51
|
Dong R, Xu Y. Glomerular cell cross talk in diabetic kidney diseases. J Diabetes 2022; 14:514-523. [PMID: 35999686 PMCID: PMC9426281 DOI: 10.1111/1753-0407.13304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes mellitus. It is the leading inducement of end-stage renal disease (ESRD), and its global incidence has been increasing at an alarming rate. The strict control of blood pressure and blood glucose can delay the progression of DKD, but intensive treatment is challenging to maintain. Studies to date have failed to find a complete cure. The glomerulus's alterations and injuries play a pivotal role in the initiation and development of DKD. A wealth of data indicates that the interdependent relationship between resident cells in the glomerulus will provide clues to the mechanism of DKD and new ways for therapeutic intervention. This review summarizes the significant findings of glomerular cell cross talk in DKD, focusing on cellular signaling pathways, regulators, and potential novel avenues for treating progressive DKD.
Collapse
Affiliation(s)
- Ruixue Dong
- Faculty of Pharmacy, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
| | - Youhua Xu
- Faculty of Pharmacy, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- Department of Endocrinology, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, People's Republic of China
| |
Collapse
|
52
|
Raghubar AM, Pham DT, Tan X, Grice LF, Crawford J, Lam PY, Andersen SB, Yoon S, Teoh SM, Matigian NA, Stewart A, Francis L, Ng MSY, Healy HG, Combes AN, Kassianos AJ, Nguyen Q, Mallett AJ. Spatially Resolved Transcriptomes of Mammalian Kidneys Illustrate the Molecular Complexity and Interactions of Functional Nephron Segments. Front Med (Lausanne) 2022; 9:873923. [PMID: 35872784 PMCID: PMC9300864 DOI: 10.3389/fmed.2022.873923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Available transcriptomes of the mammalian kidney provide limited information on the spatial interplay between different functional nephron structures due to the required dissociation of tissue with traditional transcriptome-based methodologies. A deeper understanding of the complexity of functional nephron structures requires a non-dissociative transcriptomics approach, such as spatial transcriptomics sequencing (ST-seq). We hypothesize that the application of ST-seq in normal mammalian kidneys will give transcriptomic insights within and across species of physiology at the functional structure level and cellular communication at the cell level. Here, we applied ST-seq in six mice and four human kidneys that were histologically absent of any overt pathology. We defined the location of specific nephron structures in the captured ST-seq datasets using three lines of evidence: pathologist's annotation, marker gene expression, and integration with public single-cell and/or single-nucleus RNA-sequencing datasets. We compared the mouse and human cortical kidney regions. In the human ST-seq datasets, we further investigated the cellular communication within glomeruli and regions of proximal tubules-peritubular capillaries by screening for co-expression of ligand-receptor gene pairs. Gene expression signatures of distinct nephron structures and microvascular regions were spatially resolved within the mouse and human ST-seq datasets. We identified 7,370 differentially expressed genes (p adj < 0.05) distinguishing species, suggesting changes in energy production and metabolism in mouse cortical regions relative to human kidneys. Hundreds of potential ligand-receptor interactions were identified within glomeruli and regions of proximal tubules-peritubular capillaries, including known and novel interactions relevant to kidney physiology. Our application of ST-seq to normal human and murine kidneys confirms current knowledge and localization of transcripts within the kidney. Furthermore, the generated ST-seq datasets provide a valuable resource for the kidney community that can be used to inform future research into this complex organ.
Collapse
Affiliation(s)
- Arti M. Raghubar
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Duy T. Pham
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Xiao Tan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Laura F. Grice
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Joanna Crawford
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Pui Yeng Lam
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Stacey B. Andersen
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
- UQ Sequencing Facility, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Sohye Yoon
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
| | - Siok Min Teoh
- UQ Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas A. Matigian
- QCIF Facility for Advanced Bioinformatics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Anne Stewart
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
| | - Leo Francis
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
| | - Monica S. Y. Ng
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Nephrology Department, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Helen G. Healy
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Alexander N. Combes
- Department of Anatomy and Developmental Biology, Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Andrew J. Kassianos
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Mallett
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, Queensland, QLD, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, Queensland, QLD, Australia
| |
Collapse
|
53
|
Glycation of Tie-2 Inhibits Angiopoietin-1 Signaling Activation and Angiopoietin-1-Induced Angiogenesis. Int J Mol Sci 2022; 23:ijms23137137. [PMID: 35806141 PMCID: PMC9266685 DOI: 10.3390/ijms23137137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/04/2023] Open
Abstract
The impairment of the angiopoietin-1 (Ang-1)/Tie-2 signaling pathway has been thought to play a critical role in diabetic complications. However, the underlying mechanisms remain unclear. The present study aims to investigate the effects of Tie-2 glycation on Ang-1 signaling activation and Ang-1-induced angiogenesis. We identified that Tie-2 was modified by advanced glycation end products (AGEs) in aortae derived from high fat diet (HFD)-fed mice and in methylglyoxal (MGO)-treated human umbilical vein endothelial cells (HUVECs). MGO-induced Tie-2 glycation significantly inhibited Ang-1-evoked Tie-2 and Akt phosphorylation and Ang-1-regulated endothelial cell migration and tube formation, whereas the blockade of AGE formation by aminoguanidine remarkably rescued Ang-1 signaling activation and Ang-1-induced angiogenesis in vitro. Furthermore, MGO treatment markedly increased AGE cross-linking of Tie-2 in cultured aortae ex vivo and MGO-induced Tie-2 glycation also significantly decreased Ang-1-induced vessel outgrow from aortic rings. Collectively, these data suggest that Tie-2 may be modified by AGEs in diabetes mellitus and that Tie-2 glycation inhibits Ang-1 signaling activation and Ang-1-induced angiogenesis. This may provide a novel mechanism for Ang-1/Tie-2 signal dysfunction and angiogenesis failure in diabetic ischaemic diseases.
Collapse
|
54
|
Network Pharmacology and In Vivo Experimental Validation to Uncover the Renoprotective Mechanisms of Fangji Huangqi Decoction on Nephrotic Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4223729. [PMID: 35722158 PMCID: PMC9200505 DOI: 10.1155/2022/4223729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022]
Abstract
Background Fangji Huangqi decoction (FHD) is a traditional Chinese medicine formula that has the potential efficacy for nephrotic syndrome (NS) treatment. This study aims to explore the effects and underlying mechanisms of FHD against NS via network pharmacology and in vivo experiments. Methods The bioactive compounds and targets of FHD were retrieved from the TCMSP database. NS-related targets were collected from GeneCards and DisGeNET databases. The compound-target and protein-protein interaction networks were constructed by Cytoscape 3.8 and BisoGenet, respectively. GO and KEGG analyses were performed by the DAVID online tool. The interactions between active compounds and hub genes were revealed by molecular docking. An NS rat model was established to validate the renoprotective effects and molecular mechanisms of FHD against NS in vivo. Results A total of 32 hub genes were predicted to play essential roles in FHD treating NS. Eight main bioactive compounds of FHD had the good affinity with 9 hub targets (CCL2, IL-10, PTGS2, TNF, MAPK1, IL-6, CXCL8, TP53, and VEGFA). The therapeutic effect of FHD on NS was closely involved in the regulation of inflammation and PI3K-Akt pathway. In vivo experiments confirmed the renoprotective effect of FHD on NS, evidenced by reducing the levels of proteinuria, serum creatinine, blood urea nitrogen, and inflammatory factors in NS rats. The PI3K activator 740Y-P weakened the effects of FHD against NS. Furthermore, FHD downregulated the levels of PTGS2, MAPK1, IL-6, and p-Akt in NS rats. Conclusions FHD alleviates kidney injury and inflammation in NS by targeting PTGS2, MAPK1, IL-6, and PI3K-Akt pathway.
Collapse
|
55
|
Mifune T, Tanabe K, Nakashima Y, Tanimura S, Sugiyama H, Sato Y, Wada J. Vasohibin-1 has α-tubulin detyrosinating activity in glomerular podocytes. Biochem Biophys Res Commun 2022; 599:93-99. [PMID: 35180473 DOI: 10.1016/j.bbrc.2022.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/11/2022] [Indexed: 11/27/2022]
Abstract
Podocytes are highly specialized epithelial cells in glomeruli, with a complex morphology composed of a cell body, primary processes, and foot processes, which maintain barrier function in glomerular filtration. The microtubule-based cytoskeleton is necessary for podocyte morphology. Microtubule structure and function can be affected by post-translational modification of tubulin, including detyrosination. Recent studies have shown that vasohibin-1 (VASH1), an antiangiogenic factor, has tubulin carboxypeptidase activity that causes detyrosination of α-tubulin. We aimed to examine the role of VASH1 in regulating α-tubulin detyrosination in podocytes and the potential involvement of VASH1 deficiency in renal morphology. In normal mouse kidneys, detyrosinated α-tubulin was mainly identified in glomeruli, especially in podocytes; meanwhile, in cultured immortalized podocytes, α-tubulin detyrosination was promoted with cell differentiation. Notably, α-tubulin detyrosination in glomeruli was diminished in Vash1 homozygous knockout (Vash1-/-) mice, and knockdown of VASH1 in cultured podocytes prevented α-tubulin detyrosination. Although VASH1 deficiency-induced downregulation of detyrosination caused no remarkable glomerular lesions, urinary albuminuria excretion and glomerular volume were significantly higher in Vash1-/- mice than in wild-type mice. Furthermore, decreased glomerular nephrin expression and narrower slit diaphragms width were observed in Vash1-/- mice. Taken together, we demonstrated that α-tubulin detyrosination in podocytes was mainly regulated by VASH1 and that VASH1 deficiency-mediated decreases in α-tubulin detyrosination led to minor alterations in podocyte morphology and predisposition to albuminuria. VASH1 expression and α-tubulin detyrosination may be novel targets for maintaining glomerular filtration barrier integrity.
Collapse
Affiliation(s)
- Tomoyo Mifune
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Tanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Yuri Nakashima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Tanimura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Sugiyama
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasufumi Sato
- New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
56
|
Guan Y, Davis L, Breyer MD, Hao CM. Cyclooxygenase-2 contributes to diabetic nephropathy through glomerular EP4 receptor. Prostaglandins Other Lipid Mediat 2022; 159:106621. [DOI: 10.1016/j.prostaglandins.2022.106621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 11/27/2022]
|
57
|
Abstract
The kidney maintains electrolyte, water, and acid-base balance, eliminates foreign and waste compounds, regulates blood pressure, and secretes hormones. There are at least 16 different highly specialized epithelial cell types in the mammalian kidney. The number of specialized endothelial cells, immune cells, and interstitial cell types might even be larger. The concerted interplay between different cell types is critical for kidney function. Traditionally, cells were defined by their function or microscopical morphological appearance. With the advent of new single-cell modalities such as transcriptomics, epigenetics, metabolomics, and proteomics we are entering into a new era of cell type definition. This new technological revolution provides new opportunities to classify cells in the kidney and understand their functions.
Collapse
Affiliation(s)
- Michael S Balzer
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Philadelphia, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Philadelphia, USA
| |
Collapse
|
58
|
Dai C, Waduge P, Ji L, Huang C, He Y, Tian H, Zuniga-Sanchez E, Bhatt A, Pang IH, Su G, Webster KA, Li W. Secretogranin III stringently regulates pathological but not physiological angiogenesis in oxygen-induced retinopathy. Cell Mol Life Sci 2022; 79:63. [PMID: 35006382 PMCID: PMC9007175 DOI: 10.1007/s00018-021-04111-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 01/12/2023]
Abstract
Conventional angiogenic factors, such as vascular endothelial growth factor (VEGF), regulate both pathological and physiological angiogenesis indiscriminately, and their inhibitors may elicit adverse side effects. Secretogranin III (Scg3) was recently reported to be a diabetes-restricted VEGF-independent angiogenic factor, but the disease selectivity of Scg3 in retinopathy of prematurity (ROP), a retinal disease in preterm infants with concurrent pathological and physiological angiogenesis, was not defined. Here, using oxygen-induced retinopathy (OIR) mice, a surrogate model of ROP, we quantified an exclusive binding of Scg3 to diseased versus healthy developing neovessels that contrasted sharply with the ubiquitous binding of VEGF. Functional immunohistochemistry visualized Scg3 binding exclusively to disease-related disorganized retinal neovessels and neovascular tufts, whereas VEGF bound to both disorganized and well-organized neovessels. Homozygous deletion of the Scg3 gene showed undetectable effects on physiological retinal neovascularization but markedly reduced the severity of OIR-induced pathological angiogenesis. Furthermore, anti-Scg3 humanized antibody Fab (hFab) inhibited pathological angiogenesis with similar efficacy to anti-VEGF aflibercept. Aflibercept dose-dependently blocked physiological angiogenesis in neonatal retinas, whereas anti-Scg3 hFab was without adverse effects at any dose and supported a therapeutic window at least 10X wider than that of aflibercept. Therefore, Scg3 stringently regulates pathological but not physiological angiogenesis, and anti-Scg3 hFab satisfies essential criteria for development as a safe and effective disease-targeted anti-angiogenic therapy for ROP.
Collapse
Affiliation(s)
- Chang Dai
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Prabuddha Waduge
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Liyang Ji
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Chengchi Huang
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Ye He
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Hong Tian
- Everglades Biopharma, LLC, Houston, Texas, USA
| | - Elizabeth Zuniga-Sanchez
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Amit Bhatt
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Texas Children Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Iok-Hou Pang
- Dept. of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas, Fort Worth, Texas, USA
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Keith A. Webster
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA.,Everglades Biopharma, LLC, Houston, Texas, USA
| | - Wei Li
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
59
|
Tziastoudi M, Cholevas C, Theoharides TC, Stefanidis I. Meta-Analysis and Bioinformatics Detection of Susceptibility Genes in Diabetic Nephropathy. Int J Mol Sci 2021; 23:ijms23010020. [PMID: 35008447 PMCID: PMC8744540 DOI: 10.3390/ijms23010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/16/2022] Open
Abstract
The latest meta-analysis of genome-wide linkage studies (GWLS) identified nine cytogenetic locations suggestive of a linkage with diabetic nephropathy (DN) due to type 1 diabetes mellitus (T1DM) and seven locations due to type 2 diabetes mellitus (T2DM). In order to gain biological insight about the functional role of the genes located in these regions and to prioritize the most significant genetic loci for further research, we conducted a gene ontology analysis with an over representation test for the functional annotation of the protein coding genes. Protein analysis through evolutionary relationships (PANTHER) version 16.0 software and Cytoscape with the relevant plugins were used for the gene ontology analysis, and the overrepresentation test and STRING database were used for the construction of the protein network. The findings of the over-representation test highlight the contribution of immune related molecules like immunoglobulins, cytokines, and chemokines with regard to the most overrepresented protein classes, whereas the most enriched signaling pathways include the VEGF signaling pathway, the Cadherin pathway, the Wnt pathway, the angiogenesis pathway, the p38 MAPK pathway, and the EGF receptor signaling pathway. The common section of T1DM and T2DM results include the significant over representation of immune related molecules, and the Cadherin and Wnt signaling pathways that could constitute potential therapeutic targets for the treatment of DN, irrespective of the type of diabetes.
Collapse
Affiliation(s)
- Maria Tziastoudi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece;
- Correspondence: ; Tel.: +30-2413501667; Fax: +30-2413501015
| | - Christos Cholevas
- First Department of Ophthalmology, Faculty of Health Sciences, Aristotle University of Thessaloniki School of Medicine, AHEPA Hospital, 54636 Thessaloniki, Greece;
| | | | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece;
| |
Collapse
|
60
|
Black LM, Farrell ER, Barwinska D, Osis G, Zmijewska AA, Traylor AM, Esman SK, Bolisetty S, Whipple G, Kamocka MM, Winfree S, Spangler DR, Khan S, Zarjou A, El-Achkar TM, Agarwal A. VEGFR3 tyrosine kinase inhibition aggravates cisplatin nephrotoxicity. Am J Physiol Renal Physiol 2021; 321:F675-F688. [PMID: 34658261 PMCID: PMC8714977 DOI: 10.1152/ajprenal.00186.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elisa R Farrell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daria Barwinska
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Gunars Osis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anna A Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephanie K Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Grace Whipple
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Malgorzata M Kamocka
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Seth Winfree
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Daryll R Spangler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
- Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
61
|
Liu H, Hiremath C, Patterson Q, Vora S, Shang Z, Jamieson AR, Fiolka R, Dean KM, Dellinger MT, Marciano DK. Heterozygous Mutation of Vegfr3 Reduces Renal Lymphatics without Renal Dysfunction. J Am Soc Nephrol 2021; 32:3099-3113. [PMID: 34551997 PMCID: PMC8638391 DOI: 10.1681/asn.2021010061] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Lymphatic abnormalities are observed in several types of kidney disease, but the relationship between the renal lymphatic system and renal function is unclear. The discovery of lymphatic-specific proteins, advances in microscopy, and available genetic mouse models provide the tools to help elucidate the role of renal lymphatics in physiology and disease. METHODS We utilized a mouse model containing a missense mutation in Vegfr3 (dubbed Chy ) that abrogates its kinase ability. Vegfr3 Chy/+ mice were examined for developmental abnormalities and kidney-specific outcomes. Control and Vegfr3 Chy/+ mice were subjected to cisplatin-mediated injury. We characterized renal lymphatics using tissue-clearing, light-sheet microscopy, and computational analyses. RESULTS In the kidney, VEGFR3 is expressed not only in lymphatic vessels but also, in various blood capillaries. Vegfr3 Chy/+ mice had severely reduced renal lymphatics with 100% penetrance, but we found no abnormalities in BP, serum creatinine, BUN, albuminuria, and histology. There was no difference in the degree of renal injury after low-dose cisplatin (5 mg/kg), although Vegfr3 Chy/+ mice developed perivascular inflammation. Cisplatin-treated controls had no difference in total cortical lymphatic volume and length but showed increased lymphatic density due to decreased cortical volume. CONCLUSIONS We demonstrate that VEGFR3 is required for development of renal lymphatics. Our studies reveal that reduced lymphatic density does not impair renal function at baseline and induces only modest histologic changes after mild injury. We introduce a novel quantification method to evaluate renal lymphatics in 3D and demonstrate that accurate measurement of lymphatic density in CKD requires assessment of changes to cortical volume.
Collapse
Affiliation(s)
- Hao Liu
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quinten Patterson
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Saumya Vora
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiguo Shang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew R. Jamieson
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Reto Fiolka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas,Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kevin M. Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael T. Dellinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K. Marciano
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
62
|
Anticancer Effect of Heparin-Taurocholate Conjugate on Orthotopically Induced Exocrine and Endocrine Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13225775. [PMID: 34830928 PMCID: PMC8616444 DOI: 10.3390/cancers13225775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Pancreatic cancer has a less than 9% 5-year survival rate among patients because it is very difficult to detect and diagnose early. Combinatorial chemotherapy with surgery or radiotherapy is a potential remedy to treat pancreatic cancer. However, these strategies still have side effects such as hair loss, skin soreness and fatigue. To overcome these side effects, angiogenesis inhibitors such as sunitinib are used to deliver targeted blood vessels around tumor tissues, including pancreatic cancer tumors. It is still controversial whether antiangiogenesis therapy is sufficient to treat pancreatic cancer. So far, many scientists have not been focused on the tumor types of pancreatic cancer when they have developed antipancreatic cancer medication. Here, we used heparin–taurocholate (LHT) as an anticancer drug to treat pancreatic cancer through inhibition of angiogenic growth factors. In this study, we examined the anticancer efficacy of LHT on various types of pancreatic cancer in an orthotopic model. Abstract Pancreatic cancers are classified based on where they occur, and are grouped into those derived from exocrine and those derived from neuroendocrine tumors, thereby experiencing different anticancer effects under medication. Therefore, it is necessary to develop anticancer drugs that can inhibit both types. To this end, we developed a heparin–taurocholate conjugate, i.e., LHT, to suppress tumor growth via its antiangiogenic activity. Here, we conducted a study to determine the anticancer efficacy of LHT on pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine tumor (PNET), in an orthotopic animal model. LHT reduced not only proliferation of cancer cells, but also attenuated the production of VEGF through ERK dephosphorylation. LHT effectively reduced the migration, invasion and tube formation of endothelial cells via dephosphorylation of VEGFR, ERK1/2, and FAK protein. Especially, these effects of LHT were much stronger on PNET (RINm cells) than PDAC (PANC1 and MIA PaCa-2 cells). Eventually, LHT reduced ~50% of the tumor weights and tumor volumes of all three cancer cells in the orthotopic model, via antiproliferation of cancer cells and antiangiogenesis of endothelial cells. Interestingly, LHT had a more dominant effect in the PNET-induced tumor model than in PDAC in vivo. Collectively, these findings demonstrated that LHT could be a potential antipancreatic cancer medication, regardless of pancreatic cancer types.
Collapse
|
63
|
Zhou HY, Sui H, Zhao YJ, Qian HJ, Yang N, Liu L, Guan Q, Zhou Y, Lin HL, Wang DP. The Impact of Inflammatory Immune Reactions of the Vascular Niche on Organ Fibrosis. Front Pharmacol 2021; 12:750509. [PMID: 34776968 PMCID: PMC8585779 DOI: 10.3389/fphar.2021.750509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Inflammation is a type of defense response against tissue damage, and can be mediated by lymphocytes and macrophages. Fibrosis is induced by tissue injury and inflammation, which leads to an increase in fibrous connective tissue in organs and a decrease in organ parenchyma cells, finally leading to organ dysfunction or even failure. The vascular niche is composed of endothelial cells, pericytes, macrophages, and hematopoietic stem cells. It forms a guiding microenvironment for the behavior of adjacent cells, and mainly exists in the microcirculation, including capillaries. When an organ is damaged, the vascular niche regulates inflammation and affects the repair of organ damage in a variety of ways, such as via its angiocrine function and transformation of myofibroblasts. In this paper, the main roles of vascular niche in the process of organ fibrosis and its mechanism of promoting the progress of fibrosis through inflammatory immunoregulation are summarized. It was proposed that the vascular niche should be regarded as a new therapeutic target for organ fibrosis, suggesting that antifibrotic effects could be achieved by regulating macrophages, inhibiting endothelial-mesenchymal transition, interfering with the angiocrine function of endothelial cells, and inhibiting the transformation of pericytes into myofibroblasts, thus providing new ideas for antifibrosis drug research.
Collapse
Affiliation(s)
- Hong-Yan Zhou
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hua Sui
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yang-Jianing Zhao
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Hong-Jie Qian
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Nan Yang
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Lu Liu
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Qing Guan
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yue Zhou
- Department of Nephrology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hong-Li Lin
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China.,Department of Nephrology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Da-Peng Wang
- Institude college of Integrative Medicine, Dalian Medical University, Dalian, China.,Department of Nephrology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
64
|
Shirakura K, Okada Y. Vascular Leakage Prevention by Roundabout 4 under Pathological Conditions. Biol Pharm Bull 2021; 44:1365-1370. [PMID: 34602544 DOI: 10.1248/bpb.b21-00413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular permeability is regulated mainly by the endothelial barrier and controls vascular homeostasis, proper vessel development, and immune cell trafficking. Several molecules are involved in regulating endothelial barrier function. Roundabout 4 (Robo4) is a single-pass transmembrane protein that is specifically expressed in vascular endothelial cells. Robo4 is an important regulator of vascular leakage and angiogenesis, especially under pathological conditions. The role of Robo4 in preventing vascular leakage has been studied in various disease models, including animal models of retinopathy, tumors, diabetes, and endotoxemia. The involvement of Robo4 in vascular endothelial growth factor and inflammation-mediated signaling pathways has been well studied, and recent evidence suggests that Robo4 modulates endothelial barrier function via distinct mechanisms. In this review, we discuss the role of Robo4 in endothelial barrier function and the underlying molecular mechanisms.
Collapse
Affiliation(s)
| | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
65
|
Black LM, Winfree S, Khochare SD, Kamocka MM, Traylor AM, Esman SK, Khan S, Zarjou A, Agarwal A, El-Achkar TM. Quantitative 3-dimensional imaging and tissue cytometry reveals lymphatic expansion in acute kidney injury. J Transl Med 2021; 101:1186-1196. [PMID: 34017058 PMCID: PMC8373805 DOI: 10.1038/s41374-021-00609-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/27/2022] Open
Abstract
The lymphatic system plays an integral role in physiology and has recently been identified as a key player in disease progression. Tissue injury stimulates lymphatic expansion, or lymphangiogenesis (LA), though its precise role in disease processes remains unclear. LA is associated with inflammation, which is a key component of acute kidney injury (AKI), for which there are no approved therapies. While LA research has gained traction in the last decade, there exists a significant lack of understanding of this process in the kidney. Though innovative studies have elucidated markers and models with which to study LA, the field is still evolving with ways to visualize lymphatics in vivo. Prospero-related homeobox-1 (Prox-1) is the master regulator of LA and determines lymphatic cell fate through its action on vascular endothelial growth factor receptor expression. Here, we investigate the consequences of AKI on the abundance and distribution of lymphatic endothelial cells using Prox1-tdTomato reporter mice (ProxTom) coupled with large-scale three-dimensional quantitative imaging and tissue cytometry (3DTC). Using these technologies, we describe the spatial dynamics of lymphatic vasculature in quiescence and post-AKI. We also describe the use of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) as a marker of lymphatic vessels using 3DTC in the absence of the ProxTom reporter mice as an alternative approach. The use of 3DTC for lymphatic research presents a new avenue with which to study the origin and distribution of renal lymphatic vessels. These findings will enhance our understanding of renal lymphatic function during injury and could inform the development of novel therapeutics for intervention in AKI.
Collapse
Affiliation(s)
- Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Seth Winfree
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Suraj D Khochare
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Malgorzata M Kamocka
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie K Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shehnaz Khan
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Veterans Affairs, Birmingham, AL, USA.
| | - Tarek M El-Achkar
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA.
- Indianapolis Veterans Affairs Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
66
|
He J, Zhang S, Qiu Z, Li X, Huang H, Jin W, Xu Y, Shao G, Wang L, Meng J, Wang S, Geng X, Jia Y, Li M, Yang B, Jenny Lu HA, Zhou H. Inhibiting Focal Adhesion Kinase Ameliorates Cyst Development in Polycystin-1-Deficient Polycystic Kidney Disease in Animal Model. J Am Soc Nephrol 2021; 32:2159-2174. [PMID: 34465607 PMCID: PMC8729842 DOI: 10.1681/asn.2020111560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/07/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is characterized by numerous cysts originating from renal tubules and is associated with significant tubular epithelial cell proliferation. Focal adhesion kinase (FAK) promotes tumor growth by regulating multiple proliferative pathways. METHODS We established the forskolin (FSK)-induced three-dimensional (3D) Madin-Darby Canine Kidney cystogenesis model and 8-bromoadenosine-3`,5`-cyclic monophosphate-stimulated cyst formation in ex vivo embryonic kidney culture. Cultured human renal cyst-lining cells (OX-161) and normal tubular epithelial cells were treated with FAK inhibitors or transfected with green fluorescent protein-tagged FAK mutant plasmids for proliferation study. Furthermore, we examined the role of FAK in two transgenic ADPKD animal models, the kidney-specific Pkd1 knockout and the collecting duct-specific Pkd1 knockout mouse models. RESULTS FAK activity was significantly elevated in OX-161 cells and in two ADPKD mouse models. Inhibiting FAK activity reduced cell proliferation in OX-161 cells and prevented cyst growth in ex vivo and 3D cyst models. In tissue-specific Pkd1 knockout mouse models, FAK inhibitors retarded cyst development and mitigated renal function decline. Mechanically, FSK stimulated FAK activation in tubular epithelial cells, which was blocked by a protein kinase A (PKA) inhibitor. Inhibition of FAK activation by inhibitors or transfected cells with mutant FAK constructs interrupted FSK-mediated Src activation and upregulation of ERK and mTOR pathways. CONCLUSIONS Our study demonstrates the critical involvement of FAK in renal cyst development, suggests that FAK is a potential therapeutic target in treating patients with ADPKD, and highlights the role of FAK in cAMP-PKA-regulated proliferation.
Collapse
Affiliation(s)
- Jinzhao He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Shun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiwei Qiu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaowei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Huihui Huang
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - William Jin
- Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, Massachusetts
| | - Yue Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guangying Shao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jia Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shuyuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoqiang Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingli Jia
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hua A. Jenny Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| |
Collapse
|
67
|
Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis. Clin Sci (Lond) 2021; 135:1999-2029. [PMID: 34427291 DOI: 10.1042/cs20201016] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD.
Collapse
|
68
|
Marchetti A, Rosellini M, Rizzo A, Mollica V, Battelli N, Massari F, Santoni M. An up-to-date evaluation of cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother 2021; 22:2323-2336. [PMID: 34405738 DOI: 10.1080/14656566.2021.1959548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: In the evolving treatment scenario of metastatic renal cell carcinoma, cabozantinib is gaining increasing attention, presenting as a cornerstone therapy, both as a monotherapy and in combination with immune-checkpoint inhibitors.Areas covered: In this review, the authors explore the role of cabozantinib in the treatment of metastatic clear cell and non-clear cell renal cell carcinoma, presenting data from the most recent clinical trials and investigating ongoing studies. They, furthermore, evaluate the pharmacokinetic, pharmacodynamic, and immunomodulatory effect of cabozantinib, as well as underlining the tolerability profile and patients' quality of life.Expert opinion: Cabozantinib's administration as a single agent is restricted to intermediate- and poor-risk patients (according to IMDC criteria). The further advent of anti-VEGF-receptor tyrosine kinase inhibitors combined with immune checkpoint inhibitor regimens (such as pembrolizumab + axitinib) has allowed to expand the use of cabozantinib, leading to its combination with nivolumab. In the next few years, more information is required to look for the application of cabozantinib-based combinations as a later-line approach in metastatic RCC patients, beside their use in the first-line setting.
Collapse
Affiliation(s)
- Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | | |
Collapse
|
69
|
Liu D, Du Y, Jin FY, Xu XL, Du YZ. Renal Cell-Targeted Drug Delivery Strategy for Acute Kidney Injury and Chronic Kidney Disease: A Mini-Review. Mol Pharm 2021; 18:3206-3222. [PMID: 34337953 DOI: 10.1021/acs.molpharmaceut.1c00511] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), have become a global public health concern associated with high morbidity, mortality, and healthcare costs. However, at present, very few effective and specific drug therapies are available, owing to the poor therapeutic efficacy and systemic side effects. Kidney-targeted drug delivery, as a potential strategy for solving these problems, has received great attention in the fields of AKI and CKD in recent years. Here, we review the literature on renal targeted, more specifically, renal cell-targeted formulations of AKI and CKD that offered biodistribution data. First, we provide a broad overview of the unique structural characteristics and injured cells of acute and chronic injured kidneys. We then separately summarize literature examples of renal targeted formulations according to the difference of target cells and elaborate on the appropriate formulation design criteria for AKI and CKD. Finally, we propose a hypothetic strategy to improve the renal accumulation of glomerular cell-targeted formulation by escaping the uptake of the reticuloendothelial system and provide some perspectives for future studies.
Collapse
Affiliation(s)
- Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yan Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Fei-Yang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| |
Collapse
|
70
|
Thomas MC. Targeting the Pathobiology of Diabetic Kidney Disease. Adv Chronic Kidney Dis 2021; 28:282-289. [PMID: 34922684 DOI: 10.1053/j.ackd.2021.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 07/06/2021] [Indexed: 12/23/2022]
Abstract
The pathobiology of diabetic kidney disease (DKD) involves an interplay between all the many different cell types that exist within the kidney and their shared and cumulative dysfunction in response to chronic hyperglycemia. DKD is characteriszed by morphological changes including tubular hypertrophy, podocyte dysfunction, mesangial expansion and mesangiolysis, endothelitis and capillary rarefaction, arteriolar hyalinosis, basement membrane thickening, and ultimately nephron dropout and tubulointerstitial fibrosis. These adaptive but ultimately maladaptive changes accelerate the progression of lesions in the diabetic kidney by increasing mechanical and oxidative stress, hypoxia, fibrogenesis, inflammation, senescence, and apoptosis. In particular, atrophy at the critical junction between Bowman's capsule and the proximal tubule likely represent the leading cause of nephron dropout and kidney function decline in DKD. Preventing, slowing, or reversing these changes should be the target of future "smart" therapies for patients with DKD, many of which are now under development.
Collapse
|
71
|
A Review of Traditional Chinese Medicine in Treating Renal Interstitial Fibrosis via Endoplasmic Reticulum Stress-Mediated Apoptosis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6667791. [PMID: 34055995 PMCID: PMC8147530 DOI: 10.1155/2021/6667791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/23/2021] [Accepted: 05/07/2021] [Indexed: 01/07/2023]
Abstract
Renal interstitial fibrosis (RIF) is the main pathological manifestation of end-stage renal disease. Recent studies have shown that endoplasmic reticulum (ER) stress is involved in the pathogenesis and development of RIF. Traditional Chinese medicine (TCM), as an effective treatment for kidney diseases, can improve kidney damage by affecting the apoptotic signaling pathway mediated by ER stress. This article reviews the apoptotic pathways mediated by ER stress, including the three major signaling pathways of unfolded protein response, the main functions of the transcription factor C/EBP homologous protein. We also present current research on TCM treatment of RIF, focusing on medicines that regulate ER stress. A new understanding of using TCM to treat kidney disease by regulating ER stress will promote clinical application of Chinese medicine and discovery of new drugs for the treatment of RIF.
Collapse
|
72
|
Ren Y, Zhang Y, Wang L, He F, Yan M, Liu X, Ou Y, Wu Q, Bi T, Wang S, Liu J, Ding BS, Wang L, Qing J. Selective Targeting of Vascular Endothelial YAP Activity Blocks EndMT and Ameliorates Unilateral Ureteral Obstruction-Induced Kidney Fibrosis. ACS Pharmacol Transl Sci 2021; 4:1066-1074. [PMID: 34151201 DOI: 10.1021/acsptsci.1c00010] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Kidney fibrosis is accompanied by vascular dysfunction. Discovering new ways to ameliorate dysfunctional angiogenesis may bypass kidney fibrosis. YAP (Yes-associated protein) plays a multifaceted role during angiogenesis. Here, we found that selectively targeting YAP signaling in the endothelium ameliorates unilateral ureteral obstruction (UUO)-induced kidney fibrosis. Genetic deletion of Yap1, encoding YAP protein, in VE-cadherin+ endothelial cells inhibited endothelial-to-mesenchymal transition (EndMT) and dysfunctional angiogenesis and improved obstructive nephropathy and kidney fibrosis. Treatment with the systemic YAP inhibitor verteporfin worsened kidney fibrosis symptoms because of its lack of cell specificity. In an attempt to identify endothelial-specific YAP modulators, we found that G-protein-coupled receptor coagulation factor II receptor-like 1 (F2RL1) was highly expressed in vessels after UUO-induced kidney fibrosis. The F2RL1 peptide antagonist FSLLRY-NH2 selectively blocked YAP activity in endothelial cells and ameliorated kidney fibrosis. Thus, selective antagonization of endothelial YAP activity might bypass kidney fibrosis and provide new avenues for the design of antifibrotic therapies.
Collapse
Affiliation(s)
- Yafeng Ren
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Yuwei Zhang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lu Wang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Fuqian He
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Mengli Yan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Xiaoheng Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yangying Ou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Qinkai Wu
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, Canada
| | - Tao Bi
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Shiyuan Wang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jian Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China.,Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10128, United States.,Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York 10065, United States
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jie Qing
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| |
Collapse
|
73
|
Krishnan S, Suarez-Martinez AD, Bagher P, Gonzalez A, Liu R, Murfee WL, Mohandas R. Microvascular dysfunction and kidney disease: Challenges and opportunities? Microcirculation 2021; 28:e12661. [PMID: 33025626 PMCID: PMC9990864 DOI: 10.1111/micc.12661] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
Kidneys are highly vascular organs that despite their relatively small size receive 20% of the cardiac output. The highly intricate, delicately organized structure of renal microcirculation is essential to enable renal function and glomerular filtration rate through the local modulation of renal blood flow and intraglomerular pressure. Not surprisingly, the dysregulation of blood flow within the microvessels (abnormal vasoreactivity), fibrosis driven by disordered vascular-renal cross talk, or the loss of renal microvasculature (rarefaction) is associated with kidney disease. In addition, kidney disease can cause microcirculatory dysfunction in distant organs such as the heart and brain, mediated by mechanisms that remain to be elucidated. The objective of this review is to highlight the role of renal microvasculature in kidney disease. The overview will outline the impetus to study renal microvasculature, the bidirectional relationship between kidney disease and microvascular dysfunction, the key pathways driving microvascular diseases such as vasoreactivity, the cell dynamics coordinating fibrosis, and vessel rarefaction. Finally, we will also briefly highlight new therapies targeting the renal microvasculature to improve renal function.
Collapse
Affiliation(s)
- Suraj Krishnan
- Division of Nephrology, Hypertension & Transplantation, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ariana D Suarez-Martinez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Pooneh Bagher
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Rajesh Mohandas
- Division of Nephrology, Hypertension & Transplantation, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
74
|
Nephrotoxicity of Anti-Angiogenic Therapies. Diagnostics (Basel) 2021; 11:diagnostics11040640. [PMID: 33916159 PMCID: PMC8066213 DOI: 10.3390/diagnostics11040640] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/19/2021] [Accepted: 03/29/2021] [Indexed: 12/24/2022] Open
Abstract
The use of inhibitors of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) signaling for the treatment of cancer has increased over the last decade. This signaling pathway plays a fundamental role in angiogenesis and also in kidney physiology. The emergence of anti-angiogenic therapies has led to adverse nephrotoxic effects, despite improving the outcomes of patients. In this review, we will present the different anti-angiogenic therapies targeting the VEGFR pathway in association with the incidence of renal manifestations during their use. In addition, we will discuss, in detail, the pathophysiological mechanisms of frequent renal diseases such as hypertension, proteinuria, renal dysfunction, and electrolyte disorders. Finally, we will outline the cellular damage described following these therapies.
Collapse
|
75
|
Hamroun A, Puech P, Maanaoui M, Bouyé S, Hazzan M, Lionet A. Renal Lymphangiectasia, a Rare Complication After Kidney Transplantation. Kidney Int Rep 2021; 6:1475-1479. [PMID: 34013129 PMCID: PMC8116723 DOI: 10.1016/j.ekir.2021.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/04/2021] [Accepted: 03/01/2021] [Indexed: 02/03/2023] Open
Affiliation(s)
- Aghilès Hamroun
- Lille University, Lille University Hospital Center, Department of Nephrology, Dialysis and Kidney Transplantation, Lille, France.,National Institute of Health and Medical Research, Center for research in Epidemiology and Population Health (CESP), Clinical Epidemiology Team, Villejuif, France
| | - Philippe Puech
- Lille University Hospital Center, Department of Radiology, Lille University, Lille, France.,U1189 - ONCO-THAI - Image Assisted Laser Therapy for Oncology, Lille, France
| | - Mehdi Maanaoui
- Lille University, Lille University Hospital Center, Department of Nephrology, Dialysis and Kidney Transplantation, Lille, France.,INSERM U1190, Translational Research for Diabetes, Lille, France
| | - Sébastien Bouyé
- Department of Urology, Lille University, Regional and University Hospital Center of Lille, Lille, France
| | - Marc Hazzan
- Lille University, Lille University Hospital Center, Department of Nephrology, Dialysis and Kidney Transplantation, Lille, France
| | - Arnaud Lionet
- Lille University, Lille University Hospital Center, Department of Nephrology, Dialysis and Kidney Transplantation, Lille, France
| |
Collapse
|
76
|
Mahtal N, Lenoir O, Tharaux PL. Glomerular Endothelial Cell Crosstalk With Podocytes in Diabetic Kidney Disease. Front Med (Lausanne) 2021; 8:659013. [PMID: 33842514 PMCID: PMC8024520 DOI: 10.3389/fmed.2021.659013] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes is the main cause of renal failure worldwide. Complications of the kidney micro-and macro-circulation are common in diabetic patients, leading to proteinuria and can progress to end-stage renal disease. Across the complex interplays aggravating diabetes kidney disease progression, lesions of the glomerular filtration barrier appear crucial. Among its components, glomerular endothelial cells are known to be central safeguards of plasma filtration. An array of evidence has recently pinpointed its intricate relations with podocytes, highly specialized pericytes surrounding glomerular capillaries. During diabetic nephropathy, endothelial cells and podocytes are stressed and damaged. Besides, each can communicate with the other, directly affecting the progression of glomerular injury. Here, we review recent studies showing how in vitro and in vivo studies help to understand pathological endothelial cells-podocytes crosstalk in diabetic kidney disease.
Collapse
Affiliation(s)
- Nassim Mahtal
- Université de Paris, Paris Cardiovascular Center, Inserm, Paris, France
| | - Olivia Lenoir
- Université de Paris, Paris Cardiovascular Center, Inserm, Paris, France
| | | |
Collapse
|
77
|
Xiao Y, Deng J, Li C, Gong X, Gui Z, Huang J, Zhang Y, Liu Y, Ye X, Li X. Epiberberine ameliorated diabetic nephropathy by inactivating the angiotensinogen (Agt) to repress TGFβ/Smad2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 83:153488. [PMID: 33571918 DOI: 10.1016/j.phymed.2021.153488] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe microvascular complication of diabetes with prominent morbidity and mortality. At present, there are hardly any effective drugs to treat DN. Epiberberine (EPI), an isoquinoline alkaloid, has attracted considerable attention due to its anti-hyperglycemic, anti-hyperlipidemic, and anti-inflammatory functions. However, whether there is a protective effect of EPI on DN has not been reported. PURPOSE The research was aimed to investigate the activities of EPI alleviating kidney damage in db/db mice and to explore its possible mechanisms. STUDY DESIGN The db/db mice and high-glucose (HG) induced glomerular mesangial cells (GMCs) were used to explore the protective effect of EPI on DN in vivo and in vitro. METHODS The changes in fasting blood glucose, metabolic index, renal function, and histopathological morphology in db/db mice were detected to evaluate the therapeutic effect of EPI. Then, renal transcriptome and molecular docking were used to screen the key targets. Subsequently, HG-induced GMCs through mimicing the pathological changes in DN were utilized to study the renal protective effects of EPI and its potential mechanism. RESULTS The results in vivo showed that EPI administration for 8 weeks significantly alleviated diabetes-related metabolic disorders, improved renal functions, and relieved the histopathological abnormalities of renal tissue, especially renal fibrosis in db/db mice. The results in vitro showed that EPI inhibited the proliferation and induced the G2/M phase arrest of HG-induced GMCs. Moreover, a key gene Angiotensinogen (Agt) was screen out by the RNA-seq of kidney and molecular docking, and EPI reduced Agt, TGFβ1, and Smad2 expression in vitro and in vivo. Noteworthy, Agt knockdown by siRNA significantly attenuated these beneficial efficacies exerted by EPI, indicating that Agt played a crucial role in the process of EPI improving DN. CONCLUSION These findings suggested that EPI might be a potential drug for the treatment of DN dependent on the Agt-TGFβ/Smad2 pathway.
Collapse
Affiliation(s)
- Yaping Xiao
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Jianling Deng
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Chunming Li
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Xiaobao Gong
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Zhenwei Gui
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jieyao Huang
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Yaru Zhang
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Yan Liu
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China
| | - Xiaoli Ye
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xuegang Li
- College of Pharmaceutical Sciences. Translational Pharmacy Center of Medical Research Institute. Southwest University, Chongqing 400716, China.
| |
Collapse
|
78
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
79
|
Jeong JH, Ojha U, Lee YM. Pathological angiogenesis and inflammation in tissues. Arch Pharm Res 2020; 44:1-15. [PMID: 33230600 PMCID: PMC7682773 DOI: 10.1007/s12272-020-01287-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022]
Abstract
The role of angiogenesis in the growth of organs and tumors is widely recognized. Vascular-organ interaction is a key mechanism and a concept that enables an understanding of all biological phenomena and normal physiology that is essential for human survival under pathological conditions. Recently, vascular endothelial cells have been classified as a type of innate immune cells that are dependent on the pathological situations. Moreover, inflammatory cytokines and signaling regulators activated upon exposure to infection or various stresses play crucial roles in the pathological function of parenchymal cells, peripheral immune cells, stromal cells, and cancer cells in tissues. Therefore, vascular-organ interactions as a vascular microenvironment or tissue microenvironment under physiological and pathological conditions are gaining popularity as an interesting research topic. Here, we review vascular contribution as a major factor in microenvironment homeostasis in the pathogenesis of normal as well as cancerous tissues. Furthermore, we suggest that the normalization strategy of pathological angiogenesis could be a promising therapeutic target for various diseases, including cancer.
Collapse
Affiliation(s)
- Ji-Hak Jeong
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea.,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea. .,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
80
|
Fuzheng Huayu recipe, a traditional Chinese compound herbal medicine, attenuates renal interstitial fibrosis via targeting the miR-21/PTEN/AKT axis. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:505-513. [PMID: 32912827 DOI: 10.1016/j.joim.2020.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) may be viable targets for treating renal interstitial fibrosis (RIF). Fuzheng Huayu recipe (FZHY), a traditional Chinese compound herbal medicine, is often used in China to treat fibrosis. This study sought to assess the mechanisms through which FZHY influences miRNAs to treat RIF. METHODS RIF was induced in rats by mercury chloride and treated with FZHY. Hydroxyproline content, Masson's staining and type I collagen expression were used to evaluate renal collagen deposition. Renal miRNA profiles were evaluated using a miRNA microarray. Those miRNAs that were differentially expressed following FZHY treatment were identified and subjected to bioinformatic analyses. The miR-21 target gene phosphatase and tensin homolog (PTEN) expression and AKT phosphorylation in kidney tissues were assessed via Western blotting. In addition, HK-2 human proximal tubule epithelial cells were treated using angiotensin II (Ang-II) to induce epithelial-to-mesenchymal transition (EMT), followed by FZHY exposure. miR-21 and PTEN expressions were evaluated via quantitative reverse transcription-polymerase chain reaction (qRT-PCR), while E-cadherin and α-smooth muscle actin (α-SMA) expressions were assessed by immunofluorescent staining and qRT-PCR. Western blotting was used to assess PTEN and AKT phosphorylation. RESULTS FZHY significantly decreased kidney collagen deposition, hydroxyproline content and type I collagen level. The miRNA microarray identified 20 miRNAs that were differentially expressed in response to FZHY treatment. Subsequent bioinformatic analyses found that miR-21 was the key fibrosis-related miRNA regulated by FZHY. FZHY also decreased PTEN expression and AKT phosphorylation in fibrotic kidneys. Results from in vitro tests also suggested that FZHY promoted E-cadherin upregulation and inhibited α-SMA expression in Ang-II-treated HK-2 cells, effectively reversing Ang-II-mediated EMT. We also determined that FZHY reduced miR-21 expression, increased PTEN expression and decreased AKT phosphorylation in these cells. CONCLUSION miR-21 is the key fibrosis-related miRNA regulated by FZHY. The ability of FZHY to modulate miR-21/PTEN/AKT signaling may be a viable approach for treating RIF.
Collapse
|
81
|
Genetic Deletion of Vasohibin-2 Exacerbates Ischemia-Reperfusion-Induced Acute Kidney Injury. Int J Mol Sci 2020; 21:ijms21124545. [PMID: 32604722 PMCID: PMC7352238 DOI: 10.3390/ijms21124545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) has been increasingly recognized as a risk factor for transition to chronic kidney disease. Recent evidence suggests that endothelial damage in peritubular capillaries can accelerate the progression of renal injury. Vasohibin-2 (VASH2) is a novel proangiogenic factor that promotes tumor angiogenesis. However, the pathophysiological roles of VASH2 in kidney diseases remain unknown. In the present study, we examined the effects of VASH2 deficiency on the progression of ischemia–reperfusion (I/R) injury-induced AKI. I/R injury was induced by bilaterally clamping renal pedicles for 25 min in male wild-type (WT) and Vash2 homozygous knockout mice. Twenty-four hours later, I/R injury-induced renal dysfunction and tubular damage were more severe in VASH2-deficient mice than in WT mice, with more prominent neutrophil infiltration and peritubular capillary loss. After induction of I/R injury, VASH2 expression was markedly increased in injured renal tubules. These results suggest that VASH2 expression in renal tubular epithelial cells might be essential for alleviating I/R injury-induced AKI, probably through protecting peritubular capillaries and preventing inflammatory infiltration.
Collapse
|