1
|
Addario G, Moroni L, Mota C. Kidney Fibrosis In Vitro and In Vivo Models: Path Toward Physiologically Relevant Humanized Models. Adv Healthc Mater 2025:e2403230. [PMID: 39906010 DOI: 10.1002/adhm.202403230] [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: 08/27/2024] [Revised: 01/14/2025] [Indexed: 02/06/2025]
Abstract
Chronic kidney disease (CKD) affects over 10% of the global population and is a leading cause of mortality. Kidney fibrosis, a key endpoint of CKD, disrupts nephron tubule anatomy and filtration function, and disease pathomechanisms are not fully understood. Kidney fibrosis is currently investigated with in vivo models, that gradually support the identification of possible mechanisms of fibrosis, but with limited translational research, as they do not fully recapitulate human kidney physiology, metabolism, and molecular pathways. In vitro 2D cell culture models are currently used, as a starting point in disease modeling and pharmacology, however, they lack the 3D kidney architecture complexity and functions. The failure of several therapies and drugs in clinical trials highlights the urgent need for advanced 3D in vitro models. This review discusses the urinary system's anatomy, associated diseases, and diagnostic methods, including biomarker analysis and tissue biopsy. It evaluates 2D and in vivo models, highlighting their limitations. The review explores the state-of-the-art 3D-humanized in vitro models, such as 3D cell aggregates, on-chip models, biofabrication techniques, and hybrid models, which aim to mimic kidney morphogenesis and functions. These advanced models hold promise for translating new therapies and drugs for kidney fibrosis into clinics.
Collapse
Affiliation(s)
- Gabriele Addario
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| | - Carlos Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, ER Maastricht, 6229, The Netherlands
| |
Collapse
|
2
|
Hashimoto K, Yoshida Y, Kamesawa M, Yazawa N, Tominaga H, Aisyah R, Chen S, Bumrungkit C, Kawamoto S, Kumrungsee T, Yanaka N. Glucosyl Hesperidin Supplementation Prevents Tubulointerstitial Fibrosis and Immune Activation in Diabetic Nephropathy in Mice. Nutrients 2025; 17:383. [PMID: 39940240 PMCID: PMC11820413 DOI: 10.3390/nu17030383] [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: 11/29/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a serious condition that can result in end-stage renal failure. Recent evidence has focused on the dietary effects of polyphenols on blood glucose levels and the complications of diabetes. OBJECTIVES In this study, we investigated the protective effect of glucosyl hesperidin (G-Hes), composed of glucose and hesperidin, against streptozotocin (STZ)-induced nephropathy in mice. METHODS We used an STZ-induced diabetic mouse model to investigate the preventive effect of G-Hes on renal pathology. After G-Hes supplementation for 4 weeks, we investigated the renal gene expression profiles using DNA microarray analysis and renal histology to examine the underlying molecular mechanism. RESULTS G-Hes suppressed the increase in kidney weight without any change in the blood glucose levels. This study identified 511 genes whose expression levels were substantially increased during DN development but were downregulated by G-Hes supplementation. G-Hes prevented mRNA expression associated with renal tubule injury, fibrosis, and immune responses. Notably, G-Hes supplementation considerably decreased the complement component C3 at the mRNA and protein levels in the glomeruli and ameliorated glomerular and mesangial matrix expansion in diabetic nephropathy. CONCLUSIONS G-Hes supplementation is useful in preventing tubulointerstitial fibrosis and inflammation in a mouse model of DN, without exhibiting a hypoglycemic effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan (S.K.); (T.K.)
| |
Collapse
|
3
|
Zhang H, Zhang L, Tian D, Bai Y, Feng Y, Liu W, Diao Z. CD146⁺ Endothelial Cells Facilitate Renal Interstitial Fibrosis Through Endothelial-to-Mesenchymal Transition. Ann Transplant 2024; 29:e945917. [PMID: 39654278 PMCID: PMC11645843 DOI: 10.12659/aot.945917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/15/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Endothelial cells play a crucial role in the pathogenesis of renal interstitial fibrosis (RIF), with CD146 being upregulated on injured endothelial cells. However, the precise contribution of CD146⁺ endothelial cells to RIF remains unclear. This study aimed to observe and detect the relationship between CD146 expression and endothelial cells and to explore the role and possible mechanism of CD146 promoting endothelial-mesenchymal transition in RIF. MATERIAL AND METHODS In this study, we investigated the association between CD146⁺ endothelial cells and RIF. Double-label immunofluorescence was used in patients with chronic kidney disease, whereas multiplex immunofluorescence staining was used for the analysis in unilateral ureteral obstruction (UUO) mice. Hematoxylin and eosin and Masson trichrome staining were performed to evaluate RIF. RESULTS Our results revealed an elevation of CD146⁺ endothelial cells, which positively correlated with the degree of RIF in chronic kidney disease patients and UUO mice. Notably, CD146⁺ endothelial cells undergoing endothelial-mesenchymal transition (CD146⁺ EndMT) were significantly higher in subjects with severe renal interstitial fibrosis, as observed in chronic kidney disease patients and UUO mice. Additionally, with the progression of renal interstitial fibrosis, the expression of PDGFRb, the receptor of PDGF-B signaling pathway, increased and co-localized with CD146⁺ CD31⁺ a-SMA⁺ cells. The proportion of CD146⁺ CD31⁺ alpha-SMA⁺ PDGFRß⁺ cells in CD31⁺ cells increased. CONCLUSIONS In the process of renal interstitial fibrosis, CD146 is mainly expressed in renal interstitial vascular endothelial cells and participates in endothelial-to-mesenchymal transition, which may be related to the PDGF-B/PDGFR-ß signaling pathway.
Collapse
Affiliation(s)
- Huixian Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Liling Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
- Department of Emergency, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, PR China
| | - Dongli Tian
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Yu Bai
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Yiduo Feng
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| |
Collapse
|
4
|
Gong Y, Zhu W, Li Y, Lu T, Tan J, He C, Yang L, Zhu Y, Gong L. Dynamic regulation of proximal tubular autophagy from injury to repair after ischemic kidney damage. Cell Mol Biol Lett 2024; 29:151. [PMID: 39639205 PMCID: PMC11619129 DOI: 10.1186/s11658-024-00663-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The role of proximal tubular autophagy in repairing kidney injury following ischemia remains unclear. METHODS In this study, we utilized mice with conditional deletion of the Atg5 gene in proximal tubules and monitored the long-term dynamic regulation of autophagy following ischemic acute kidney injury (AKI). RESULTS The results showed that Atg5-deficient proximal tubule epithelial cells exhibited damaged mitochondria, concentric membranes, and lysosomal accumulation 24 h after ischemia/reperfusion. However, 28 days after ischemia/reperfusion, concentric membrane bodies remained, but lysosomal accumulation was no longer observed. Notably, the absence of Atg5 in renal tubular epithelial cells impaired renal function and led to increased tubular cell proliferation and oxidative stress in the early stage of injury. However, during the repair period following AKI, Atg5 deficiency exhibited no significant difference in the expression of proliferating cell nuclear antigen (PCNA) and 4-hydoxynonenal (4HNE), suggesting that the improvement in renal fibrosis associated with Atg5 deficiency is unlikely to result from its effect on cell proliferation or reactive oxygen species levels. Additionally, Atg5 deficiency inhibits the secretion of profibrotic factor fibroblast growth factor 2 (FGF2) from the early stage of renal injury to the recovery stage of AKI, indicating that autophagy-specific regulation of FGF2 secretion is a dynamic process overlapping with other stages of injury. Furthermore, increased co-localization of ATG5 with 4HNE and FGF2 was observed in patient samples. CONCLUSION In summary, our results suggest that the dynamic regulation of autophagy on key molecules involved in kidney injury and repair varies with the stage of kidney injury.
Collapse
Affiliation(s)
- Yuhong Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wei Zhu
- Department of Infectious Diseases, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yongqiang Li
- Department of General Practice, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Tao Lu
- Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No. 7 People's Hospital, Changzhou, 213011, China
| | - Jiexing Tan
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Changsheng He
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Luodan Yang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Yufeng Zhu
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Li Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. Sci Rep 2024; 14:29307. [PMID: 39592775 PMCID: PMC11599588 DOI: 10.1038/s41598-024-80930-0] [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: 07/16/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Davy Vanhoutte
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaitlynn Stowers
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Michelle Sargent
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mike Adam
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Anne Karina T Perl
- Division of Neonatology and Pulmonary biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander G Miethke
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Burg
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tiffany Shi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - E Steve S Woodle
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA.
| |
Collapse
|
6
|
Bai M, Xu S, Jiang M, Guo Y, Hu D, He J, Wang T, Zhang Y, Guo Y, Zhang Y, Huang S, Jia Z, Zhang A. Meis1 Targets Protein Tyrosine Phosphatase Receptor J in Fibroblast to Retard Chronic Kidney Disease Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309754. [PMID: 39162106 PMCID: PMC11497016 DOI: 10.1002/advs.202309754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/11/2024] [Indexed: 08/21/2024]
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) with the proliferation and activation of myofibroblasts being definite effectors and drivers. Here, increased expression of Meis1 (myeloid ecotropic viral integration site 1) is observed, predominantly in the nucleus of the kidney of CKD patients and mice, and negatively correlates with serum creatinine. Fibroblast-specific knock-in of Meis1 inhibits myofibroblast activation and attenuates renal fibrosis and kidney dysfunction in CKD models. Overexpression of Meis1 in NRK-49F cells suppresses the pro-fibrotic response induced by transforming growth factor-β1 but accelerates by its knockdown. Mechanistically, Meis1 targets protein tyrosine phosphatase receptor J (Ptprj) to block renal fibrosis by inhibiting the proliferation and activation of fibroblasts. Finally, a new activator of Ptprj is identified through computer-aided virtual screening, which has the effect of alleviating renal fibrosis. Collectively, these results illustrate that the Meis1/Ptprj axis has therapeutic potential for clinically treating CKD.
Collapse
Affiliation(s)
- Mi Bai
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Shuang Xu
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Mingzhu Jiang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yuxian Guo
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Dandan Hu
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Jia He
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Ting Wang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yu Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Medical School of Nanjing UniversityNanjing210093China
| | - Yan Guo
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yue Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Songming Huang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Zhanjun Jia
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Aihua Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| |
Collapse
|
7
|
Muns JA, Schooten E, van Dasselaar RDJ, Noordman YE, Adamzek K, Eibergen AC, Pronk SD, Cali S, Sijbrandi NJ, Merkul E, Oliveira S, van Bergen En Henegouwen PMP, Takkenberg RB, Verheij J, van de Graaf SFJ, Nijmeijer BA, van Dongen GAMS. Preclinical targeting of liver fibrosis with a 89Zr-labeled Fibrobody® directed against platelet derived growth factor receptor-β. Eur J Nucl Med Mol Imaging 2024; 51:3545-3558. [PMID: 38888612 PMCID: PMC11445362 DOI: 10.1007/s00259-024-06785-9] [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: 04/17/2024] [Accepted: 06/01/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE Hepatic fibrosis develops as a response to chronic liver injury, resulting in the formation of fibrous scars. This process is initiated and driven by collagen-producing activated myofibroblasts which reportedly express high levels of platelet derived growth factor receptor-β (PDGFRβ). We therefore regard PDGFRβ as an anchor for diagnosis and therapy. The Fibrobody® SP02SP26-ABD is a biparatopic VHH-construct targeting PDGFRβ. Here, we explore its potential as a theranostic vector for liver fibrosis. METHODS Specificity, cross-species binding, and cellular uptake of SP02SP26-ABD was assessed using human, mouse and rat PDGFRβ ectodomains and PDGFRβ-expressing cells. Cellular uptake by PDGFRβ-expressing cells was also evaluated by equipping the Fibrobody® with auristatinF and reading out in vitro cytotoxicity. The validity of PDGFRβ as a marker for active fibrosis was confirmed in human liver samples and 3 mouse models of liver fibrosis (DDC, CCl4, CDA-HFD) through immunohistochemistry and RT-PCR. After radiolabeling of DFO*-SP02SP26-ABD with 89Zr, its in vivo targeting ability was assessed in healthy mice and mice with liver fibrosis by PET-CT imaging, ex vivo biodistribution and autoradiography. RESULTS SP02SP26-ABD shows similar nanomolar affinity for human, mouse and rat PDGFRβ. Cellular uptake and hence subnanomolar cytotoxic potency of auristatinF-conjugated SP02SP26-ABD was observed in PDGFRβ-expressing cell lines. Immunohistochemistry of mouse and human fibrotic livers confirmed co-localization of PDGFRβ with markers of active fibrosis. In all three liver fibrosis models, PET-CT imaging and biodistribution analysis of [89Zr]Zr-SP02SP26-ABD revealed increased PDGFRβ-specific uptake in fibrotic livers. In the DDC model, liver uptake was 12.15 ± 0.45, 15.07 ± 0.90, 20.23 ± 1.34, and 20.93 ± 4.35%ID/g after 1,2,3 and 4 weeks of fibrogenesis, respectively, compared to 7.56 ± 0.85%ID/g in healthy mice. Autoradiography revealed preferential uptake in the fibrotic (PDGFRβ-expressing) periportal areas. CONCLUSION The anti-PDGFRβ Fibrobody® SP02SP26-ABD shows selective and high-degree targeting of activated myofibroblasts in liver fibrosis, and qualifies as a vector for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Joey A Muns
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands
| | - Erik Schooten
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands
| | | | | | - Kevin Adamzek
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands
| | | | - Sebas D Pronk
- Department of Biology, Division of Cell Biology, Neurology and Biophysics, Science Faculty, Utrecht University, Utrecht, the Netherlands
| | - Sagel Cali
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands
| | | | - Eugen Merkul
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands
| | - Sabrina Oliveira
- Department of Biology, Division of Cell Biology, Neurology and Biophysics, Science Faculty, Utrecht University, Utrecht, the Netherlands
- Department of Pharmaceutical Sciences, Pharmaceutics Devision, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Paul M P van Bergen En Henegouwen
- Department of Biology, Division of Cell Biology, Neurology and Biophysics, Science Faculty, Utrecht University, Utrecht, the Netherlands
| | - R Bart Takkenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Guus A M S van Dongen
- LinXis Biopharmaceuticals, Amsterdam, the Netherlands.
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Yoshida T, Latt KZ, Rosenberg AZ, Santo BA, Myakala K, Ishimoto Y, Zhao Y, Shrivastav S, Jones BA, Yang X, Wang XX, Tutino VM, Sarder P, Levi M, Okamoto K, Winkler CA, Kopp JB. PKR activation-induced mitochondrial dysfunction in HIV-transgenic mice with nephropathy. eLife 2024; 12:RP91260. [PMID: 39207915 PMCID: PMC11361708 DOI: 10.7554/elife.91260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
HIV disease remains prevalent in the USA and chronic kidney disease remains a major cause of morbidity in HIV-1-positive patients. Host double-stranded RNA (dsRNA)-activated protein kinase (PKR) is a sensor for viral dsRNA, including HIV-1. We show that PKR inhibition by compound C16 ameliorates the HIV-associated nephropathy (HIVAN) kidney phenotype in the Tg26 transgenic mouse model, with reversal of mitochondrial dysfunction. Combined analysis of single-nucleus RNA-seq and bulk RNA-seq data revealed that oxidative phosphorylation was one of the most downregulated pathways and identified signal transducer and activator of transcription (STAT3) as a potential mediating factor. We identified in Tg26 mice a novel proximal tubular cell cluster enriched in mitochondrial transcripts. Podocytes showed high levels of HIV-1 gene expression and dysregulation of cytoskeleton-related genes, and these cells dedifferentiated. In injured proximal tubules, cell-cell interaction analysis indicated activation of the pro-fibrogenic PKR-STAT3-platelet-derived growth factor (PDGF)-D pathway. These findings suggest that PKR inhibition and mitochondrial rescue are potential novel therapeutic approaches for HIVAN.
Collapse
Affiliation(s)
- Teruhiko Yoshida
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
| | - Khun Zaw Latt
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Briana A Santo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at BuffaloBuffaloUnited States
| | - Komuraiah Myakala
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown UniversityWashingtonUnited States
| | - Yu Ishimoto
- Polycystic Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
| | - Yongmei Zhao
- Frederick National Laboratory for Cancer Research, NCI, NIHFrederickUnited States
| | - Shashi Shrivastav
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
| | - Bryce A Jones
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown UniversityWashingtonUnited States
| | - Xiaoping Yang
- Department of Pathology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown UniversityWashingtonUnited States
| | - Vincent M Tutino
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at BuffaloBuffaloUnited States
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at BuffaloBuffaloUnited States
- College of Medicine, University of FloridaGainesvilleUnited States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown UniversityWashingtonUnited States
| | - Koji Okamoto
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
- Nephrology Endocrinology and Vascular Medicine, Tohoku University HospitalSendaiJapan
| | - Cheryl A Winkler
- Frederick National Laboratory for Cancer Research, NCI, NIHFrederickUnited States
| | - Jeffrey B Kopp
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIHBethesdaUnited States
| |
Collapse
|
9
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. RESEARCH SQUARE 2024:rs.3.rs-4746078. [PMID: 39184103 PMCID: PMC11343171 DOI: 10.21203/rs.3.rs-4746078/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
|
10
|
Cimini M, Hansmann UHE, Gonzalez C, Chesney AD, Truongcao MM, Gao E, Wang T, Roy R, Forte E, Mallaredy V, Thej C, Magadum A, Joladarashi D, Benedict C, Koch WJ, Tükel Ç, Kishore R. Podoplanin Positive Cell-derived Extracellular Vesicles Contribute to Cardiac Amyloidosis After Myocardial Infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601297. [PMID: 39005419 PMCID: PMC11244852 DOI: 10.1101/2024.06.28.601297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Amyloidosis is a major long-term complication of chronic disease; however, whether it represents one of the complications of post-myocardial infarction (MI) is yet to be fully understood. Methods Using wild-type and knocked-out MI mouse models and characterizing in vitro the exosomal communication between bone marrow-derived macrophages and activated mesenchymal stromal cells (MSC) isolated after MI, we investigated the mechanism behind Serum Amyloid A 3 (SAA3) protein overproduction in injured hearts. Results Here, we show that amyloidosis occurs after MI and that amyloid fibers are composed of macrophage-derived SAA3 monomers. SAA3 overproduction in macrophages is triggered by exosomal communication from a subset of activated MSC, which, in response to MI, acquire the expression of a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin (PDPN). Cardiac MSC PDPN+ communicate with and activate macrophages through their extracellular vesicles or exosomes. Specifically, MSC PDPN+ derived exosomes (MSC PDPN+ Exosomes) are enriched in SAA3 and exosomal SAA3 protein engages with Toll-like receptor 2 (TRL2) on macrophages, triggering an overproduction and impaired clearance of SAA3 proteins, resulting in aggregation of SAA3 monomers as rigid amyloid deposits in the extracellular space. The onset of amyloid fibers deposition alongside extra-cellular-matrix (ECM) proteins in the ischemic heart exacerbates the rigidity and stiffness of the scar, hindering the contractility of viable myocardium and overall impairing organ function. Using SAA3 and TLR2 deficient mouse models, we show that SAA3 delivered by MSC PDPN+ exosomes promotes post-MI amyloidosis. Inhibition of SAA3 aggregation via administration of a retro-inverso D-peptide, specifically designed to bind SAA3 monomers, prevents the deposition of SAA3 amyloid fibrils, positively modulates the scar formation, and improves heart function post-MI. Conclusion Overall, our findings provide mechanistic insights into post-MI amyloidosis and suggest that SAA3 may be an attractive target for effective scar reversal after ischemic injury and a potential target in multiple diseases characterized by a similar pattern of inflammation and amyloid deposition. NOVELTY AND SIGNIFICANCE What is known? Accumulation of rigid amyloid structures in the left ventricular wall impairs ventricle contractility.After myocardial infarction cardiac Mesenchymal Stromal Cells (MSC) acquire Podoplanin (PDPN) to better interact with immune cells.Amyloid structures can accumulate in the heart after chronic inflammatory conditions. What information does this article contribute? Whether accumulation of cumbersome amyloid structures in the ischemic scar impairs left ventricle contractility, and scar reversal after myocardial infarction (MI) has never been investigated.The pathophysiological relevance of PDPN acquirement by MSC and the functional role of their secreted exosomes in the context of post-MI cardiac remodeling has not been investigated.Amyloid structures are present in the scar after ischemia and are composed of macrophage-derived Serum Amyloid A (SAA) 3 monomers, although mechanisms of SAA3 overproduction is not established. SUMMARY OF NOVELTY AND SIGNIFICANCE Here, we report that amyloidosis, a secondary phenomenon of an already preexisting and prolonged chronic inflammatory condition, occurs after MI and that amyloid structures are composed of macrophage-derived SAA3 monomers. Frequently studied cardiac amyloidosis are caused by aggregation of immunoglobulin light chains, transthyretin, fibrinogen, and apolipoprotein in a healthy heart as a consequence of systemic chronic inflammation leading to congestive heart failure with various types of arrhythmias and tissue stiffness. Although chronic MI is considered a systemic inflammatory condition, studies regarding the possible accumulation of amyloidogenic proteins after MI and the mechanisms involved in that process are yet to be reported. Here, we show that SAA3 overproduction in macrophages is triggered in a Toll-like Receptor 2 (TLR2)-p38MAP Kinase-dependent manner by exosomal communication from a subset of activated MSC, which, in response to MI, express a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin. We provide the full mechanism of this phenomenon in murine models and confirm SAA3 amyloidosis in failing human heart samples. Moreover, we developed a retro-inverso D-peptide therapeutic approach, "DRI-R5S," specifically designed to bind SAA3 monomers and prevent post-MI aggregation and deposition of SAA3 amyloid fibrils without interfering with the innate immune response.
Collapse
|
11
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594404. [PMID: 38798483 PMCID: PMC11118280 DOI: 10.1101/2024.05.15.594404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
|
12
|
Sakuma H, Maruyama K, Aonuma T, Kobayashi Y, Hayasaka T, Kano K, Kawaguchi S, Nakajima KI, Kawabe JI, Hasebe N, Nakagawa N. Inducible deletion of microRNA activity in kidney mesenchymal cells exacerbates renal fibrosis. Sci Rep 2024; 14:10963. [PMID: 38745066 PMCID: PMC11094108 DOI: 10.1038/s41598-024-61560-y] [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: 09/14/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
MicroRNAs (miRNAs) are sequence-specific inhibitors of post-transcriptional gene expression. However, the physiological functions of these non-coding RNAs in renal interstitial mesenchymal cells remain unclear. To conclusively evaluate the role of miRNAs, we generated conditional knockout (cKO) mice with platelet-derived growth factor receptor-β (PDGFR-β)-specific inactivation of the key miRNA pathway gene Dicer. The cKO mice were subjected to unilateral ureteral ligation, and renal interstitial fibrosis was quantitatively evaluated using real-time polymerase chain reaction and immunofluorescence staining. Compared with control mice, cKO mice had exacerbated interstitial fibrosis exhibited by immunofluorescence staining and mRNA expression of PDGFR-β. A microarray analysis showed decreased expressions of miR-9-5p, miR-344g-3p, and miR-7074-3p in cKO mice compared with those in control mice, suggesting an association with the increased expression of PDGFR-β. An analysis of the signaling pathways showed that the major transcriptional changes in cKO mice were related to smooth muscle cell differentiation, regulation of DNA metabolic processes and the actin cytoskeleton, positive regulation of fibroblast proliferation and Ras protein signal transduction, and focal adhesion-PI3K/Akt/mTOR signaling pathways. Depletion of Dicer in mesenchymal cells may downregulate the signaling pathway related to miR-9-5p, miR-344g-3p, and miR-7074-3p, which can lead to the progression of chronic kidney disease. These findings highlight the possibility for future diagnostic or therapeutic developments for renal fibrosis using miR-9-5p, miR-344g-3p, and miR-7074-3p.
Collapse
Affiliation(s)
- Hirofumi Sakuma
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Keisuke Maruyama
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Tatsuya Aonuma
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Yuya Kobayashi
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Taiki Hayasaka
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Kohei Kano
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Satoshi Kawaguchi
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Naoyuki Hasebe
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan
| | - Naoki Nakagawa
- Division of Cardiology and Nephrology, Department of Internal Medicine, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Japan.
| |
Collapse
|
13
|
Tamiato A, Tombor LS, Fischer A, Muhly-Reinholz M, Vanicek LR, Toğru BN, Neitz J, Glaser SF, Merten M, Rodriguez Morales D, Kwon J, Klatt S, Schumacher B, Günther S, Abplanalp WT, John D, Fleming I, Wettschureck N, Dimmeler S, Luxán G. Age-Dependent RGS5 Loss in Pericytes Induces Cardiac Dysfunction and Fibrosis. Circ Res 2024; 134:1240-1255. [PMID: 38563133 PMCID: PMC11081481 DOI: 10.1161/circresaha.123.324183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Pericytes are capillary-associated mural cells involved in the maintenance and stability of the vascular network. Although aging is one of the main risk factors for cardiovascular disease, the consequences of aging on cardiac pericytes are unknown. METHODS In this study, we have combined single-nucleus RNA sequencing and histological analysis to determine the effects of aging on cardiac pericytes. Furthermore, we have conducted in vivo and in vitro analysis of RGS5 (regulator of G-protein signaling 5) loss of function and finally have performed pericytes-fibroblasts coculture studies to understand the effect of RGS5 deletion in pericytes on the neighboring fibroblasts. RESULTS Aging reduced the pericyte area and capillary coverage in the murine heart. Single-nucleus RNA sequencing analysis further revealed that the expression of Rgs5 was reduced in cardiac pericytes from aged mice. In vivo and in vitro studies showed that the deletion of RGS5 impaired cardiac function, induced fibrosis, and morphological changes in pericytes characterized by a profibrotic gene expression signature and the expression of different ECM (extracellular matrix) components and growth factors, for example, TGFB2 and PDGFB. Indeed, culturing fibroblasts with the supernatant of RGS5-deficient pericytes induced their activation as evidenced by the increased expression of αSMA (alpha smooth muscle actin) in a TGFβ (transforming growth factor beta)2-dependent mechanism. CONCLUSIONS Our results have identified RGS5 as a crucial regulator of pericyte function during cardiac aging. The deletion of RGS5 causes cardiac dysfunction and induces myocardial fibrosis, one of the hallmarks of cardiac aging.
Collapse
Affiliation(s)
- Anita Tamiato
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Lukas S. Tombor
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Ariane Fischer
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Marion Muhly-Reinholz
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Leah Rebecca Vanicek
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Büşra Nur Toğru
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Jessica Neitz
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Simone Franziska Glaser
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Maximilian Merten
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - David Rodriguez Morales
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Jeonghyeon Kwon
- Department of Pharmacology (J.K., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stephan Klatt
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- Institute for Vascular Signalling, Center of Molecular Medicine (S.K., I.F.), Goethe University Frankfurt, Germany
| | - Bianca Schumacher
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Stefan Günther
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
- Bioinformatics and Deep Sequencing Platform (S.G.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Wesley T. Abplanalp
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - David John
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Ingrid Fleming
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- Institute for Vascular Signalling, Center of Molecular Medicine (S.K., I.F.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Nina Wettschureck
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
- Department of Pharmacology (J.K., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Guillermo Luxán
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| |
Collapse
|
14
|
Chen X, Wang T, Chen L, Zhao Y, Deng Y, Shen W, Li L, Yin Z, Zhang C, Cai G, Zhang M, Chen X. Cross-species single-cell analysis uncovers the immunopathological mechanisms associated with IgA nephropathy progression. JCI Insight 2024; 9:e173651. [PMID: 38716725 PMCID: PMC11141938 DOI: 10.1172/jci.insight.173651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/19/2024] [Indexed: 05/12/2024] Open
Abstract
IgA nephropathy (IgAN) represents the main cause of renal failure, while the precise pathogenetic mechanisms have not been fully determined. Herein, we conducted a cross-species single-cell survey on human IgAN and mouse and rat IgAN models to explore the pathogenic programs. Cross-species single-cell RNA sequencing (scRNA-Seq) revealed that the IgAN mesangial cells (MCs) expressed high levels of inflammatory signatures CXCL12, CCL2, CSF1, and IL-34 and specifically interacted with IgAN macrophages via the CXCL12/CXCR4, CSF1/IL-34/CSF1 receptor, and integrin subunit alpha X/integrin subunit alpha M/complement C3 (C3) axes. IgAN macrophages expressed high levels of CXCR4, PDGFB, triggering receptor expressed on myeloid cells 2, TNF, and C3, and the trajectory analysis suggested that these cells derived from the differentiation of infiltrating blood monocytes. Additionally, protein profiling of 21 progression and 28 nonprogression IgAN samples revealed that proteins CXCL12, C3, mannose receptor C-type 1, and CD163 were negatively correlated with estimated glomerular filtration rate (eGFR) value and poor prognosis (30% eGFR as composite end point). Last, a functional experiment revealed that specific blockade of the Cxcl12/Cxcr4 pathway substantially attenuated the glomerulus and tubule inflammatory injury, fibrosis, and renal function decline in the mouse IgAN model. This study provides insights into IgAN progression and may aid in the refinement of IgAN diagnosis and the optimization of treatment strategies.
Collapse
Affiliation(s)
- Xizhao Chen
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Tiantian Wang
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Lei Chen
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yinghua Zhao
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yiyao Deng
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Wanjun Shen
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Lin Li
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Zhong Yin
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Chaoran Zhang
- Department of Stomatology, The First Medical Center of People’s Liberation Army General Hospital, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Min Zhang
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
15
|
Firat EAM, Buhl EM, Bouteldja N, Smeets B, Eriksson U, Boor P, Klinkhammer BM. PDGF-D Is Dispensable for the Development and Progression of Murine Alport Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:641-655. [PMID: 38309427 DOI: 10.1016/j.ajpath.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/05/2024]
Abstract
Alport syndrome is an inherited kidney disease, which can lead to glomerulosclerosis and fibrosis, as well as end-stage kidney disease in children and adults. Platelet-derived growth factor-D (PDGF-D) mediates glomerulosclerosis and interstitial fibrosis in various models of kidney disease, prompting investigation of its role in a murine model of Alport syndrome. In vitro, PDGF-D induced proliferation and profibrotic activation of conditionally immortalized human parietal epithelial cells. In Col4a3-/- mice, a model of Alport syndrome, PDGF-D mRNA and protein were significantly up-regulated compared with non-diseased wild-type mice. To analyze the therapeutic potential of PDGF-D inhibition, Col4a3-/- mice were treated with a PDGF-D neutralizing antibody. Surprisingly, PDGF-D antibody treatment had no effect on renal function, glomerulosclerosis, fibrosis, or other indices of kidney injury compared with control treatment with unspecific IgG. To characterize the role of PDGF-D in disease development, Col4a3-/- mice with a constitutive genetic deletion of Pdgfd were generated and analyzed. No difference in pathologic features or kidney function was observed in Col4a3-/-Pdgfd-/- mice compared with Col4a3-/-Pdgfd+/+ littermates, confirming the antibody treatment data. Mechanistically, lack of proteolytic PDGF-D activation in Col4a3-/- mice might explain the lack of effects in vivo. In conclusion, despite its established role in kidney fibrosis, PDGF-D, without further activation, does not mediate the development and progression of Alport syndrome in mice.
Collapse
Affiliation(s)
| | - Eva Miriam Buhl
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Nassim Bouteldja
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany; Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
| | | |
Collapse
|
16
|
Mebrahtu A, Aniander G, Mega A, Moradi Barzadd M, Berndt Thalén N, Gudmundsdotter L, Backström Rydin E, Sandegren A, Frejd FY, Rockberg J. Co-culture platform for tuning of cancer receptor density allows for evaluation of bispecific immune cell engagers. N Biotechnol 2024; 79:120-126. [PMID: 38159596 DOI: 10.1016/j.nbt.2023.12.012] [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: 02/09/2023] [Revised: 11/30/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Cancer immunotherapy, where a patient's immune system is harnessed to eradicate cancer cells selectively, is a leading strategy for cancer treatment. However, successes with immune checkpoint inhibitors (ICI) are hampered by reported systemic and organ-specific toxicities and by two-thirds of the patients being non-responders or subsequently acquiring resistance to approved ICIs. Hence substantial efforts are invested in discovering novel targeted immunotherapies aimed at reduced side-effects and improved potency. One way is utilizing the dual targeting feature of bispecific antibodies, which have made them increasingly popular for cancer immunotherapy. Easy and predictive screening methods for activation ranking of candidate drugs in tumor contra non-tumor environments are however lacking. Herein, we present a cell-based assay mimicking the tumor microenvironment by co-culturing B cells with engineered human embryonic kidney 293 T cells (HEK293T), presenting a controllable density of platelet-derived growth factor receptor β (PDGFRβ). A target density panel with three different surface protein levels on HEK293T cells was established by genetic constructs carrying regulatory elements limiting RNA translation of PDGFRβ. We employed a bispecific antibody-affibody construct called an AffiMab capable of binding PDGFRβ on cancer cells and CD40 expressed by B cells as a model. Specific activation of CD40-mediated signaling of immune cells was demonstrated with the two highest receptor-expressing cell lines, Level 2/3 and Level 4, while low-to-none in the low-expressing cell lines. The concept of receptor tuning and the presented co-culture protocol may be of general utility for assessing and developing novel bi-specific antibodies for immuno-oncology applications.
Collapse
Affiliation(s)
- Aman Mebrahtu
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Gustav Aniander
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Alessandro Mega
- Affibody Medical AB, Scheeles väg 2, SE-171 65 Solna, Sweden
| | - Mona Moradi Barzadd
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Niklas Berndt Thalén
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | | | | | - Anna Sandegren
- Affibody Medical AB, Scheeles väg 2, SE-171 65 Solna, Sweden
| | - Fredrik Y Frejd
- Affibody Medical AB, Scheeles väg 2, SE-171 65 Solna, Sweden
| | - Johan Rockberg
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden.
| |
Collapse
|
17
|
Zhang X, Li L, Tan H, Hong X, Yuan Q, Hou FF, Zhou L, Liu Y. Klotho-derived peptide 1 inhibits cellular senescence in the fibrotic kidney by restoring Klotho expression via posttranscriptional regulation. Theranostics 2024; 14:420-435. [PMID: 38164143 PMCID: PMC10750200 DOI: 10.7150/thno.89105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
Background: Klotho deficiency is a common feature of premature aging and chronic kidney disease (CKD). As such, restoring Klotho expression could be a logic strategy for protecting against various nephropathies. In this study, we demonstrate that KP1, a Klotho-derived peptide, inhibits cellular senescence by restoring endogenous Klotho expression. Methods: The effects of KP1 on cellular senescence and Klotho expression were assessed in mouse models of CKD. RNA-sequencing was employed to identify the microRNA involved in regulating Klotho by KP1. Gain- or loss-of-function approaches were used to assess the role of miR-223-3p and IncRNA-TUG1 in regulating Klotho and cellular senescence. Results: KP1 inhibited senescence markers p21, p16 and γ-H2AX in tubular epithelial cells of diseased kidneys, which was associated with its restoration of Klotho expression at the posttranscriptional level. Profiling of kidney microRNAs by RNA sequencing identified miR-223-3p that bound to Klotho mRNA and inhibited its protein expression. Overexpression of miR-223-3p inhibited Klotho and induced p21, p16 and γ-H2AX, which were negated by KP1. Conversely, inhibition of miR-223-3p restored Klotho expression, inhibited cellular senescence. Furthermore, miR-223-3p interacted with lncRNA-TUG1 and inhibited its expression. Knockdown of lncRNA-TUG1 increased miR-223-3p, aggravated Klotho loss and worsened cellular senescence, whereas KP1 mitigated all these changes. Conclusion: These studies demonstrate that KP1 inhibits cellular senescence and induces Klotho expression via posttranscriptional regulation mediated by miR-223-3p and lncRNA-TUG1. By restoring endogenous Klotho, KP1 elicits a broad spectrum of protective actions and could serve as a promising therapeutic agent for fibrotic kidney disorders.
Collapse
Affiliation(s)
- Xiaoyao Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huishi Tan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Hong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Yuan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
18
|
Yamashita N, Kramann R. Mechanisms of kidney fibrosis and routes towards therapy. Trends Endocrinol Metab 2024; 35:31-48. [PMID: 37775469 DOI: 10.1016/j.tem.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023]
Abstract
Kidney fibrosis is the final common pathway of virtually all chronic kidney diseases (CKDs) and is therefore considered to be a promising therapeutic target for these conditions. However, despite great progress in recent years, no targeted antifibrotic therapies for the kidney have been approved, likely because the complex mechanisms that initiate and drive fibrosis are not yet completely understood. Recent single-cell genomic approaches have allowed novel insights into kidney fibrosis mechanisms in mouse and human, particularly the heterogeneity and differentiation processes of myofibroblasts, the role of injured epithelial cells and immune cells, and their crosstalk mechanisms. In this review we summarize the key mechanisms that drive kidney fibrosis, including recent advances in understanding the mechanisms, as well as potential routes for developing novel targeted antifibrotic therapeutics.
Collapse
Affiliation(s)
- Noriyuki Yamashita
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Packer M. Mechanisms of enhanced renal and hepatic erythropoietin synthesis by sodium-glucose cotransporter 2 inhibitors. Eur Heart J 2023; 44:5027-5035. [PMID: 37086098 PMCID: PMC10733737 DOI: 10.1093/eurheartj/ehad235] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/06/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of major heart failure events, an action that is statistically linked to enhanced erythropoiesis, suggesting that stimulation of erythropoietin and cardioprotection are related to a shared mechanism. Four hypotheses have been proposed to explain how these drugs increase erythropoietin production: (i) renal cortical reoxygenation with rejuvenation of erythropoietin-producing cells; (ii) counterregulatory distal sodium reabsorption leading to increased tubular workload and oxygen consumption, and thus, to localized hypoxia; (iii) increased iron mobilization as a stimulus of hypoxia-inducible factor-2α (HIF-2α)-mediated erythropoietin synthesis; and (iv) direct HIF-2α activation and enhanced erythropoietin gene transcription due to increased sirtuin-1 (SIRT1) signaling. The first two hypotheses assume that the source of increased erythropoietin is the interstitial fibroblast-like cells in the deep renal cortex. However, SGLT2 inhibitors do not alter regional tissue oxygen tension in the non-diabetic kidney, and renal erythropoietin synthesis is markedly impaired in patients with anemia due to chronic kidney disease, and yet, SGLT2 inhibitors produce an unattenuated erythrocytic response in these patients. This observation raises the possibility that the liver contributes to the production of erythropoietin during SGLT2 inhibition. Hypoxia-inducible factor-2α and erythropoietin are coexpressed not only in the kidney but also in hepatocytes; the liver is a major site of production when erythropoietin stimulation is maintained for prolonged periods. The ability of SGLT2 inhibitors to improve iron mobilization by derepressing hepcidin and ferritin would be expected to increase cytosolic ferrous iron, which might stimulate HIF-2α expression in both the kidney and liver through the action of iron regulatory protein 1. Alternatively, the established ability of SGLT2 inhibitors to enhance SIRT1 might be the mechanism of enhanced erythropoietin production with these drugs. In hepatic cell lines, SIRT1 can directly activate HIF-2α by deacetylation, and additionally, through an effect of SIRT in the liver, peroxisome proliferator-activated receptor-γ coactivator-1α binds to hepatic nuclear factor 4 to promote transcription of the erythropoietin gene and synthesis of erythropoietin. Since SIRT1 up-regulation exerts direct cytoprotective effects on the heart and stimulates erythropoietin, it is well-positioned to represent the shared mechanism that links erythropoiesis to cardioprotection during SGLT2 inhibition.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, 621 North Hall Street, Dallas, TX 75226, USA
- Imperial College, London, UK
| |
Collapse
|
20
|
Wang S, Broder A, Shao D, Kesarwani V, Boderman B, Aguilan J, Sidoli S, Suzuki M, Greally JM, Saenger YM, Rovin BH, Michelle Kahlenberg J. Urine Proteomics Link Complement Activation with Interstitial Fibrosis/Tubular Atrophy in Lupus Nephritis Patients. Semin Arthritis Rheum 2023; 63:152263. [PMID: 37802003 PMCID: PMC10783434 DOI: 10.1016/j.semarthrit.2023.152263] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Intrarenal complement activation has been implicated in the pathogenesis of tubulointerstitial fibrosis in lupus nephritis (LN) based on prior animal studies. The assembly of the membrane attack complex (MAC) by complement C5b to C9 on the cell membrane leads to cytotoxic pores and cell lysis, while CD59 inhibits MAC formation by preventing C9 from joining the complex. We hypothesize that complement activation and imbalance between complement activation and inhibition, as defined by increased production of individual complement components and uncontrolled MAC activation relative to CD59 inhibition, are associated with interstitial fibrosis and tubular atrophy (IFTA) in LN and correlate with the key mediators of kidney fibrosis- transforming growth factor receptors beta (TGFRβ), platelet-derived growth factor beta (PDGFβ) and platelet-derived growth factor receptor beta (PDGFRβ). METHODS We included urine samples from 46 adults and pediatric biopsy-proven lupus nephritis patients who underwent clinically indicated kidney biopsies between 2010 and 2019. We compared individual urinary complement components and the urinary C9-to-CD59 ratio between LN patients with moderate/severe IFTA and none/mild IFTA. IFTA was defined as none/mild (<25% of interstitium affected) versus moderate/severe (≥ 25% of interstitium affected). Proteomics analysis was performed using mass spectrometry (Orbitrap Fusion Lumos, Thermo Scientific) and processed by the Proteome Discoverer. Urinary complement proteins enriched in LN patients with moderate/severe IFTA were correlated with serum creatinine, TGFβR1, TGFβR2, PDGFβ, and PDGFRβ. RESULTS Of the 46 LN patients included in the study, 41 (89.1%) were women, 20 (43.5%) self-identified as Hispanic or Latino, and 26 (56.5%) self-identified as Black or African American. Ten of the 46 (21.7%) LN patients had moderate/severe IFTA on kidney biopsy. LN patients with moderate/severe IFTA had an increased urinary C9-to-CD59 ratio [median 0.91 (0.83-1.05) vs 0.81 (0.76-0.91), p=0.01]. Urinary C3 and CFI levels in LN patients with moderate/severe IFTA were higher compared to those with none/mild IFTA [C3 median (IQR) 24.4(23.5-25.5) vs. 20.2 (18.5-22.2), p= 0.02], [CFI medium (IQR) 28.8 (21.8-30.6) vs. 20.4 (18.5-22.9), p=0.01]. Complement C9, CD59, C3 and CFI correlated with TGFβR1, PDGFβ, and PDGFRβ, while C9, CD59 and C3 correlated with TGFβR2. CONCLUSION This study is one of the first to compare the urinary complement profile in LN patients with moderate/severe IFTA and none/mild IFTA in human tissues. This study identified C3, CFI, and C9-to-CD59 ratio as potential markers of tubulointerstitial fibrosis in LN.
Collapse
Affiliation(s)
- Shudan Wang
- Division of Rheumatology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - Anna Broder
- Division of Rheumatology, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Daming Shao
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - Vartika Kesarwani
- Department of Medicine, University of Wisconsin Hospital and Clinics, WI, USA
| | - Brianna Boderman
- Department of Medicine, University of Connecticut School of Medicine, CT, USA
| | - Jennifer Aguilan
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Masako Suzuki
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY USA
| | - John M Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY USA
| | - Yvonne M Saenger
- Department of Oncology and Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| | - Brad H Rovin
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | |
Collapse
|
21
|
Jo YS, Kim KJ, Rhee CK, Kim YH. Effects of comorbid chronic kidney disease on mortality in idiopathic pulmonary fibrosis patients and influence of pirfenidone. Sci Rep 2023; 13:19238. [PMID: 37935732 PMCID: PMC10630477 DOI: 10.1038/s41598-023-46506-0] [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: 08/16/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
Chronic kidney disease (CKD) is a comorbidity in idiopathic pulmonary fibrosis (IPF), and managing IPF with CKD is challenging due to limited options for antifibrotic therapy. The aim of this study was to examine the prevalence of CKD and prescription status of pirfenidone in IPF patients and to analyze its impact on mortality. Data from the Korean National Health Insurance Service (NHIS) database between October 2015 and September 2021 were used. IPF and CKD were defined based on both International Classification of Diseases 10th Revision (ICD-10) codes and Rare Intractable Disease (RID) codes. The risk of mortality was assessed based on accompanying CKD with or without antifibrotic therapy. Among 5038 patients with IPF, 8.4% had comorbid CKD and 83.3% with CKD did not receive renal replacement therapy (RRT). Patients with IPF and CKD were older, predominantly male, and had more frequent comorbidities such as cardiovascular disease and diabetes mellitus than subjects without CKD. Pirfenidone was prescribed to 105 (24.6%) of 426 CKD patients, and 89.5% of them did not receive RRT. Pirfenidone was also prescribed to 775 (16.8%) of 4612 IPF patients without CKD. Significant difference was not found in all-cause mortality between the IPF patients with or without CKD regardless of pirfenidone treatment. The use of antifibrotics in IPF patients with CKD is limited due to CKD severity; however, evidence is lacking. Mortality did not increase with accompanying CKD regardless of antifibrotic use. Further research on IPF and CKD is needed.
Collapse
Affiliation(s)
- Yong Suk Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyung Joo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Hyun Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Naas S, Schiffer M, Schödel J. Hypoxia and renal fibrosis. Am J Physiol Cell Physiol 2023; 325:C999-C1016. [PMID: 37661918 DOI: 10.1152/ajpcell.00201.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Renal fibrosis is the final stage of most progressive kidney diseases. Chronic kidney disease (CKD) is associated with high comorbidity and mortality. Thus, preventing fibrosis and thereby preserving kidney function increases the quality of life and prolongs the survival of patients with CKD. Many processes such as inflammation or metabolic stress modulate the progression of kidney fibrosis. Hypoxia has also been implicated in the pathogenesis of renal fibrosis, and oxygen sensing in the kidney is of outstanding importance for the body. The dysregulation of oxygen sensing in the diseased kidney is best exemplified by the loss of stimulation of erythropoietin production from interstitial cells in the fibrotic kidney despite anemia. Furthermore, hypoxia is present in acute or chronic kidney diseases and may affect all cell types present in the kidney including tubular and glomerular cells as well as resident immune cells. Pro- and antifibrotic effects of the transcription factors hypoxia-inducible factors 1 and 2 have been described in a plethora of animal models of acute and chronic kidney diseases, but recent advances in sequencing technologies now allow for novel and deeper insights into the role of hypoxia and its cell type-specific effects on the progression of renal fibrosis, especially in humans. Here, we review existing literature on how hypoxia impacts the development and progression of renal fibrosis.
Collapse
Affiliation(s)
- Stephanie Naas
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
23
|
Yu S, Kalinin AA, Paraskevopoulou MD, Maruggi M, Cheng J, Tang J, Icke I, Luo Y, Wei Q, Scheibe D, Hunter J, Singh S, Nguyen D, Carpenter AE, Horman SR. Integrating inflammatory biomarker analysis and artificial-intelligence-enabled image-based profiling to identify drug targets for intestinal fibrosis. Cell Chem Biol 2023; 30:1169-1182.e8. [PMID: 37437569 PMCID: PMC10529501 DOI: 10.1016/j.chembiol.2023.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/11/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
Intestinal fibrosis, often caused by inflammatory bowel disease, can lead to intestinal stenosis and obstruction, but there are no approved treatments. Drug discovery has been hindered by the lack of screenable cellular phenotypes. To address this, we used a scalable image-based morphology assay called Cell Painting, augmented with machine learning algorithms, to identify small molecules that could reverse the activated fibrotic phenotype of intestinal myofibroblasts. We then conducted a high-throughput small molecule chemogenomics screen of approximately 5,000 compounds with known targets or mechanisms, which have achieved clinical stage or approval by the FDA. By integrating morphological analyses and AI using pathologically relevant cells and disease-relevant stimuli, we identified several compounds and target classes that are potentially able to treat intestinal fibrosis. This phenotypic screening platform offers significant improvements over conventional methods for identifying a wide range of drug targets.
Collapse
Affiliation(s)
- Shan Yu
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| | | | | | - Marco Maruggi
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Jie Cheng
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Jie Tang
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Ilknur Icke
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Yi Luo
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Qun Wei
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Dan Scheibe
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Joel Hunter
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Shantanu Singh
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Deborah Nguyen
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | | | - Shane R Horman
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| |
Collapse
|
24
|
Nangaku M, Kitching AR, Boor P, Fornoni A, Floege J, Coates PT, Himmelfarb J, Lennon R, Anders HJ, Humphreys BD, Caskey FJ, Fogo AB. International Society of Nephrology first consensus guidance for preclinical animal studies in translational nephrology. Kidney Int 2023; 104:36-45. [PMID: 37001557 DOI: 10.1016/j.kint.2023.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
Preclinical tests in animal models are key steps in early drug development. Consequently, the International Society of Nephrology held a consensus meeting that connected experts in the global kidney community in order to provide guidance on optimal management of translational animal studies for the development of new drugs to treat kidney disease, entitled "TRANSFORM; TRAnslational Nephrology Science FOR new Medications." The meeting covered various themes, including the following: (i) selection of disease model; (ii) pharmacokinetics; (iii) interventions in late preclinical models; (iv) choice of animal; (v) statistical power; (vi) organoids and organ-on-a-chip models; and (vii) reporting of results. This guidance is the first to be provided on the optimal conduct of translational animal studies for the development of new drugs to treat kidney disease. These recommendations are designed to accelerate development of new drugs for efficacious treatment of kidney diseases, and to improve the prognosis and quality of life of patients with a variety of kidney diseases.
Collapse
Affiliation(s)
- Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Peter Boor
- Institute of Pathology, Rheinisch Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jürgen Floege
- Division of Nephrology and Rheumatology, Rheinisch Westfälische Technische Hochschule (RWTH) University Hospital, Aachen, Germany
| | - P Toby Coates
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, South Australia, Australia; University of Adelaide, Medical Specialties, Adelaide, Australia; South Australian Medical Research Institute, Adelaide, South Australia, Australia
| | - Jonathan Himmelfarb
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Hans-Joachim Anders
- Department of Medicine IV, Hospital of Ludwig Maximilian University of Munich, Munich, Germany
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Fergus J Caskey
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
25
|
Altrieth AL, O’Keefe KJ, Gellatly VA, Tavarez JR, Feminella SM, Moskwa NL, Cordi CV, Turrieta JC, Nelson DA, Larsen M. Identifying fibrogenic cells following salivary gland obstructive injury. Front Cell Dev Biol 2023; 11:1190386. [PMID: 37287453 PMCID: PMC10242138 DOI: 10.3389/fcell.2023.1190386] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/11/2023] [Indexed: 06/09/2023] Open
Abstract
Fibrosis results from excess extracellular matrix accumulation, which alters normal tissue architecture and impedes function. In the salivary gland, fibrosis can be induced by irradiation treatment for cancer therapy, Sjögren's Disease, and other causes; however, it is unclear which stromal cells and signals participate in injury responses and disease progression. As hedgehog signaling has been implicated in fibrosis of the salivary gland and other organs, we examined contributions of the hedgehog effector, Gli1, to fibrotic responses in salivary glands. To experimentally induce a fibrotic response in female murine submandibular salivary glands, we performed ductal ligation surgery. We detected a progressive fibrotic response where both extracellular matrix accumulation and actively remodeled collagen significantly increased at 14 days post-ligation. Macrophages, which participate in extracellular matrix remodeling, and Gli1+ and PDGFRα+ stromal cells, which may deposit extracellular matrix, both increased with injury. Using single-cell RNA-sequencing, Gli1 + cells were not found in discrete clusters at embryonic day 16 but were found in clusters expressing the stromal genes Pdgfra and/or Pdgfrb. In adult mice, Gli1+ cells were similarly heterogeneous but more cells co-expressed PDGFRα and PDGFRβ. Using Gli1-CreERT2; ROSA26tdTomato lineage-tracing mice, we found that Gli1-derived cells expand with ductal ligation injury. Although some of the Gli1 lineage-traced tdTomato+ cells expressed vimentin and PDGFRβ following injury, there was no increase in the classic myofibroblast marker, smooth muscle alpha-actin. Additionally, there was little change in extracellular matrix area, remodeled collagen area, PDGFRα, PDGFRβ, endothelial cells, neurons, or macrophages in Gli1 null salivary glands following injury when compared with controls, suggesting that Gli1 signaling and Gli1+ cells have only a minor contribution to mechanical injury-induced fibrotic changes in the salivary gland. We used scRNA-seq to examine cell populations that expand with ligation and/or showed increased expression of matrisome genes. Some Pdgfra + /Pdgfrb + stromal cell subpopulations expanded in response to ligation, with two stromal cell subpopulations showing increased expression of Col1a1 and a greater diversity of matrisome genes, consistent with these cells being fibrogenic. However, only a few cells in these subpopulations expressed Gli1, consistent with a minor contribution of these cells to extracellular matrix production. Defining the signaling pathways driving fibrotic responses in stromal cell sub-types could reveal future therapeutic targets.
Collapse
Affiliation(s)
- Amber L. Altrieth
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Kevin J. O’Keefe
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Victoria A. Gellatly
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Joey R. Tavarez
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Sage M. Feminella
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
| | - Nicholas L. Moskwa
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Carmalena V. Cordi
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
| | - Judy C. Turrieta
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
| | - Deirdre A. Nelson
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
| | - Melinda Larsen
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY, United States
- Molecular, Cellular, Developmental and Neural Biology Graduate Program, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
26
|
Jia T, Xu T, Smeets B, Buhl EM, Moeller MJ, Floege J, Klinkhammer BM, Boor P. The Role of Platelet-Derived Growth Factor in Focal Segmental Glomerulosclerosis. J Am Soc Nephrol 2023; 34:241-257. [PMID: 36351762 PMCID: PMC10103089 DOI: 10.1681/asn.2022040491] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND FSGS is the final common pathway to nephron loss in most forms of severe or progressive glomerular injury. Although podocyte injury initiates FSGS, parietal epithelial cells (PECs) are the main effectors. Because PDGF takes part in fibrotic processes, we hypothesized that the ligand PDGF-B and its receptor PDGFR- β participate in the origin and progression of FSGS. METHODS We challenged Thy1.1 transgenic mice, which express Thy1.1 in the podocytes, with anti-Thy1.1 antibody to study the progression of FSGS. We investigated the role of PDGF in FSGS using challenged Thy1.1 mice, 5/6 nephrectomized mice, Col4 -/- (Alport) mice, patient kidney biopsies, and primary murine PECs, and challenged Thy1.1 mice treated with neutralizing anti-PDGF-B antibody therapy. RESULTS The unchallenged Thy1.1 mice developed only mild spontaneous FSGS, whereas challenged mice developed progressive FSGS accompanied by a decline in kidney function. PEC activation, proliferation, and profibrotic phenotypic switch drove the FSGS. During disease, PDGF-B was upregulated in podocytes, whereas PDGFR- β was upregulated in PECs from both mice and patients with FSGS. Short- and long-term treatment with PDGF-B neutralizing antibody improved kidney function and reduced FSGS, PEC proliferation, and profibrotic activation. In vitro , stimulation of primary murine PECs with PDGF-B recapitulated in vivo findings with PEC activation and proliferation, which was inhibited by PDGF-B antibody or imatinib. CONCLUSION PDGF-B-PDGFR- β molecular crosstalk between podocytes and PECs drives glomerulosclerosis and the progression of FSGS. PODCAST This article contains a podcast at.
Collapse
Affiliation(s)
- Ting Jia
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tong Xu
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Eva Miriam Buhl
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Marcus Johannes Moeller
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
- Heisenberg Chair for Preventive and Translational Nephrology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Barbara Mara Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
27
|
He A, Sarwar A, Thole LML, Siegle J, Sattler A, Ashraf MI, Proß V, Stahl C, Dornieden T, Bergmann Y, Ritschl PV, Ebner S, Hublitz KW, Stamatiades EG, Bülow RD, Boor P, Kotsch K. Renal inflamm-aging provokes intra-graft inflammation following experimental kidney transplantation. Am J Transplant 2022; 22:2529-2547. [PMID: 35851547 DOI: 10.1111/ajt.17154] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/14/2022] [Accepted: 07/14/2022] [Indexed: 01/25/2023]
Abstract
Donor age is a major risk factor for allograft outcome in kidney transplantation. The underlying cellular mechanisms and the recipient's immune response within an aged allograft have yet not been analyzed. A comprehensive immunophenotyping of naïve and transplanted young versus aged kidneys revealed that naïve aged murine kidneys harbor significantly higher frequencies of effector/memory T cells, whereas regulatory T cells were reduced. Aged kidney-derived CD8+ T cells produced more IFNγ than their young counterparts. Senescent renal CD8+ T and NK cells upregulated the cytotoxicity receptor NKG2D and the enrichment of memory-like CD49a+ CXCR6+ NK cells was documented in aged naïve kidneys. In the C57BL/6 to BALB/c kidney transplantation model, recipient-derived T cells infiltrating an aged graft produced significantly more IFNγ, granzyme B and perforin on day 7 post-transplantation, indicating an enhanced inflammatory, cytotoxic response towards the graft. Pre-treatment of aged kidney donors with the senolytic drug ABT-263 changed the recipient-derived effector molecule profile to significantly reduced levels of IFNγ and IL-10 compared to controls. Graft function after ABT-263 pre-treatment was significantly improved 28 days post kidney transplantation. In conclusion, renal senescence also occurs at the immunological level (inflamm-aging) and aged organs provoke an altered recipient-dominated immune response in the graft.
Collapse
Affiliation(s)
- An He
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Attia Sarwar
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Linda Marie Laura Thole
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Janine Siegle
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Arne Sattler
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Vanessa Proß
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Carolin Stahl
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Theresa Dornieden
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Yasmin Bergmann
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Paul Viktor Ritschl
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Susanne Ebner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Karolin Wiebke Hublitz
- Institute of Microbiology, Infectious Diseases and Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Efstathios Gregorios Stamatiades
- Institute of Microbiology, Infectious Diseases and Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Roman David Bülow
- Institute of Pathology & Department of Nephrology, University Clinic of RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology & Department of Nephrology, University Clinic of RWTH Aachen, Aachen, Germany
| | - Katja Kotsch
- Department of General- and Visceral Surgery, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
28
|
Wang Q, Deng J, Sun J, Zhang H, Liu D, Gao C, Qiu J, Liu W, Qu G, Wen D, Du J, Zhang A, Zeng L, Jiang J. PDGFR kinase inhibitor protects against septic death via regulation of BTLA. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1917-1928. [PMID: 35918604 PMCID: PMC9345782 DOI: 10.1007/s11427-021-2136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Sepsis, defined as life-threatening organ failure caused by a dysregulated host response to severe infection, is a major cause of death among intensive care unit patients. Therapies targeting on immunomodulatory is a new research field in sepsis treatment. B- and T-lymphocyte attenuator (BTLA) is an inhibitory costimulatory factor molecule of B and T lymphocytes. Studies have shown that elevated expression of BTLA in lymphocytes can reduce mortality in sepsis, but its regulatory compounds and the underlying mechanism remains to be elucidated. Here, we show that treatment with CP-673451 significantly decreases mortality of septic mouse. CP-673451 is a PDGFR kinase inhibitor which can promote the expression of BTLA, inhibit the release of chemokines such as CXCL13, and reduce first the chemotaxis of B cells to the peripheral blood and vital organs. CP-673451 also inhibits both the release of cytokines and chemokines such as IL-1β, IL-6, IL-10, TNF-α, CCL1, CCL2 and CCL7 and reduces both the chemotactic ability of T cells. This suggests that CP-673451 may prevent septic death by inhibiting lymphocyte chemotaxis and alleviating "cytokine storm". In conclusion, our study provides a new therapeutic target and an effective compound for sepsis treatment.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Jin Deng
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Jianhui Sun
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Huacai Zhang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Di Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Chu Gao
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Jinchao Qiu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Wenyi Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Guoxin Qu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Dalin Wen
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Juan Du
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Anqiang Zhang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Ling Zeng
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China.
| | - Jianxin Jiang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
29
|
Jang HS, Noh MR, Plumb T, Lee K, He JC, Ferrer FA, Padanilam BJ. Hepatic and proximal tubule angiotensinogen play distinct roles in kidney dysfunction, glomerular and tubular injury, and fibrosis progression. Am J Physiol Renal Physiol 2022; 323:F435-F446. [PMID: 35924445 PMCID: PMC9485008 DOI: 10.1152/ajprenal.00029.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/21/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Components of the renin-angiotensin system, including angiotensinogen (AGT), are critical contributors to chronic kidney disease (CKD) development and progression. However, the specific role of tissue-derived AGTs in CKD has not been fully understood. To define the contribution of liver versus kidney AGT in the CKD development, we performed 5/6 nephrectomy (Nx), an established CKD model, in wild-type (WT), proximal tubule (PT)- or liver-specific AGT knockout (KO) mice. Nx significantly elevated intrarenal AGT expression and elevated blood pressure (BP) in WT mice. The increase of intrarenal AGT protein was completely blocked in liver-specific AGT KO mice with BP reduction, suggesting a crucial role for liver AGT in BP regulation during CKD. Nx-induced glomerular and kidney injury and dysfunction, as well as fibrosis, were all attenuated to a greater extent in liver-specific AGT KO mice compared with PT-specific AGT KO and WT mice. However, the suppression of interstitial fibrosis in PT- and liver-specific AGT KO mouse kidneys was comparable. Our findings demonstrate that liver AGT acts as a critical contributor in driving glomerular and tubular injury, renal dysfunction, and fibrosis progression, whereas the role of PT AGT was limited to interstitial fibrosis progression in chronic renal insufficiency. Our results provide new insights for the development of tissue-targeted renin-angiotensin system intervention in the treatment of CKD.NEW & NOTEWORTHY Chronic kidney disease (CKD) is a major unmet medical need with no effective treatment. Current findings demonstrate that hepatic and proximal tubule angiotensinogen have distinct roles in tubular and glomerular injury, fibrogenesis, and renal dysfunction during CKD development. As renin-angiotensin system components, including angiotensinogen, are important targets for treating CKD in the clinic, the results from our study may be applied to developing better tissue-targeted treatment strategies for CKD and other fibroproliferative diseases.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mi Ra Noh
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Troy Plumb
- Division of Nephrology, Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fernando A Ferrer
- Department of Urology, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Babu J Padanilam
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
30
|
Mishra M, Nichols L, Dave AA, Pittman EH, Cheek JP, Caroland AJV, Lotwala P, Drummond J, Bridges CC. Molecular Mechanisms of Cellular Injury and Role of Toxic Heavy Metals in Chronic Kidney Disease. Int J Mol Sci 2022; 23:11105. [PMID: 36232403 PMCID: PMC9569673 DOI: 10.3390/ijms231911105] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive disease that affects millions of adults every year. Major risk factors include diabetes, hypertension, and obesity, which affect millions of adults worldwide. CKD is characterized by cellular injury followed by permanent loss of functional nephrons. As injured cells die and nephrons become sclerotic, remaining healthy nephrons attempt to compensate by undergoing various structural, molecular, and functional changes. While these changes are designed to maintain appropriate renal function, they may lead to additional cellular injury and progression of disease. As CKD progresses and filtration decreases, the ability to eliminate metabolic wastes and environmental toxicants declines. The inability to eliminate environmental toxicants such as arsenic, cadmium, and mercury may contribute to cellular injury and enhance the progression of CKD. The present review describes major molecular alterations that contribute to the pathogenesis of CKD and the effects of arsenic, cadmium, and mercury on the progression of CKD.
Collapse
Affiliation(s)
- Manish Mishra
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Larry Nichols
- Department of Pathology and Clinical Sciences Education, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Aditi A. Dave
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Elizabeth H Pittman
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - John P. Cheek
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Anasalea J. V. Caroland
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Purva Lotwala
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - James Drummond
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Christy C. Bridges
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
31
|
Hsu PH, Chen YH, Huang PI, Hwang PA. Skin proteomic profiling of irradiation-induced fibrosis and its modulation by low molecular weight fucoidan via tight junction pathway. Biomed Pharmacother 2022; 153:113417. [DOI: 10.1016/j.biopha.2022.113417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 11/02/2022] Open
|
32
|
Ruiz-Ortega M, Lamas S, Ortiz A. Antifibrotic Agents for the Management of CKD: A Review. Am J Kidney Dis 2022; 80:251-263. [PMID: 34999158 DOI: 10.1053/j.ajkd.2021.11.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 11/18/2021] [Indexed: 01/27/2023]
Abstract
Kidney fibrosis is a hallmark of chronic kidney disease (CKD) and a potential therapeutic target. However, there are conceptual and practical challenges to directly targeting kidney fibrosis. Whether fibrosis is mainly a cause or a consequence of CKD progression has been disputed. It is unclear whether specifically targeting fibrosis is feasible in clinical practice because most drugs that decrease fibrosis in preclinical models target additional and often multiple pathogenic pathways (eg, renin-angiotensin-aldosterone system blockade). Moreover, tools to assess whole-kidney fibrosis in routine clinical practice are lacking. Pirfenidone, a drug used for idiopathic pulmonary fibrosis, is undergoing a phase 2 trial for kidney fibrosis. Other drugs in use or being tested for idiopathic pulmonary fibrosis (eg, nintedanib, PRM-151, epigallocatechin gallate) are also potential candidates to treat kidney fibrosis. Novel therapeutic approaches may include antagomirs (eg, lademirsen) or drugs targeting interleukin 11 or NKD2 (WNT signaling pathway inhibitor). Reversing the dysfunctional tubular cell metabolism that leads to kidney fibrosis offers additional therapeutic opportunities. However, any future drug targeting fibrosis of the kidneys should demonstrate added benefit to a standard of care that combines renin-angiotensin system with mineralocorticoid receptor (eg, finerenone) blockade or with sodium/glucose cotransporter 2 inhibitors.
Collapse
Affiliation(s)
- Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid; Red de Investigación Renal, Madrid, Spain
| | - Santiago Lamas
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid; Red de Investigación Renal, Madrid, Spain; Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Alberto Ortiz
- Nephrology and Hypertension, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid; Red de Investigación Renal, Madrid, Spain.
| |
Collapse
|
33
|
Moeller MJ, Kramann R, Lammers T, Hoppe B, Latz E, Ludwig-Portugall I, Boor P, Floege J, Kurts C, Weiskirchen R, Ostendorf T. New Aspects of Kidney Fibrosis-From Mechanisms of Injury to Modulation of Disease. Front Med (Lausanne) 2022; 8:814497. [PMID: 35096904 PMCID: PMC8790098 DOI: 10.3389/fmed.2021.814497] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 02/02/2023] Open
Abstract
Organ fibrogenesis is characterized by a common pathophysiological final pathway independent of the underlying progressive disease of the respective organ. This makes it particularly suitable as a therapeutic target. The Transregional Collaborative Research Center “Organ Fibrosis: From Mechanisms of Injury to Modulation of Disease” (referred to as SFB/TRR57) was hosted from 2009 to 2021 by the Medical Faculties of RWTH Aachen University and the University of Bonn. This consortium had the ultimate goal of discovering new common but also different fibrosis pathways in the liver and kidneys. It finally successfully identified new mechanisms and established novel therapeutic approaches to interfere with hepatic and renal fibrosis. This review covers the consortium's key kidney-related findings, where three overarching questions were addressed: (i) What are new relevant mechanisms and signaling pathways triggering renal fibrosis? (ii) What are new immunological mechanisms, cells and molecules that contribute to renal fibrosis?, and finally (iii) How can renal fibrosis be modulated?
Collapse
Affiliation(s)
- Marcus J Moeller
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Heisenberg Chair for Preventive and Translational Nephrology, Aachen, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Faculty of Medicine, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Bernd Hoppe
- Division of Pediatric Nephrology and Kidney Transplantation, University Hospital of Bonn, Bonn, Germany.,German Hyperoxaluria Center, Pediatric Kidney Care Center, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital of Bonn, Bonn, Germany
| | - Isis Ludwig-Portugall
- Institute for Molecular Medicine and Experimental Immunology, University Hospital of Bonn, Bonn, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Kurts
- Institute for Molecular Medicine and Experimental Immunology, University Hospital of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital RWTH Aachen, Aachen, Germany
| | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
34
|
Li H, Sun F, Bai S, Chang G, Wu R, Wei Y, Wen X, Xi Y, Hao J, Zaid A. The DR1‑CSE/H 2S system inhibits renal fibrosis by downregulating the ERK1/2 signaling pathway in diabetic mice. Int J Mol Med 2022; 49:7. [PMID: 34779492 PMCID: PMC8651227 DOI: 10.3892/ijmm.2021.5062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Glomerular mesangial cell (MC) proliferation and extracellular matrix deposition are the main pathological changes in diabetic nephropathy. Hydrogen sulfide (H2S) inhibits the proliferation of MCs. Dopamine 1 receptors (DR1) are expressed in MCs and serve important physiological roles. However, it is unclear whether DR1 activation inhibits MC proliferation by increasing endogenous H2S. The present study found that the production of H2S and the expression of DR1 and cystathionine‑γ‑lyase (CSE) were decreased in the renal tissues of diabetic mice and high glucose (HG)‑induced MCs. SKF38393 (a DR1 agonist) increased the production of H2S and the expression of DR1 and CSE and NaHS (an exogenous H2S donor) only increased H2S production and CSE expression but not DR1 expression. HG increased the thickness of the glomerular basement membrane, cell viability and proliferation, the expression of cyclin D1, PCNA, collagen 1 and α‑smooth muscle actin and the activity of phosphorylated ERK1/2 and decreased the expression of P21 and MMP9. SKF38393 and NaHS reversed the effects of HG. PPG (a CSE inhibitor) abolished the beneficial effects of SKF38393. The beneficial effects of SKF38393 were similar to those of PD98059 (an ERK1/2 inhibitor). Taken together, the findings suggested that the DR1‑CSE/H2S pathway activation attenuated diabetic MC proliferation and extracellular matrix deposition by downregulating the ERK1/2 signaling pathway.
Collapse
MESH Headings
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- Animals
- Cell Line
- Cell Proliferation
- Collagen/metabolism
- Cystathionine gamma-Lyase/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Female
- Fibrosis
- Glucose/pharmacology
- Hydrogen Sulfide/metabolism
- Kidney/metabolism
- Kidney/pathology
- MAP Kinase Signaling System/physiology
- Male
- Mesangial Cells/drug effects
- Mesangial Cells/pathology
- Mice, Inbred C57BL
- Receptors, Dopamine D1/agonists
- Receptors, Dopamine D1/metabolism
- Mice
Collapse
Affiliation(s)
- Hongzhu Li
- Department of Pathophysiology, School of Medicine, Xiamen University, Xiamen, Fujian 361100, P.R. China
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Fengqi Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shuzhi Bai
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Guiquan Chang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Ren Wu
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yaxin Wei
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xin Wen
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yuxin Xi
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jinghui Hao
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Altaany Zaid
- Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan
| |
Collapse
|
35
|
Progression of Myeloproliferative Neoplasms (MPN): Diagnostic and Therapeutic Perspectives. Cells 2021; 10:cells10123551. [PMID: 34944059 PMCID: PMC8700229 DOI: 10.3390/cells10123551] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPN) are a heterogeneous group of hematologic malignancies, including essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), as well as post-PV-MF and post-ET-MF. Progression to more symptomatic disease, such as overt MF or acute leukemia, represents one of the major causes of morbidity and mortality. There are clinically evident but also subclinical types of MPN progression. Clinically evident progression includes evolution from ET to PV, ET to post-ET-MF, PV to post-PV-MF, or pre-PMF to overt PMF, and transformation of any of these subtypes to myelodysplastic neoplasms or acute leukemia. Thrombosis, major hemorrhage, severe infections, or increasing symptom burden (e.g., pruritus, night sweats) may herald progression. Subclinical types of progression may include increases in the extent of bone marrow fibrosis, increases of driver gene mutational allele burden, and clonal evolution. The underlying causes of MPN progression are diverse and can be attributed to genetic alterations and chronic inflammation. Particularly, bystander mutations in genes encoding epigenetic regulators or splicing factors were associated with progression. Finally, comorbidities such as systemic inflammation, cardiovascular diseases, and organ fibrosis may augment the risk of progression. The aim of this review was to discuss types and mechanisms of MPN progression and how their knowledge might improve risk stratification and therapeutic intervention. In view of these aspects, we discuss the potential benefits of early diagnosis using molecular and functional imaging and exploitable therapeutic strategies that may prevent progression, but also highlight current challenges and methodological pitfalls.
Collapse
|
36
|
Kwon HR, Kim JH, Woods JP, Olson LE. Skeletal stem cell fate defects caused by Pdgfrb activating mutation. Development 2021; 148:272709. [PMID: 34738614 DOI: 10.1242/dev.199607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/28/2021] [Indexed: 11/20/2022]
Abstract
Autosomal dominant PDGFRβ gain-of-function mutations in mice and humans cause a spectrum of wasting and overgrowth disorders afflicting the skeleton and other connective tissues, but the cellular origin of these disorders remains unknown. We demonstrate that skeletal stem cells (SSCs) isolated from mice with a gain-of-function D849V point mutation in PDGFRβ exhibit colony formation defects that parallel the wasting or overgrowth phenotypes of the mice. Single-cell RNA transcriptomics with SSC-derived polyclonal colonies demonstrates alterations in osteogenic and chondrogenic precursors caused by PDGFRβD849V. Mutant cells undergo poor osteogenesis in vitro with increased expression of Sox9 and other chondrogenic markers. Mice with PDGFRβD849V exhibit osteopenia. Increased STAT5 phosphorylation and overexpression of Igf1 and Socs2 in PDGFRβD849V cells suggests that overgrowth in mice involves PDGFRβD849V activating the STAT5-IGF1 axis locally in the skeleton. Our study establishes that PDGFRβD849V causes osteopenic skeletal phenotypes that are associated with intrinsic changes in SSCs, promoting chondrogenesis over osteogenesis.
Collapse
Affiliation(s)
- Hae Ryong Kwon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Jang H Kim
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - John P Woods
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lorin E Olson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
37
|
Geis L, Boudriot FF, Wagner C. Connexin mRNA distribution in adult mouse kidneys. Pflugers Arch 2021; 473:1737-1747. [PMID: 34365513 PMCID: PMC8528753 DOI: 10.1007/s00424-021-02608-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 11/25/2022]
Abstract
Kidneys are thought to express eight different connexin isoforms (i.e., Cx 26, 30, 32, 37, 40, 43, 45, and 46), which form either hemichannels or gap junctions serving to intercellular communication and functional synchronization. Proper function of connexins has already been shown to be crucial for regulation of renal hemodynamics and renin secretion, and there is also growing evidence for connexins to play a role in pathologic conditions such as renal fibrosis or diabetic nephropathy. Therefore, exact intrarenal localization of the different connexin isoforms gains particular interest. Until now intrarenal expression of connexins has mainly been examined by immunohistochemistry, which in part generated conflicting results depending on antibodies and fixation protocols used. In this work, we used fluorescent RNAscope as an alternative technical approach to localize renal connexin mRNAs in healthy mouse kidneys. Addition of RNAscope probes for cell type specific mRNAs was used to assign connexin mRNA signals to specific cell types. We hereby found Cx26 mRNA strongly expressed in proximal tubules, Cx30 mRNA was selectively detected in the urothelium, and Cx32 mRNA was found in proximal tubules and to a lesser extent also in collecting ducts. Cx37 mRNA was mainly associated with vascular endothelium, Cx40 mRNA was largely found in glomerular mesangial and less in vascular endothelial cells, Cx43 mRNA was sparsely expressed by interstitial cells of all kidney zones, and Cx45 mRNA was predominantly found in smooth muscle cell layers of both blood vessels and ureter as well as in mesangial and interstitial (fibroblastic) cells. Cx46 mRNA could not be detected. In summary our results essentially confirm previous data on connexin expression in the renal vasculature and in glomeruli. In addition, they demonstrate strong connexin gene expression in proximal tubules, and they suggest significant connexin expression in resident tubulointerstitial cells.
Collapse
Affiliation(s)
- Lisa Geis
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany.
| | | | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
38
|
Jiang M, Bai M, Xu S, Wang T, Lei J, Xu M, Huang S, Jia Z, Zhang A. Blocking AURKA with MK-5108 attenuates renal fibrosis in chronic kidney disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166227. [PMID: 34311081 DOI: 10.1016/j.bbadis.2021.166227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 12/25/2022]
Abstract
Renal fibrosis, a common feature of chronic kidney disease (CKD), is characterized by excessive deposition of extracellular matrix (ECM) leading to scar formation in the renal parenchyma. Active epithelial-mesenchymal communication (EMC), and the proliferation and activation of fibroblasts are implicated in the causation of renal fibrosis. Aurora-A kinase (AURKA) is a serine/threonine kinase required for the process of mitosis. Dysregulation of AURKA has been demonstrated in the context of various cancers. However, the role of AURKA in CKD-associated fibrosis has not been elucidated. MK-5108, a potent and highly selective AURKA inhibitor, was shown to exhibit anti-cancer activity in recent preclinical and clinical studies. In the present study, we investigated the role of MK-5108 in renal fibrosis employing animal and cell models. In vivo, AURKA was highly expressed in fibrotic kidneys of CKD patients and in mouse kidneys with unilateral ureteral obstruction (UUO). Post treatment with MK-5108 at the 3rd day after UUO remarkably alleviated renal fibrosis, possibly by inhibiting the proliferation and activation of fibroblasts and suppressing the phenotypic transition of renal cells. Moreover, the enhanced inflammatory factors in obstructive kidneys were also repressed. In vitro, MK-5108 treatment inhibited the pro-fibrotic response in renal cells induced by transforming growth factor-β1. Finally, overexpression of AURKA in renal fibroblasts promoted fibrotic response, while silencing AURKA showed anti-fibrotic effect, further confirming the pro-fibrotic role of AURKA. In this study, inhibition of AURKA by MK-5108 markedly attenuated renal fibrosis. MK-5108 is a potential therapeutic agent for treatment of renal fibrosis in CKD.
Collapse
Affiliation(s)
- Mingzhu Jiang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China; Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, China
| | - Shuang Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Ting Wang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Juan Lei
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Man Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China; Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China; Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, China.
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China; Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, China.
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China; Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, China.
| |
Collapse
|
39
|
Jia X, Zhang A, Li Z, Peng X, Tian X, Gao F. Activation of spinal PDGFRβ in microglia promotes neuronal autophagy via p38 MAPK pathway in morphine-tolerant rats. J Neurochem 2021; 158:373-390. [PMID: 33950542 DOI: 10.1111/jnc.15383] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022]
Abstract
The adverse side effects of opioids, especially antinociceptive tolerance, limit their clinical application. A recent study reported that platelet-derived growth factor receptor β (PDGFRβ) blockage selectively inhibited morphine tolerance. Autophagy has been reported to contribute to the cellular and behavioral responses to morphine. However, little is known about the relationship between PDGFRβ and autophagy in the mechanisms of morphine tolerance. In this study, rats were intrathecally administered with morphine twice daily for 7 days to induce antinociceptive tolerance, which was evaluated using a tail-flick latency test. By administration autophagy inhibitor 3-Methyladenine, PDGFRβ inhibitor imatinib, p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 hydrochloride and minocycline hydrochloride, western blot, immunofluorescence, and transmission electron microscopy techniques were used to elucidate the roles of PDGFRβ, autophagy, and related signaling pathways in morphine tolerance. This study demonstrated for the first time that spinal PDGFRβ in microglia promotes autophagy in gamma-aminobutyric acid (GABA) interneurons through activating p38 MAPK pathway during the development of morphine tolerance, which suggest a potential strategy for preventing the development of morphine tolerance clinically, thereby improving the use of opioids in pain management.
Collapse
Affiliation(s)
- Xiaoqian Jia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anqi Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Li
- Department of Anesthesiology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Apelt K, Bijkerk R, Lebrin F, Rabelink TJ. Imaging the Renal Microcirculation in Cell Therapy. Cells 2021; 10:cells10051087. [PMID: 34063200 PMCID: PMC8147454 DOI: 10.3390/cells10051087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Renal microvascular rarefaction plays a pivotal role in progressive kidney disease. Therefore, modalities to visualize the microcirculation of the kidney will increase our understanding of disease mechanisms and consequently may provide new approaches for evaluating cell-based therapy. At the moment, however, clinical practice is lacking non-invasive, safe, and efficient imaging modalities to monitor renal microvascular changes over time in patients suffering from renal disease. To emphasize the importance, we summarize current knowledge of the renal microcirculation and discussed the involvement in progressive kidney disease. Moreover, an overview of available imaging techniques to uncover renal microvascular morphology, function, and behavior is presented with the associated benefits and limitations. Ultimately, the necessity to assess and investigate renal disease based on in vivo readouts with a resolution up to capillary level may provide a paradigm shift for diagnosis and therapy in the field of nephrology.
Collapse
Affiliation(s)
- Katerina Apelt
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (K.A.); (R.B.); (F.L.)
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (K.A.); (R.B.); (F.L.)
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Franck Lebrin
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (K.A.); (R.B.); (F.L.)
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Physics for Medicine Paris, Inserm, CNRS, ESPCI Paris, Paris Sciences et Lettres University, 75005 Paris, France
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (K.A.); (R.B.); (F.L.)
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
41
|
Cimini M, Kishore R. Role of Podoplanin-Positive Cells in Cardiac Fibrosis and Angiogenesis After Ischemia. Front Physiol 2021; 12:667278. [PMID: 33912076 PMCID: PMC8072458 DOI: 10.3389/fphys.2021.667278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/15/2021] [Indexed: 01/05/2023] Open
Abstract
New insights into the cellular and extra-cellular composition of scar tissue after myocardial infarction (MI) have been identified. Recently, a heterogeneous podoplanin-expressing cell population has been associated with fibrogenic and inflammatory responses and lymphatic vessel growth during scar formation. Podoplanin is a mucin-like transmembrane glycoprotein that plays an important role in heart development, cell motility, tumorigenesis, and metastasis. In the adult mouse heart, podoplanin is expressed only by cardiac lymphatic endothelial cells; after MI, it is acquired with an unexpected heterogeneity by PDGFRα-, PDGFRβ-, and CD34-positive cells. Podoplanin may therefore represent a sign of activation of a cohort of progenitor cells during different phases of post-ischemic myocardial wound repair. Podoplanin binds to C-type lectin-like receptor 2 (CLEC-2) which is exclusively expressed by platelets and a variety of immune cells. CLEC-2 is upregulated in CD11bhigh cells, including monocytes and macrophages, following inflammatory stimuli. We recently published that inhibition of the interaction between podoplanin-expressing cells and podoplanin-binding cells using podoplanin-neutralizing antibodies reduces but does not fully suppress inflammation post-MI while improving heart function and scar composition after ischemic injury. These data support an emerging and alternative mechanism of interactome in the heart that, when neutralized, leads to altered inflammatory response and preservation of cardiac function and structure. The overarching objective of this review is to assimilate and discuss the available evidence on the functional role of podoplanin-positive cells on cardiac fibrosis and remodeling. A detailed characterization of cell-to-cell interactions and paracrine signals between podoplanin-expressing cells and the other type of cells that compose the heart tissue is needed to open a new line of investigation extending beyond the known function of these cells. This review attempts to discuss the role and biology of podoplanin-positive cells in the context of cardiac injury, repair, and remodeling.
Collapse
Affiliation(s)
- Maria Cimini
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
42
|
Guérit E, Arts F, Dachy G, Boulouadnine B, Demoulin JB. PDGF receptor mutations in human diseases. Cell Mol Life Sci 2021; 78:3867-3881. [PMID: 33449152 PMCID: PMC11072557 DOI: 10.1007/s00018-020-03753-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/16/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022]
Abstract
PDGFRA and PDGFRB are classical proto-oncogenes that encode receptor tyrosine kinases responding to platelet-derived growth factor (PDGF). PDGFRA mutations are found in gastrointestinal stromal tumors (GISTs), inflammatory fibroid polyps and gliomas, and PDGFRB mutations drive myofibroma development. In addition, chromosomal rearrangement of either gene causes myeloid neoplasms associated with hypereosinophilia. Recently, mutations in PDGFRB were linked to several noncancerous diseases. Germline heterozygous variants that reduce receptor activity have been identified in primary familial brain calcification, whereas gain-of-function mutants are present in patients with fusiform aneurysms, Kosaki overgrowth syndrome or Penttinen premature aging syndrome. Functional analysis of these variants has led to the preclinical validation of tyrosine kinase inhibitors targeting PDGF receptors, such as imatinib, as a treatment for some of these conditions. This review summarizes the rapidly expanding knowledge in this field.
Collapse
Affiliation(s)
- Emilie Guérit
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Florence Arts
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Guillaume Dachy
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Boutaina Boulouadnine
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium.
| |
Collapse
|
43
|
Hewitson TD, Smith ER. A Metabolic Reprogramming of Glycolysis and Glutamine Metabolism Is a Requisite for Renal Fibrogenesis-Why and How? Front Physiol 2021; 12:645857. [PMID: 33815149 PMCID: PMC8010236 DOI: 10.3389/fphys.2021.645857] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/22/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic Kidney Disease (CKD) is characterized by organ remodeling and fibrosis due to failed wound repair after on-going or severe injury. Key to this process is the continued activation and presence of matrix-producing renal fibroblasts. In cancer, metabolic alterations help cells to acquire and maintain a malignant phenotype. More recent evidence suggests that something similar occurs in the fibroblast during activation. To support these functions, pro-fibrotic signals released in response to injury induce metabolic reprograming to meet the high bioenergetic and biosynthetic demands of the (myo)fibroblastic phenotype. Fibrogenic signals such as TGF-β1 trigger a rewiring of cellular metabolism with a shift toward glycolysis, uncoupling from mitochondrial oxidative phosphorylation, and enhanced glutamine metabolism. These adaptations may also have more widespread implications with redirection of acetyl-CoA directly linking changes in cellular metabolism and regulatory protein acetylation. Evidence also suggests that injury primes cells to these metabolic responses. In this review we discuss the key metabolic events that have led to a reappraisal of the regulation of fibroblast differentiation and function in CKD.
Collapse
Affiliation(s)
- Timothy D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital (RMH), Melbourne, VIC, Australia.,Department of Medicine-RMH, The University of Melbourne, Melbourne, VIC, Australia
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital (RMH), Melbourne, VIC, Australia.,Department of Medicine-RMH, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
44
|
Xie T, Xia Z, Wang W, Zhou X, Xu C. BMPER Ameliorates Renal Fibrosis by Inhibiting Tubular Dedifferentiation and Fibroblast Activation. Front Cell Dev Biol 2021; 9:608396. [PMID: 33644047 PMCID: PMC7905093 DOI: 10.3389/fcell.2021.608396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/08/2021] [Indexed: 12/02/2022] Open
Abstract
Tubulointerstitial fibrosis is both a pathological manifestation of chronic kidney disease and a driving force for the progression of kidney disease. A previous study has shown that bone morphogenetic protein-binding endothelial cell precursor-derived regulator (BMPER) is involved in lung fibrogenesis. However, the role of BMPER in renal fibrosis remains unknown. In the present study, the expression of BMPER was examined by real-time PCR, Western blot and immunohistochemical staining. The in vitro effects of BMPER on tubular dedifferentiation and fibroblast activation were analyzed in cultured HK-2 and NRK-49F cells. The in vivo effects of BMPER were dissected in unilateral ureteral obstruction (UUO) mice by delivery of BMPER gene via systemic administration of plasmid vector. We reported that the expression of BMPER decreased in the kidneys of UUO mice and HK-2 cells. TGF-β1 increased inhibitor of differentiation-1 (Id-1) and induced epithelial mesenchymal transition in HK-2 cells, and knockdown of BMPER aggravated Id-1 up-regulation, E-cadherin loss, and tubular dedifferentiation. On the contrary, exogenous BMPER inhibited Id-1 up-regulation, prevented E-cadherin loss and tubular dedifferentiation after TGF-β1 exposure. In addition, exogenous BMPER suppressed fibroblast activation by hindering Erk1/2 phosphorylation. Knockdown of low-density lipoprotein receptor-related protein 1 abolished the inhibitory effect of BMPER on Erk1/2 phosphorylation and fibroblast activation. Moreover, delivery of BMPER gene improved renal tubular damage and interstitial fibrosis in UUO mice. Therefore, BMPER inhibits TGF-β1-induced tubular dedifferentiation and fibroblast activation and may hold therapeutic potential for tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Ting Xie
- Department of Woman's Health Care, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Zunen Xia
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Changgeng Xu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
GYY4137 alleviates sepsis-induced acute lung injury in mice by inhibiting the PDGFRβ/Akt/NF-κB/NLRP3 pathway. Life Sci 2021; 271:119192. [PMID: 33577850 DOI: 10.1016/j.lfs.2021.119192] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/15/2021] [Accepted: 01/31/2021] [Indexed: 12/29/2022]
Abstract
AIMS GYY4137 [GYY, morpholin-4-ium-4-methoxyphenyl (morpholino) phosphinodithioate] is a novel and perfect hydrogen sulfide (H2S) donor that is stable in vivo and in vitro. H2S, along with CO and NO, has been recognized as the third physiological gas signaling molecule that plays an active role in fighting various lung infections. However, the mechanism by which GYY4137 affects cecal ligation and puncture (CLP)-induced acute lung injury (ALI) is not understood. This study aimed to investigate whether GYY4137 inhibits the activation of the pyrin domain-containing protein 3 (NLRP3) inflammasome by inhibiting the PDGFRβ/Akt/NF-κB pathway. MAIN METHODS The model of CLP-induced ALI was established in vivo. The mice were subsequently treated with GYY4137 (25 μg/g and 50 μg/g) to simulate the realistic conditions of pathogenesis. Western blotting and immunohistochemical staining were used to examine protein expression, hematoxylin and eosin staining was used for the histopathological analysis, and the levels of inflammatory factors were determined using enzyme-linked immunosorbent assays (ELISAs). KEY FINDINGS GYY4137 significantly increased the 7-day survival of mice with septic peritonitis and protected against CLP-induced ALI, including decreasing neutrophil infiltration, improving sepsis-induced lung histopathological changes, diminishing lung tissue damage, and attenuating the severity of lung injury in mice. The protective effect of GYY4137 was undoubtedly dose-dependent. We discovered that GYY4137 reduced the levels of the p-PDGFRβ, p-NF-κB, ASC, NLRP3, caspase-1, and p-Akt proteins in septic mouse lung tissue. Akt regulates the generation of proinflammatory cytokines in endotoxemia-associated ALI by enhancing the nuclear translocation of NF-κB. SIGNIFICANCE These results indicate a new molecular mechanism explaining the effect of GYY4137 on the treatment of CLP-induced ALI in mice.
Collapse
|
46
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
47
|
Addario G, Djudjaj S, Farè S, Boor P, Moroni L, Mota C. Microfluidic bioprinting towards a renal in vitro model. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
48
|
Cha JJ, Mandal C, Ghee JY, Yoo JA, Lee MJ, Kang YS, Hyun YY, Lee JE, Kim HW, Han SY, Han JY, Chung AY, Yoon DW, Rhyu IJ, Oh J, Cha DR. Inhibition of Renal Stellate Cell Activation Reduces Renal Fibrosis. Biomedicines 2020; 8:biomedicines8100431. [PMID: 33086608 PMCID: PMC7603238 DOI: 10.3390/biomedicines8100431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Interstitial fibrosis is a common feature of chronic kidney disease, and platelet-derived growth factor receptor-β (PDGFR-β)-positive mesenchymal cells are reportedly the major source of scar-producing myofibroblasts. We had previously demonstrated that albumin and its derivative R-III (a retinol-binding protein-albumin domain III fusion protein) inhibited the transdifferentiation/activation of hepatic stellate cells (HSCs) to myofibroblasts and that R-III administration reduced liver fibrosis. In this study, we isolated cells (referred to as renal stellate cells, RSCs) from rat kidney tissues using the HSC isolation protocol and compared their morphological and biochemical characteristics with those of HSCs. RSCs shared many characteristics with HSCs, such as storage of vitamin A-containing lipid droplets and expression of HSC markers as well as pericyte markers. RSCs underwent spontaneous transdifferentiation into myofibroblasts in in vitro culture, which was inhibited by albumin expression or R-III treatment. We also evaluated the therapeutic effects of R-III in unilateral ureteral obstruction (UUO)-induced renal fibrosis in mice. Injected R-III localized predominantly in cytoglobin/stellate cell activation-associated protein (Cygb/STAP)-positive cells in the kidney and reduced renal fibrosis. These findings suggest that RSCs can be recognized as the renal counterparts of HSCs and that RSCs represent an attractive therapeutic target for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Jin Joo Cha
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
| | - Chanchal Mandal
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Korea; (C.M.); (A.Y.C.); (D.W.Y.); (I.J.R.)
| | - Jung Yeon Ghee
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
| | - Ji Ae Yoo
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
| | - Mi Jin Lee
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
| | - Young Sun Kang
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
| | - Young Youl Hyun
- Department of Nephrology, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul 03181, Korea;
| | - Ji Eun Lee
- Department of Nephrology, Wonkwang University Sanbon Hospital, Gunpo 15865, Korea; (J.E.L.); (H.W.K.)
| | - Hyun Wook Kim
- Department of Nephrology, Wonkwang University Sanbon Hospital, Gunpo 15865, Korea; (J.E.L.); (H.W.K.)
| | - Sang Youb Han
- Department of Nephrology, Inje University Ilsan Paik Hospital, Ilsan 10380, Korea;
| | - Jee Young Han
- Department of Pathology, Inha University Hospital, Incheon 22332, Korea;
| | - Ah Young Chung
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Korea; (C.M.); (A.Y.C.); (D.W.Y.); (I.J.R.)
| | - Dae Wui Yoon
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Korea; (C.M.); (A.Y.C.); (D.W.Y.); (I.J.R.)
| | - Im Joo Rhyu
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Korea; (C.M.); (A.Y.C.); (D.W.Y.); (I.J.R.)
| | - Junseo Oh
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Korea; (C.M.); (A.Y.C.); (D.W.Y.); (I.J.R.)
- Correspondence: (J.O.); (D.R.C.); Tel.: +82-2-2286-1389 (J.O.); +82-31-412-5572 (D.R.C.)
| | - Dae Ryong Cha
- Department of Nephrology, Korea University Ansan Hospital, Ansan 15355, Korea; (J.J.C.); (J.Y.G.); (J.A.Y.); (M.J.L.); (Y.S.K.)
- Correspondence: (J.O.); (D.R.C.); Tel.: +82-2-2286-1389 (J.O.); +82-31-412-5572 (D.R.C.)
| |
Collapse
|
49
|
Buhl EM, Djudjaj S, Klinkhammer BM, Ermert K, Puelles VG, Lindenmeyer MT, Cohen CD, He C, Borkham‐Kamphorst E, Weiskirchen R, Denecke B, Trairatphisan P, Saez‐Rodriguez J, Huber TB, Olson LE, Floege J, Boor P. Dysregulated mesenchymal PDGFR-β drives kidney fibrosis. EMBO Mol Med 2020; 12:e11021. [PMID: 31943786 PMCID: PMC7059015 DOI: 10.15252/emmm.201911021] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Kidney fibrosis is characterized by expansion and activation of platelet-derived growth factor receptor-β (PDGFR-β)-positive mesenchymal cells. To study the consequences of PDGFR-β activation, we developed a model of primary renal fibrosis using transgenic mice with PDGFR-β activation specifically in renal mesenchymal cells, driving their pathological proliferation and phenotypic switch toward myofibroblasts. This resulted in progressive mesangioproliferative glomerulonephritis, mesangial sclerosis, and interstitial fibrosis with progressive anemia due to loss of erythropoietin production by fibroblasts. Fibrosis induced secondary tubular epithelial injury at later stages, coinciding with microinflammation, and aggravated the progression of hypertensive and obstructive nephropathy. Inhibition of PDGFR activation reversed fibrosis more effectively in the tubulointerstitium compared to glomeruli. Gene expression signatures in mice with PDGFR-β activation resembled those found in patients. In conclusion, PDGFR-β activation alone is sufficient to induce progressive renal fibrosis and failure, mimicking key aspects of chronic kidney disease in humans. Our data provide direct proof that fibrosis per se can drive chronic organ damage and establish a model of primary fibrosis allowing specific studies targeting fibrosis progression and regression.
Collapse
Affiliation(s)
- Eva M Buhl
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
- Electron Microscopy FacilityRWTH University of AachenAachenGermany
| | - Sonja Djudjaj
- Institute of PathologyRWTH University of AachenAachenGermany
| | | | - Katja Ermert
- Institute of PathologyRWTH University of AachenAachenGermany
| | - Victor G Puelles
- Division of NephrologyRWTH University of AachenAachenGermany
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of NephrologyMonash Health, and Center for Inflammatory DiseasesMonash UniversityMelbourneVic.Australia
| | - Maja T Lindenmeyer
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Clemens D Cohen
- Nephrological CenterMedical Clinic and Policlinic IVUniversity of MunichMunichGermany
| | - Chaoyong He
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Erawan Borkham‐Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research (IZKF)RWTH University of AachenAachenGermany
| | - Panuwat Trairatphisan
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Julio Saez‐Rodriguez
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Tobias B Huber
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Lorin E Olson
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Jürgen Floege
- Division of NephrologyRWTH University of AachenAachenGermany
| | - Peter Boor
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
| |
Collapse
|
50
|
Abstract
Chronic kidney disease (CKD) is one of the fastest growing global causes of death, estimated to rank among the top five by 2040 (Foreman et al, 2018). This illustrates current pitfalls in diagnosis and management of CKD. Advanced CKD requires renal function replacement by dialysis or transplantation. However, earlier CKD stages, even when renal function is still normal, are already associated with an increased risk of premature death (Perez‐Gomez et al, 2019). Thus, novel approaches to diagnose and treat CKD are needed. The histopathological hallmark of CKD is kidney fibrosis, which is closely associated with local inflammation and loss of kidney parenchymal cells. Thus, kidney fibrosis is an attractive process to develop tests allowing an earlier diagnosis of CKD and represents a potential therapeutic target to slow CKD progression or promote regression.
Collapse
Affiliation(s)
- Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, Department of Medicine, School of Medicine, Universidad Autonoma de Madrid, Madrid, Spain.,Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|