivo experimental models to address our knowledge gaps about MCC specification and differentiation. In this review, we summarize recent discoveries from our lab and others that have illuminated new insights regarding the genetic pathways that direct MCC ontogeny in the embryonic kidney using the power of the zebrafish animal model.
Collapse
Affiliation(s)
| | | | | | | | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
18
|
Wang Y, Han K, Li Z, Tang X, Wang C, Zhao Y, Zhang H, Geng Z, Kong J, Luan X, Xiong Y. Protective effect of hydroxysafflor yellow A on renal ischemia‑-reperfusion injury by targeting the Akt‑Nrf2 axis in mice. Exp Ther Med 2022; 24:741. [PMID: 36478883 PMCID: PMC9716340 DOI: 10.3892/etm.2022.11677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 01/25/2023] Open
Abstract
Ischemic/reperfusion (I/R) injury is the primary cause of acute kidney injury (AKI). Hydroxysafflor yellow A (HSYA), a natural compound isolated from Carthamus tinctorius L., has been found to possess anti-inflammatory and antioxidant properties. However, the protective effects and potential mechanism of HSYA on I/R-induced AKI remains unclear. In the present study, the in vitro hypoxia/reoxygenation (H/R) and in vivo renal I/R models were employed to investigate the renal protective effects and molecular mechanisms of HSYA on I/R-induced AKI. The present results indicated that HSYA pretreatment significantly ameliorated renal damage and dysfunction in the I/R injury mice via enhancing the antioxidant capacity and suppressing the oxidative stress injury, inflammatory response, and apoptosis. Mechanistic studies showed that HSYA could upregulate Akt/GSK-3β/Fyn-Nrf2 axis-mediated antioxidant gene expression both in vitro and in vivo. Moreover, HSYA-mediated improvement in antioxidant, anti-inflammatory, and anti-apoptotic effects in H/R-treated HK-2 cells was abrogated by Akt inhibitor LY294002 supplementation. In summary, the present results demonstrated that HSYA attenuated kidney oxidative stress, inflammation response, and apoptosis induced by I/R, at least in part, via activating the Akt/GSK-3β/Fyn-Nrf2 axis pathway. These findings provided evidence that HSYA may be applied as a potential therapeutic agent in the treatment of I/R induced AKI.
Collapse
Affiliation(s)
- Yueming Wang
- Department of Pathogen Biology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Kaiyue Han
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Zile Li
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Xiaoxuan Tang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Chen Wang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Yaxuan Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Hengchao Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Ziran Geng
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Jie Kong
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China
| | - Xiying Luan
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China,Correspondence to: Professor Xiying Luan or Professor Yanlian Xiong, Department of Immunology, School of Basic Medicine, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China
| | - Yanlian Xiong
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai 264003, P.R. China,Correspondence to: Professor Xiying Luan or Professor Yanlian Xiong, Department of Immunology, School of Basic Medicine, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
19
|
Pichler R, Rizzo L, Tröndle K, Bühler M, Brucker H, Müller AL, Grand K, Farè S, Viau A, Kaminski MM, Kuehn EW, Koch F, Zimmermann S, Koltay P, Lienkamp SS. Tuning the 3D microenvironment of reprogrammed tubule cells enhances biomimetic modeling of polycystic kidney disease. Biomaterials 2022; 291:121910. [DOI: 10.1016/j.biomaterials.2022.121910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
|
20
|
Yang XL, Wang P, Ye H, Jiang M, Su YB, Peng XX, Li H, Zhang JY. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with esophageal cancer. Front Oncol 2022; 12:938234. [PMID: 36176418 PMCID: PMC9513043 DOI: 10.3389/fonc.2022.938234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolomics has been reported as an efficient tool to screen biomarkers that are related to esophageal cancer. However, the metabolic biomarkers identifying malignant degrees and therapeutic efficacy are still largely unknown in the disease. Here, GC-MS-based metabolomics was used to understand metabolic alteration in 137 serum specimens from patients with esophageal cancer, which is approximately two- to fivefold as many plasma specimens as the previous reports. The elevated amino acid metabolism is in sharp contrast to the reduced carbohydrate as a characteristic feature of esophageal cancer. Comparative metabolomics showed that most metabolic differences were determined between the early stage (0–II) and the late stage (III and IV) among the 0–IV stages of esophageal cancer and between patients who received treatment and those who did not receive treatment. Glycine, serine, and threonine metabolism and glycine were identified as the potentially overlapped metabolic pathway and metabolite, respectively, in both disease progress and treatment effect. Glycine, fructose, ornithine, and threonine can be a potential array for the evaluation of disease prognosis and therapy in esophageal cancer. These results highlight the means of identifying previously unknown biomarkers related to esophageal cancer by a metabolomics approach.
Collapse
Affiliation(s)
- Xiao-li Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Peng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ming Jiang
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Yu-bin Su
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuan-xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| | - Jian-ying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| |
Collapse
|
21
|
Zhuang M, Scholz A, Walz G, Yakulov TA. Histone Deacetylases Cooperate with NF-κB to Support the Immediate Migratory Response after Zebrafish Pronephros Injury. Int J Mol Sci 2022; 23:ijms23179582. [PMID: 36076983 PMCID: PMC9455417 DOI: 10.3390/ijms23179582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Acute kidney injury (AKI) is commonly associated with severe human diseases, and often worsens the outcome in hospitalized patients. The mammalian kidney has the ability to recover spontaneously from AKI; however, little progress has been made in the development of supportive treatments. Increasing evidence suggest that histone deacetylases (HDAC) and NF-κB promote the pathogenesis of AKI, and inhibition of Hdac activity has a protective effect in murine models of AKI. However, the role of HDAC at the early stages of recovery is unknown. We used the zebrafish pronephros model to study the role of epigenetic modifiers in the immediate repair response after injury to the tubular epithelium. Using specific inhibitors, we found that the histone deacetylase Hdac2, Hdac6, and Hdac8 activities are required for the repair via collective cell migration. We found that hdac6, hdac8, and nfkbiaa expression levels were upregulated in the repairing epithelial cells shortly after injury. Depletion of hdac6, hdac8, or nfkbiaa with morpholino oligonucleotides impaired the repair process, whereas the combined depletion of all three genes synergistically suppressed the recovery process. Furthermore, time-lapse video microscopy revealed that the lamellipodia and filopodia formation in the flanking cells was strongly reduced in hdac6-depleted embryos. Our findings suggest that Hdac activity and NF-κB are synergistically required for the immediate repair response in the zebrafish pronephros model of AKI, and the timing of HDAC inhibition might be important in developing supportive protocols in the human disease.
Collapse
Affiliation(s)
- Mingyue Zhuang
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Alexander Scholz
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Gerd Walz
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, 79104 Freiburg, Germany
| | - Toma Antonov Yakulov
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
- Correspondence:
| |
Collapse
|
22
|
Abdelhamid SS, Scioscia J, Vodovotz Y, Wu J, Rosengart A, Sung E, Rahman S, Voinchet R, Bonaroti J, Li S, Darby JL, Kar UK, Neal MD, Sperry J, Das J, Billiar TR. Multi-Omic Admission-Based Prognostic Biomarkers Identified by Machine Learning Algorithms Predict Patient Recovery and 30-Day Survival in Trauma Patients. Metabolites 2022; 12:774. [PMID: 36144179 PMCID: PMC9500723 DOI: 10.3390/metabo12090774] [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: 07/08/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 12/04/2022] Open
Abstract
Admission-based circulating biomarkers for the prediction of outcomes in trauma patients could be useful for clinical decision support. It is unknown which molecular classes of biomolecules can contribute biomarkers to predictive modeling. Here, we analyzed a large multi-omic database of over 8500 markers (proteomics, metabolomics, and lipidomics) to identify prognostic biomarkers in the circulating compartment for adverse outcomes, including mortality and slow recovery, in severely injured trauma patients. Admission plasma samples from patients (n = 129) enrolled in the Prehospital Air Medical Plasma (PAMPer) trial were analyzed using mass spectrometry (metabolomics and lipidomics) and aptamer-based (proteomics) assays. Biomarkers were selected via Least Absolute Shrinkage and Selection Operator (LASSO) regression modeling and machine learning analysis. A combination of five proteins from the proteomic layer was best at discriminating resolvers from non-resolvers from critical illness with an Area Under the Receiver Operating Characteristic curve (AUC) of 0.74, while 26 multi-omic features predicted 30-day survival with an AUC of 0.77. Patients with traumatic brain injury as part of their injury complex had a unique subset of features that predicted 30-day survival. Our findings indicate that multi-omic analyses can identify novel admission-based prognostic biomarkers for outcomes in trauma patients. Unique biomarker discovery also has the potential to provide biologic insights.
Collapse
Affiliation(s)
- Sultan S. Abdelhamid
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jacob Scioscia
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Junru Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Eight-Year Program of Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Anna Rosengart
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eunseo Sung
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Syed Rahman
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robert Voinchet
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jillian Bonaroti
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shimena Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jennifer L. Darby
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Upendra K. Kar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jason Sperry
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
23
|
Kayser N, Zaiser F, Veenstra AC, Wang H, Göcmen B, Eckert P, Franz H, Köttgen A, Walz G, Yakulov TA. Clock genes rescue nphp mutations in zebrafish. Hum Mol Genet 2022; 31:4143-4158. [PMID: 35861640 PMCID: PMC9759334 DOI: 10.1093/hmg/ddac160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 01/21/2023] Open
Abstract
The zebrafish pronephros model, using morpholino oligonucleotides (MO) to deplete target genes, has been extensively used to characterize human ciliopathy phenotypes. Recently, discrepancies between MO and genetically defined mutants have questioned this approach. We analyzed zebrafish with mutations in the nphp1-4-8 module to determine the validity of MO-based results. While MO-mediated depletion resulted in glomerular cyst and cloaca malformation, these ciliopathy-typical manifestations were observed at a much lower frequency in zebrafish embryos with defined nphp mutations. All nphp1-4-8 mutant zebrafish were viable and displayed decreased manifestations in the next (F2) generation, lacking maternal RNA contribution. While genetic compensation was further supported by the observation that nphp4-deficient mutants became partially refractory to MO-based nphp4 depletion, zebrafish embryos, lacking one nphp gene, became more sensitive to MO-based depletion of additional nphp genes. Transcriptome analysis of nphp8 mutant embryos revealed an upregulation of the circadian clock genes cry1a and cry5. MO-mediated depletion of cry1a and cry5 caused ciliopathy phenotypes in wild-type embryos, while cry1a and cry5 depletion in maternal zygotic nphp8 mutant embryos increased the frequency of glomerular cysts compared to controls. Importantly, cry1a and cry5 rescued the nephropathy-related phenotypes in nphp1, nphp4 or nphp8-depleted zebrafish embryos. Our results reveal that nphp mutant zebrafish resemble the MO-based phenotypes, albeit at a much lower frequency. Rapid adaption through upregulation of circadian clock genes seems to ameliorate the loss of nphp genes, contributing to phenotypic differences.
Collapse
Affiliation(s)
| | | | | | | | - Burulca Göcmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79106, Germany
| | - Priska Eckert
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Pestalozzistr. 20, Basel CH-4056, Switzerland
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79106, Germany
| | - Gerd Walz
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, Hugstetter Str. 55, Freiburg 79106, Germany,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, Freiburg 79104, Germany
| | - Toma A Yakulov
- To whom correspondence should be addressed. Tel: +49 76127063036;
| |
Collapse
|
24
|
Gessler S, Guthmann C, Schuler V, Lilienkamp M, Walz G, Yakulov TA. Control of Directed Cell Migration after Tubular Cell Injury by Nucleotide Signaling. Int J Mol Sci 2022; 23:ijms23147870. [PMID: 35887219 PMCID: PMC9322613 DOI: 10.3390/ijms23147870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common complication of severe human diseases, resulting in increased morbidity and mortality as well as unfavorable long-term outcomes. Although the mammalian kidney is endowed with an amazing capacity to recover from AKI, little progress has been made in recent decades to facilitate recovery from AKI. To elucidate the early repair mechanisms after AKI, we employed the zebrafish pronephros injury model. Since damaged cells release large amounts of ATP and ATP-degradation products to signal apoptosis or necrosis to neighboring cells, we examined how depletion of purinergic and adenosine receptors impacts the directed cell migration that ensues immediately after a laser-induced tubular injury. We found that depletion of the zebrafish adenosine receptors adora1a, adora1b, adora2aa, and adora2ab significantly affected the repair process. Similar results were obtained after depletion of the purinergic p2ry2 receptor, which is highly expressed during zebrafish pronephros development. Released ATP is finally metabolized to inosine by adenosine deaminase. Depletion of zebrafish adenosine deaminases ada and ada2b interfered with the repair process; furthermore, combinations of ada and ada2b, or ada2a and ada2b displayed synergistic effects at low concentrations, supporting the involvement of inosine signaling in the repair process after a tubular injury. Our findings suggest that nucleotide-dependent signaling controls immediate migratory responses after tubular injury.
Collapse
Affiliation(s)
- Sabrina Gessler
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
| | - Clara Guthmann
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
| | - Vera Schuler
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
| | - Miriam Lilienkamp
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
| | - Gerd Walz
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, 79104 Freiburg, Germany
| | - Toma Antonov Yakulov
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (S.G.); (C.G.); (V.S.); (M.L.); (G.W.)
- Correspondence:
| |
Collapse
|
25
|
GSK3β Exacerbates Myocardial Ischemia/Reperfusion Injury by Inhibiting Myc. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2588891. [PMID: 35528516 PMCID: PMC9076327 DOI: 10.1155/2022/2588891] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/01/2023]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a life-threatening disease with high morbidity and mortality. Herein, the present study is conducted to explore the regulatory mechanism of GSK3β in MI/R injury regarding cardiomyocyte apoptosis and oxidative stress. The MI/R injury mouse model and hypoxic reoxygenation (H/R) cell model were established. The expression pattern of GSK3β, FTO, KLF5, and Myc was determined followed by their relation validation. Next, loss-of-function experiments were implemented to verify the effect of GSK3β/FTO/KLF5/Myc on cardiomyocyte apoptosis and oxidative stress in the MI/R injury mouse model and H/R cell model. High expression of GSK3β and low expression of FTO, KLF5, and Myc were observed in the MI/R injury mouse model and H/R cell model. GSK3β promoted phosphorylation of FTO and KLF5, thus increasing the ubiquitination degradation of FTO and KLF5. A decrease of FTO and KLF5 was able to downregulate Myc expression, resulting in enhanced cardiomyocyte apoptosis and oxidative stress. These data together supported the crucial role that GSK3β played in facilitating cardiomyocyte apoptosis and oxidative stress so as to accelerate MI/R injury, which highlights a promising therapeutic strategy against MI/R injury.
Collapse
|
26
|
Zhang ZL, Wang D, Chen FS. MicroRNA-101a-3p mimic ameliorates spinal cord ischemia/reperfusion injury. Neural Regen Res 2022; 17:2022-2028. [PMID: 35142692 PMCID: PMC8848611 DOI: 10.4103/1673-5374.335164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
miR-101a-3p is expressed in a variety of organs and tissues and plays a regulatory role in many diseases, but its role in spinal cord ischemia/reperfusion injury remains unclear. In this study, we established a rat model of spinal cord ischemia/reperfusion injury by clamping the aortic arch for 14 minutes followed by reperfusion for 24 hours. Results showed that miR-101a-3p expression in L4–L6 spinal cord was greatly decreased, whereas MYCN expression was greatly increased. Dual-luciferase reporter assay results showed that miR-101a-3p targeted MYCN. MYCN immunoreactivity, which was primarily colocalized with neurons in L4–L6 spinal tissue, greatly increased after spinal cord ischemia/reperfusion injury. However, intrathecal injection of an miR-101a-3p mimic within 24 hours before injury decreased MYCN, p53, caspase-9 and interleukin-1β expression, reduced p53 immunoreactivity, reduced the number of MYCN/NeuN-positive cells and the number of necrotic cells in L4–L6 spinal tissue, and increased Tarlov scores. These findings suggest that the miR-101a-3p mimic improved spinal ischemia/reperfusion injury-induced nerve cell apoptosis and inflammation by inhibiting MYCN and the p53 signaling pathway. Therefore, miR-101a-3p mimic therapy may be a potential treatment option for spinal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zai-Li Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Feng-Shou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
27
|
Klingbeil K, Nguyen TQ, Fahrner A, Guthmann C, Wang H, Schoels M, Lilienkamp M, Franz H, Eckert P, Walz G, Yakulov TA. Corpuscles of Stannius development requires FGF signaling. Dev Biol 2021; 481:160-171. [PMID: 34666023 DOI: 10.1016/j.ydbio.2021.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023]
Abstract
The corpuscles of Stannius (CS) represent a unique endocrine organ of teleostean fish that secrets stanniocalcin-1 (Stc1) to maintain calcium homeostasis. Appearing at 20-25 somite stage in the distal zebrafish pronephros, stc1-expressing cells undergo apical constriction, and are subsequently extruded to form a distinct gland on top of the distal pronephric tubules at 50 h post fertilization (hpf). Several transcription factors (e.g. Hnf1b, Irx3b, Tbx2a/b) and signaling pathways (e.g. Notch) control CS development. We report now that Fgf signaling is required to commit tubular epithelial cells to differentiate into stc1-expressing CS cells. Inhibition of Fgf signaling by SU5402, dominant-negative Fgfr1, or depletion of fgf8a prevented CS formation and stc1 expression. Ablation experiments revealed that CS have the ability to partially regenerate via active cell migration involving extensive filopodia and lamellipodia formation. Activation of Wnt signaling curtailed stc1 expression, but had no effect on CS formation. Thus, our observations identify Fgf signaling as a crucial component of CS cell fate commitment.
Collapse
Affiliation(s)
- Konstantin Klingbeil
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Thanh Quang Nguyen
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Andreas Fahrner
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Clara Guthmann
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Hui Wang
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Maximilian Schoels
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Miriam Lilienkamp
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Priska Eckert
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
| | - Toma Antonov Yakulov
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
28
|
Torosyan R, Huang S, Bommi PV, Tiwari R, An SY, Schonfeld M, Rajendran G, Kavanaugh MA, Gibbs B, Truax AD, Bohney S, Calcutt MW, Kerr EW, Leonardi R, Gao P, Chandel NS, Kapitsinou PP. Hypoxic preconditioning protects against ischemic kidney injury through the IDO1/kynurenine pathway. Cell Rep 2021; 36:109547. [PMID: 34407414 PMCID: PMC8487442 DOI: 10.1016/j.celrep.2021.109547] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 05/06/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Prolonged cellular hypoxia leads to energetic failure and death. However, sublethal hypoxia can trigger an adaptive response called hypoxic preconditioning. While prolyl-hydroxylase (PHD) enzymes and hypoxia-inducible factors (HIFs) have been identified as key elements of oxygen-sensing machinery, the mechanisms by which hypoxic preconditioning protects against insults remain unclear. Here, we perform serum metabolomic profiling to assess alterations induced by two potent cytoprotective approaches, hypoxic preconditioning and pharmacologic PHD inhibition. We discover that both approaches increase serum kynurenine levels and enhance kynurenine biotransformation, leading to preservation of NAD+ in the post-ischemic kidney. Furthermore, we show that indoleamine 2,3-dioxygenase 1 (Ido1) deficiency abolishes the systemic increase of kynurenine and the subsequent renoprotection generated by hypoxic preconditioning and PHD inhibition. Importantly, exogenous administration of kynurenine restores the hypoxic preconditioning in the context of Ido1 deficiency. Collectively, our findings demonstrate a critical role of the IDO1-kynurenine axis in mediating hypoxic preconditioning.
Collapse
Affiliation(s)
- Rafael Torosyan
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shengping Huang
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Prashant V Bommi
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ratnakar Tiwari
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Si Young An
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael Schonfeld
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ganeshkumar Rajendran
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Matthew A Kavanaugh
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Benjamin Gibbs
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | - M Wade Calcutt
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Evan W Kerr
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Roberta Leonardi
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Medicine and Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pinelopi P Kapitsinou
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Medicine and Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
29
|
Chakraborty S, Balan M, Sabarwal A, Choueiri TK, Pal S. Metabolic reprogramming in renal cancer: Events of a metabolic disease. Biochim Biophys Acta Rev Cancer 2021; 1876:188559. [PMID: 33965513 PMCID: PMC8349779 DOI: 10.1016/j.bbcan.2021.188559] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
Recent studies have established that tumors can reprogram the pathways involved in nutrient uptake and metabolism to withstand the altered biosynthetic, bioenergetics and redox requirements of cancer cells. This phenomenon is called metabolic reprogramming, which is promoted by the loss of tumor suppressor genes and activation of oncogenes. Because of alterations and perturbations in multiple metabolic pathways, renal cell carcinoma (RCC) is sometimes termed as a "metabolic disease". The majority of metabolic reprogramming in renal cancer is caused by the inactivation of von Hippel-Lindau (VHL) gene and activation of the Ras-PI3K-AKT-mTOR pathway. Hypoxia-inducible factor (HIF) and Myc are other important players in the metabolic reprogramming of RCC. All types of RCCs are associated with reprogramming of glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle. Metabolism of glutamine, tryptophan and arginine is also reprogrammed in renal cancer to favor tumor growth and oncogenesis. Together, understanding these modifications or reprogramming of the metabolic pathways in detail offer ample opportunities for the development of new therapeutic targets and strategies, discovery of biomarkers and identification of effective tumor detection methods.
Collapse
Affiliation(s)
- Samik Chakraborty
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Akash Sabarwal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Toni K Choueiri
- Dana Farber Cancer Institute, Boston, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Soumitro Pal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
30
|
Song A, Jiang A, Xiong W, Zhang C. The Role of CXCL12 in Kidney Diseases: A Friend or Foe? KIDNEY DISEASES 2021; 7:176-185. [PMID: 34179113 DOI: 10.1159/000514913] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/28/2021] [Indexed: 12/25/2022]
Abstract
Background Chemokines are a family of proteins mainly mediating the homing and migration of various cells. The CXC chemokine CXCL12 is a member of low-weight-molecular chemokines. In the kidney, CXCL12 is pivotal for renal development and exerts a modulatory effect in kidney diseases under different etiologic settings by binding with CXC chemokine receptor 4 (CXCR4) or CXC chemokine receptor 7 (CXCR7). Besides, CXCL12 also exerts homeostasis influence in diverse physical conditions and various pathological situations. Thus, we conclude the complicated relationship between CXCL12 and kidney diseases in this review. Summary In renal development, CXCL12 contributes a lot to nephrogenesis and the formation of renal vasculature via correlating with CXCR4. CXCL12 also plays an essential role in renal recovery from acute kidney injury. However, the CXCL12/CXCR4 axis plays a dual regulatory role in the initiation and development of diabetic kidney disease as well as chronic allogeneic nephropathy after kidney transplantation through dialectical consideration. Additionally, the CXCL12/CXCR4 link is considered as a new risk factor for lupus nephritis and renal cell carcinoma. Key Messages Plenty of studies have presented the influence of CXCL12 and the relation with corresponding receptors in diverse biological and pathological statuses. Simultaneously, some drugs and antagonists targeting CXCL12/CXCR4 axis effectively treat various kidney diseases. However, more researches are needed to explore thorough influence and mechanisms, providing more cues for clinical treatments.
Collapse
Affiliation(s)
- Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anni Jiang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiong
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Schoels M, Zhuang M, Fahrner A, Küchlin S, Sagar, Franz H, Schmitt A, Walz G, Yakulov TA. Single-cell mRNA profiling reveals changes in solute carrier expression and suggests a metabolic switch during zebrafish pronephros development. Am J Physiol Renal Physiol 2021; 320:F826-F837. [PMID: 33749326 DOI: 10.1152/ajprenal.00610.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Developing organisms need to adapt to environmental variations as well as to rapid changes in substrate availability and energy demands imposed by fast-growing tissues and organs. Little is known about the adjustments that kidneys undergo in response to these challenges. We performed single-cell RNA sequencing of zebrafish pronephric duct cells to understand how the developing kidney responds to changes in filtered substrates and intrinsic energy requirements. We found high levels of glucose transporters early in development and increased expression of monocarboxylate transporters at later times. This indicates that the zebrafish embryonic kidney displays a high glucose transporting capacity during early development, which is replaced by the ability to absorb monocarboxylates and amino acids at later stages. This change in transport capacity was accompanied by the upregulation of mitochondrial carriers, indicating a switch to increased oxidative phosphorylation to meet the increasing energy demand of a developing kidney.NEW & NOTEWORTHY The zebrafish embryonic kidney has high levels of glucose transporters during early development, which are replaced by monocarboxylate and amino acid transporters later on. Inhibition of Na+-glucose cotransporter-dependent glucose transport by sotagliflozin also increased slc2a1a expression, supporting the idea that the glucose transport capacity is dynamically adjusted during zebrafish pronephros development. Concurrent upregulation of mitochondrial SCL25 transporters at later stages supports the idea that the pronephros adjusts to changing substrate supplies and/or energy demands during embryonic development.
Collapse
Affiliation(s)
- Maximilian Schoels
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Mingyue Zhuang
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Andreas Fahrner
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sebastian Küchlin
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.,Department of Ophthamology, Faculty of Medicine, University Freiburg Medical Center, University of Freiburg, Freiburg, Germany
| | - Sagar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Annette Schmitt
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Toma A Yakulov
- Renal Division, Department of Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
32
|
Nicotinamide Attenuates the Progression of Renal Failure in a Mouse Model of Adenine-Induced Chronic Kidney Disease. Toxins (Basel) 2021; 13:toxins13010050. [PMID: 33440677 PMCID: PMC7827863 DOI: 10.3390/toxins13010050] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) supplies energy for deoxidation and anti-inflammatory reactions fostering the production of adenosine triphosphate (ATP). The kidney is an essential regulator of body fluids through the excretion of numerous metabolites. Chronic kidney disease (CKD) leads to the accumulation of uremic toxins, which induces chronic inflammation. In this study, the role of NAD+ in kidney disease was investigated through the supplementation of nicotinamide (Nam), a precursor of NAD+, to an adenine-induced CKD mouse model. Nam supplementation reduced kidney inflammation and fibrosis and, therefore, prevented the progression of kidney disease. Notably, Nam supplementation also attenuated the accumulation of glycolysis and Krebs cycle metabolites that occurs in renal failure. These effects were due to increased NAD+ supply, which accelerated NAD+-consuming metabolic pathways. Our study suggests that Nam administration may be a novel therapeutic approach for CKD prevention.
Collapse
|
33
|
Liu C, Chen K, Wang H, Zhang Y, Duan X, Xue Y, He H, Huang Y, Chen Z, Ren H, Wang H, Zeng C. Gastrin Attenuates Renal Ischemia/Reperfusion Injury by a PI3K/Akt/Bad-Mediated Anti-apoptosis Signaling. Front Pharmacol 2020; 11:540479. [PMID: 33343341 PMCID: PMC7740972 DOI: 10.3389/fphar.2020.540479] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/24/2020] [Indexed: 12/25/2022] Open
Abstract
Ischemic/reperfusion (I/R) injury is the primary cause of acute kidney injury (AKI). Gastrin, a gastrointestinal hormone, is involved in the regulation of kidney function of sodium excretion. However, whether gastrin has an effect on kidney I/R injury is unknown. Here we show that cholecystokinin B receptor (CCKBR), the gastrin receptor, was significantly up-regulated in I/R-injured mouse kidneys. While pre-administration of gastrin ameliorated I/R-induced renal pathological damage, as reflected by the levels of serum creatinine and blood urea nitrogen, hematoxylin and eosin staining and periodic acid-Schiff staining. The protective effect could be ascribed to the reduced apoptosis for gastrin reduced tubular cell apoptosis both in vivo and in vitro. In vitro studies also showed gastrin preserved the viability of hypoxia/reoxygenation (H/R)-treated human kidney 2 (HK-2) cells and reduced the lactate dehydrogenase release, which were blocked by CI-988, a specific CCKBR antagonist. Mechanistically, the PI3K/Akt/Bad pathway participates in the pathological process, because gastrin treatment increased phosphorylation of PI3K, Akt and Bad. While in the presence of wortmannin (1 μM), a PI3K inhibitor, the gastrin-induced phosphorylation of Akt after H/R treatment was blocked. Additionally, wortmannin and Akt inhibitor VIII blocked the protective effect of gastrin on viability of HK-2 cells subjected to H/R treatment. These studies reveals that gastrin attenuates kidney I/R injury via a PI3K/Akt/Bad-mediated anti-apoptosis signaling. Thus, gastrin can be considered as a promising drug candidate to prevent AKI.
Collapse
Affiliation(s)
- Chao Liu
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Ken Chen
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Huaixiang Wang
- Department of Lishilu Outpatient, General Hospital of the PLA Rocket Force, Beijing, China
| | - Ye Zhang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Xudong Duan
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yuanzheng Xue
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongye He
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Yu Huang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Zhi Chen
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongyong Wang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China.,Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Chambers JM, Wingert RA. Advances in understanding vertebrate nephrogenesis. Tissue Barriers 2020; 8:1832844. [PMID: 33092489 PMCID: PMC7714473 DOI: 10.1080/21688370.2020.1832844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
The kidney is a complex organ that performs essential functions such as blood filtration and fluid homeostasis, among others. Recent years have heralded significant advancements in our knowledge of the mechanisms that control kidney formation. Here, we provide an overview of vertebrate renal development with a focus on nephrogenesis, the process of generating the epithelialized functional units of the kidney. These steps begin with intermediate mesoderm specification and proceed all the way to the terminally differentiated nephron cell, with many detailed stages in between. The establishment of nephron architecture with proper cellular barriers is vital throughout these processes. Continuously striving to gain further insights into nephrogenesis can ultimately lead to a better understanding and potential treatments for developmental maladies such as Congenital Anomalies of the Kidney and Urinary Tract (CAKUT).
Collapse
Affiliation(s)
- Joseph M. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
35
|
Lu XJ, Zhu K, Shen HX, Nie L, Chen J. CXCR4s in Teleosts: Two Paralogous Chemokine Receptors and Their Roles in Hematopoietic Stem/Progenitor Cell Homeostasis. THE JOURNAL OF IMMUNOLOGY 2020; 204:1225-1241. [DOI: 10.4049/jimmunol.1901100] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
|
36
|
Kleinhans DS, Lecaudey V. Standardized mounting method of (zebrafish) embryos using a 3D-printed stamp for high-content, semi-automated confocal imaging. BMC Biotechnol 2019; 19:68. [PMID: 31640669 PMCID: PMC6805687 DOI: 10.1186/s12896-019-0558-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/04/2019] [Indexed: 01/24/2023] Open
Abstract
Background Developmental biology relies to a large extent on the observation and comparison of phenotypic traits through time using high resolution microscopes. In this context, transparent model organisms such as the zebrafish Danio rerio in which developing tissues and organs can be easily observed and imaged using fluorescent proteins have become very popular. One limiting factor however is the acquisition of a sufficient amount of data, in standardized and reproducible conditions, to allow robust quantitative analysis. One way to improve this is by developing mounting methods to increase the number of embryos that can be imaged simultaneously in near-to-identical orientation. Results Here we present an improved mounting method allowing semi-automated and high-content imaging of zebrafish embryos. It is based on a 3D-printed stamp which is used to create a 2D coordinate system of multiple μ-wells in an agarose cast. Each μ-well models a negative of the average zebrafish embryo morphology between 22 and 96 h-post-fertilization. Due to this standardized and reproducible arrangement, it is possible to define a custom well plate in the respective imaging software that allows for a semi-automated imaging process. Furthermore, the improvement in Z-orientation significantly reduces post-processing and improves comparability of volumetric data while reducing light exposure and thus photo-bleaching and photo-toxicity, and improving signal-to-noise ratio (SNR). Conclusions We present here a new method that allows to standardize and improve mounting and imaging of embryos. The 3D-printed stamp creates a 2D coordinate system of μ-wells in an agarose cast thus standardizing specimen mounting and allowing high-content imaging of up to 44 live or mounted zebrafish embryos simultaneously in a semi-automated, well-plate like manner on inverted confocal microscopes. In summary, image data quality and acquisition efficiency (amount of data per time) are significantly improved. The latter might also be crucial when using the services of a microscopy facility.
Collapse
Affiliation(s)
- David Simon Kleinhans
- Department of Developmental Biology of Vertebrates, Institute for Cell biology and Neuroscience, Goethe University, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany
| | - Virginie Lecaudey
- Department of Developmental Biology of Vertebrates, Institute for Cell biology and Neuroscience, Goethe University, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Abstract
The vertebrate kidney is comprised of functional units known as nephrons. Defects in nephron development or activity are a common feature of kidney disease. Current medical treatments are unable to ameliorate the dire consequences of nephron deficit or injury. Although there have been tremendous advancements in our understanding of nephron ontogeny and the response to damage, many significant knowledge gaps still remain. The zebrafish embryo kidney, or pronephros, is an ideal model for many renal development and regeneration studies because it is comprised of nephrons that share conserved features with the nephron units that comprise the mammalian metanephric kidney. In this chapter, we provide an overview about the benefits of using the zebrafish pronephros to study the mechanisms underlying nephrogenesis as well as epithelial repair and regeneration. We subsequently detail methods for the spatiotemporal assessment of gene and protein expression in zebrafish embryos that can be used to extend the understanding of nephron development and disease, and thereby create new opportunities to identify therapeutic strategies for regenerative medicine.
Collapse
|
38
|
Sander V, Naylor RW, Davidson AJ. Mind the gap: renal tubule responses to injury and the role of Cxcl12 and Myc. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S30. [PMID: 31032310 DOI: 10.21037/atm.2019.01.80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Veronika Sander
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
| | - Richard W Naylor
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Alan J Davidson
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Chambers BE, Wingert RA. Nephron repair: powered by anaerobic energy metabolism. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S28. [PMID: 31032308 DOI: 10.21037/atm.2019.01.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Brooke E Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
40
|
Liu Q, Sheng W, Ma Y, Zhen J, Roy S, Alvira Jafar C, Xin W, Wan Q. USP36 protects proximal tubule cells from ischemic injury by stabilizing c-Myc and SOD2. Biochem Biophys Res Commun 2019; 513:502-508. [PMID: 30975468 DOI: 10.1016/j.bbrc.2019.03.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 03/08/2019] [Indexed: 12/19/2022]
Abstract
Acute kidney injury (AKI) is a progressive renal injury with high morbidity and mortality, however, the mechanism is far from being clarified and effective clinical interventions are lacking. USP36 is a deubiquitination enzyme involved in a variety of cellular biological processes, but its involvement in renal cell apoptosis and kidney disease is largely unknown. In the present study, we confirmed the decreased expression of USP36 both in vivo in mouse and human AKI samples and in vitro ischemic human renal proximal tubular cells, which are extremely sensitive to the damage of ischemic injury. Importantly, we found that overexpression of USP36 markedly decreased ischemia-induced apoptosis and oxidative stress in HK-2 cells, which was accompanied by elevated c-Myc and SOD2 protein levels with alleviated ischemia-induced ubiquitination of both proteins. Our findings revealed a novel role of USP36 in inhibiting apoptosis of human renal tubular cells induced by ischemia, and provided a potential therapeutic target for AKI treatment.
Collapse
Affiliation(s)
- Qing Liu
- Weifang Medical University, 261000, Weifang, Shandong Province, China
| | - Wei Sheng
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China
| | - Yuan Ma
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Junhui Zhen
- Department of Pathology, Shandong University School of Medicine, Jinan, 250012, China
| | - Satyajit Roy
- Department of Nephrology and Dialysis Unit, Bangabondhu Memorial Hospital Affiliated to University of Science & Technology, Chittagong, Bangladesh
| | | | - Wei Xin
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China.
| | - Qiang Wan
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China.
| |
Collapse
|
41
|
Prostaglandin signaling regulates renal multiciliated cell specification and maturation. Proc Natl Acad Sci U S A 2019; 116:8409-8418. [PMID: 30948642 PMCID: PMC6486750 DOI: 10.1073/pnas.1813492116] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multiciliated cells (MCCs) have core roles in organ formation and function, where they control fluid flow and particle displacement. MCCs direct fluid movement in the brain and spinal cord, clearance of respiratory mucus, and ovum transport from the ovary to the uterus. Deficiencies in MCC functionality lead to hydrocephalus, chronic respiratory infections, and infertility. Prostaglandins are lipids that are used to coordinate cellular functions. Here, we discovered that prostaglandin signaling is required for MCC development in the embryonic zebrafish kidney. Understanding renal MCC genesis can lend insights into the puzzling origins of MCCs in several chronic kidney diseases, where it is unclear whether MCCs are a cause or phenotypic outcome of the condition. Multiciliated cells (MCCs) are specialized epithelia with apical bundles of motile cilia that direct fluid flow. MCC dysfunction is associated with human diseases of the respiratory, reproductive, and central nervous systems. Further, the appearance of renal MCCs has been cataloged in several kidney conditions, where their function is unknown. Despite their pivotal health importance, many aspects of MCC development remain poorly understood. Here, we utilized a chemical screen to identify molecules that affect MCC ontogeny in the zebrafish embryo kidney, and found prostaglandin signaling is essential both for renal MCC progenitor formation and terminal differentiation. Moreover, we show that prostaglandin activity is required downstream of the transcription factor ets variant 5a (etv5a) during MCC fate choice, where modulating prostaglandin E2 (PGE2) levels rescued MCC number. The discovery that prostaglandin signaling mediates renal MCC development has broad implications for other tissues, and could provide insight into a multitude of pathological states.
Collapse
|