1
|
Perutina I, Kelam N, Maglica M, Racetin A, Rizikalo A, Filipović N, Prusac IK, Bošnjak M, Mišković J, Kablar B, Ghahramani N, Vukojević K. Spatiotemporal distribution of Wnt signaling pathway markers in human congenital anomalies of kidney and urinary tract. Acta Histochem 2025; 127:152235. [PMID: 39908631 DOI: 10.1016/j.acthis.2025.152235] [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: 11/15/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
This study aimed to investigate the spatiotemporal expression patterns of key markers involved in regulating the canonical and non-canonical Wnt pathway during human fetal kidney development, comparing healthy (CTRL) and congenital anomalies of the kidney and urinary tract (CAKUT) affected kidneys. Human fetal kidneys, ranging from the 18th to the 38th developmental weeks, including various CAKUT phenotypes (horseshoe, dysplastic, duplex and hypoplastic), underwent double immunofluorescence microscopy analysis following antibody staining. Immunoreactivity levels were quantified in different kidney structures, and expression dynamics were assessed using linear and nonlinear regression modeling techniques. The study revealed a decrease in the overall protein expression of acetylated α-tubulin during normal kidney development, while the highest percentage of positive cells was observed in the horseshoe kidney (HK), thus disturbing microtubule composition in normal cell division and differentiation. Additionally, a continuous decrease of inversin-positive cells in hypoplastic (HYP) and duplex kidneys (UD), but the exponential growth of DVL-1 expression score in dysplastic kidneys (DYS) with developmental age, result in suppression of final kidney differentiation by continuous canonical Wnt signaling activation, thus supporting the essential role of the switch from canonical to non-canonical Wnt pathway in nephrogenesis. Furthermore β-catenin-positive cells in dysplastic and hypoplastic kidney exhibited the highest percentage of positive signal, with a decline in β-catenin positive cells over time in the control group, indicating disturbances in transition from canonical to non-canonical Wnt pathway in CAKUT-affected kidneys. The findings suggest that the crosstalk between canonical and non-canonical Wnt signaling pathways is crucial for normal nephrogenesis, highlighting their potential roles in normal and dysfunctional kidney development.
Collapse
Affiliation(s)
- Ilija Perutina
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina; Department of Neurosurgery, University Clinical Hospital Mostar, Mostar 88 000, Bosnia and Herzegovina.
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split 21000, Croatia; Center for translational research in biomedicine, University of Split School of Medicine, Split 21000, Croatia.
| | - Mirko Maglica
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina.
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split 21000, Croatia; Center for translational research in biomedicine, University of Split School of Medicine, Split 21000, Croatia.
| | - Azer Rizikalo
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina.
| | - Natalija Filipović
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina; Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split 21000, Croatia.
| | - Ivana Kuzmić Prusac
- Department of Pathology, University Hospital Center Split, Split 21000, Croatia.
| | - Marko Bošnjak
- Department of Neurosurgery, University Clinical Hospital Mostar, Mostar 88 000, Bosnia and Herzegovina.
| | - Josip Mišković
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina.
| | - Boris Kablar
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Nasrollah Ghahramani
- Department of Medicine, Division of Nephrology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States.
| | - Katarina Vukojević
- Department of Anatomy, University of Mostar, School of Medicine, Mostar 88000, Bosnia and Herzegovina; Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split 21000, Croatia; Center for translational research in biomedicine, University of Split School of Medicine, Split 21000, Croatia.
| |
Collapse
|
2
|
Maknis TR, Fussi MF, Pariani AP, Huhn V, Vena R, Favre C, Molinas SM, Larocca MC. Activation of angiotensin II type 2 receptor leads to preservation of primary cilia in tubular cells during renal ischaemia-reperfusion injury. J Physiol 2024; 602:5083-5103. [PMID: 39146457 DOI: 10.1113/jp286514] [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: 03/06/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Ischaemia-reperfusion (IR)-associated acute kidney injury (AKI) is a severe clinical condition that lacks effective pharmacological treatments. Our recent research revealed that pretreatment with the angiotensin II type 2 receptor (AT2R) agonist C21 alleviates kidney damage during IR. Primary cilia are organelles crucial for regulation of epithelial cell homeostasis, which are significantly affected by IR injury. This study aimed to evaluate the impact of AT2R activation on cilia integrity during IR and to identify pathways involved in the nephroprotective effect of C21. Rats were subjected to 40 min of unilateral ischaemia followed by 24 h of reperfusion. Immunofluorescence analysis of the kidneys showed that the nephroprotective effect of C21 was associated with preservation of cilia integrity in tubular cells. AT2R agonists increased α-tubulin acetylation in primary cilia in tubular cells in vivo and in a cell model. Analysis of ERK phosphorylation indicated that AT2R activation led to diminished activation of ERK1/2 in tubular cells. Similar to AT2R agonists, inhibitors of α-tubulin deacetylase HDAC6 or inhibitors of ERK activation ameliorated IR-induced cell death and preserved cilia integrity. Immunofluorescence analysis of tubular cells revealed significant ERK localization at primary cilia and demonstrated that ERK inhibition increased cilia levels of acetylated α-tubulin. Overall, our findings demonstrate that C21 elicits a preconditioning effect that enhances cilia stability in renal tubular cells, thereby preserving their integrity when exposed to IR injury. Furthermore, our results indicate that this effect might be mediated by AT2R-induced inhibition of ERK activation. These findings offer potential insights for the development of pharmacological interventions to mitigate IR-associated AKI. KEY POINTS: The AT2R agonist C21 prevents primary cilia shortening and tubular cell deciliation during renal ischaemia-reperfusion. AT2R activation inhibits ERK1/2 in renal tubular cells. Both AT2R agonists and ERK1/2 inhibitors increase alpha-tubulin acetylation at the primary cilium in tubular cells. AT2R activation, ERK1/2 inhibition or inhibition of alpha-tubulin deacetylation elicit protective effects in tubular cells subjected to ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Tomás Rivabella Maknis
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - M Fernanda Fussi
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - Alejandro P Pariani
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - Victoria Huhn
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - Rodrigo Vena
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, Rosario, Argentina
| | - Cristián Favre
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - Sara M Molinas
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| | - M Cecilia Larocca
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas (FBIOyF), Universidad Nacional de Rosario (UNR), Rosario, Argentina
| |
Collapse
|
3
|
Loukil A, Ebright E, Uezu A, Gao Y, Soderling SH, Goetz SC. Identification of new ciliary signaling pathways in the brain and insights into neurological disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572700. [PMID: 38187761 PMCID: PMC10769350 DOI: 10.1101/2023.12.20.572700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Primary cilia are conserved sensory hubs essential for signaling transduction and embryonic development. Ciliary dysfunction causes a variety of developmental syndromes with neurological features and cognitive impairment, whose basis mostly remains unknown. Despite connections to neural function, the primary cilium remains an overlooked organelle in the brain. Most neurons have a primary cilium; however, it is still unclear how this organelle modulates brain architecture and function, given the lack of any systemic dissection of neuronal ciliary signaling. Here, we present the first in vivo glance at the molecular composition of cilia in the mouse brain. We have adapted in vivo BioID (iBioID), targeting the biotin ligase BioID2 to primary cilia in neurons. We identified tissue-specific signaling networks enriched in neuronal cilia, including Eph/Ephrin and GABA receptor signaling pathways. Our iBioID ciliary network presents a wealth of neural ciliary hits that provides new insights into neurological disorders. Our findings are a promising first step in defining the fundamentals of ciliary signaling and their roles in shaping neural circuits and behavior. This work can be extended to pathological conditions of the brain, aiming to identify the molecular pathways disrupted in the brain cilium. Hence, finding novel therapeutic strategies will help uncover and leverage the therapeutic potential of the neuronal cilium.
Collapse
Affiliation(s)
- Abdelhalim Loukil
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emma Ebright
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Akiyoshi Uezu
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Yudong Gao
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Scott H Soderling
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Sarah C. Goetz
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
4
|
Phillips AT, Boumil EF, Venkatesan A, Tilstra-Smith C, Castro N, Knox BE, Henty-Ridilla JL, Bernstein AM. The formin DAAM1 regulates the deubiquitinase activity of USP10 and integrin homeostasis. Eur J Cell Biol 2023; 102:151347. [PMID: 37562219 PMCID: PMC10839120 DOI: 10.1016/j.ejcb.2023.151347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
The differentiation of fibroblasts into pathological myofibroblasts during wound healing is characterized by increased cell surface expression of αv-integrins. Our previous studies found that the deubiquitinase (DUB) USP10 removes ubiquitin from αv-integrins, leading to cell surface integrin accumulation, subsequent TGFβ1 activation, and pathological myofibroblast differentiation. In this study, a yeast two-hybrid screen revealed a novel binding partner for USP10, the formin, DAAM1. We found that DAAM1 binds to and inhibits USP10's DUB activity through the FH2 domain of DAAM1 independent of its actin functions. The USP10/DAAM1 interaction was also supported by proximity ligation assay (PLA) in primary human corneal fibroblasts. Treatment with TGFβ1 significantly increased USP10 and DAAM1 protein expression, PLA signal, and co-localization to actin stress fibers. DAAM1 siRNA knockdown significantly reduced co-precipitation of USP10 and DAAM1 on purified actin stress fibers, and β1- and β5-integrin ubiquitination. This resulted in increased αv-, β1-, and β5-integrin total protein levels, αv-integrin recycling, and extracellular fibronectin (FN) deposition. Together, our data demonstrate that DAAM1 inhibits USP10's DUB activity on integrins subsequently regulating cell surface αv-integrin localization and FN accumulation.
Collapse
Affiliation(s)
- Andrew T Phillips
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Edward F Boumil
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Arunkumar Venkatesan
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Christine Tilstra-Smith
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Nileyma Castro
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA
| | - Barry E Knox
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Jessica L Henty-Ridilla
- SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Audrey M Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA.
| |
Collapse
|
5
|
Wang J, Zhou P, Zhu L, Guan H, Gou J, Liu X. Maternal protein deficiency alters primary cilia length in renal tubular and impairs kidney development in fetal rat. Front Nutr 2023; 10:1156029. [PMID: 37485393 PMCID: PMC10358357 DOI: 10.3389/fnut.2023.1156029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Intrauterine malnutrition impairs embryo kidney development and leads to kidney disease and hypertension in adulthood, yet the underlying mechanism remains unclear. Methods With a maternal protein restriction (MPR) rat model, we investigated the critical ciliogenesis factors and β-catenin pathway in FGR fetal kidneys and analyzed the impact of aberrant primary cilia on renal tubular epithelium. Results The data showed decreased nephron number and renal tubular dysgenesis in FGR fetus. FGR fetus showed deregulated expression of ciliogenesis factors including upregulation of IFT88 and downregulation of DYNLT1, accompanied with cilia elongation in renal tubular epithelial cells. Wnt7b, the key ligand for Wnt/β-catenin signaling, was downregulated and nuclear translocation of β-catenin was decreased. The proapoptotic protein was upregulated. In vitro study with HK-2 cells showed that overexpression of IFT88 lengthened the cilia, inhibited β-catenin signaling. Besides, IFT88 overexpression suppressed cell proliferation, activated autophagy, and induced cell apoptosis. Inhibition of autophagy partly restored the cilia length and cell viability. Likewise, knockdown of DYNLT1 led to cilia elongation, suppressed cell proliferation, and promoted apoptosis in HK-2 cell. However, the cilia elongation induced by DYNLT1 knockdown was not autophagy-dependent, but associated with reactive oxygen species (ROS) accumulation. Discussion We elucidated that intrauterine protein malnutrition led to deregulation of ciliogenesis factors and cilia elongation in renal tubular epithelial, inhibited β-catenin signaling, and induced cell apoptosis and ultimately, compromised kidney development.
Collapse
Affiliation(s)
- Jun Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Pei Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Liangliang Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongbo Guan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jian Gou
- Department of Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaomei Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Perutina I, Kelam N, Maglica M, Racetin A, Ogorevc M, Filipović N, Katsuyama Y, Mišković J, Vukojević K. Disturbances in Switching between Canonical and Non-Canonical Wnt Signaling Characterize Developing and Postnatal Kidneys of Dab1-/- ( yotari) Mice. Biomedicines 2023; 11:biomedicines11051321. [PMID: 37238991 DOI: 10.3390/biomedicines11051321] [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: 03/07/2023] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
This study aims to determine the protein expression patterns of acetylated α-tubulin, inversin, dishevelled-1, Wnt5a/b, and β-catenin in developing (E13.5 and E15.5) and early postnatal (P4 and P14) kidneys of Dab1-/- (yotari) mice, their role in regulating the Wnt signaling pathway, and the possible relation to congenital anomalies of kidney and urinary tract (CAKUT). The analysis of target protein co-expression, observed in the renal vesicles/immature glomeruli, ampullae/collecting ducts, convoluted tubules, metanephric mesenchyme of developing kidneys, but proximal convoluted tubules, distal convoluted tubules and glomeruli of postnatal kidneys, was performed using double immunofluorescence and semi-quantitative methods. The overall expression of acetylated α-tubulin and inversin during normal kidney development increases with higher expression in yotari mice as the kidney acquires mature morphology. An increase in β-catenin and cytosolic DVL-1 levels, indicating a switch from non-canonical to canonical Wnt signaling, is found in the postnatal kidney of yotari mice. In contrast, healthy mouse kidney expresses inversin and Wnt5a/b in the postnatal period, thus activating non-canonical Wnt signaling. Target protein expression patterns in kidney development and the early postnatal period observed in this study could indicate that switching between canonical and non-canonical Wnt signaling is crucial for normal nephrogenesis, while the defective Dab1 gene product in yotari mice may promote CAKUT due to interfering with this process.
Collapse
Affiliation(s)
- Ilija Perutina
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Mirko Maglica
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Josip Mišković
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Katarina Vukojević
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, University of Split School of Medicine, 21000 Split, Croatia
| |
Collapse
|
7
|
Bea-Mascato B, Neira-Goyanes E, Iglesias-Rodríguez A, Valverde D. Depletion of ALMS1 affects TGF-β signalling pathway and downstream processes such as cell migration and adhesion capacity. Front Mol Biosci 2022; 9:992313. [PMID: 36325276 PMCID: PMC9621122 DOI: 10.3389/fmolb.2022.992313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/13/2022] [Indexed: 12/23/2023] Open
Abstract
Background: ALMS1 is a ubiquitous gene associated with Alström syndrome (ALMS). The main symptoms of ALMS affect multiple organs and tissues, generating at last, multi-organic fibrosis in the lungs, kidneys and liver. TGF-β is one of the main pathways implicated in fibrosis, controlling the cell cycle, apoptosis, cell migration, cell adhesion and epithelial-mesenchymal transition (EMT). Nevertheless, the role of ALMS1 gene in fibrosis generation and other implicated processes such as cell migration or cell adhesion via the TGF- β pathway has not been elucidated yet. Methods: Initially, we evaluated how depletion of ALMS1 affects different processes like apoptosis, cell cycle and mitochondrial activity in HeLa cells. Then, we performed proteomic profiling with TGF-β stimuli in HeLa ALMS1 -/- cells and validated the results by examining different EMT biomarkers using qPCR. The expression of these EMT biomarkers were also studied in hTERT-BJ-5ta ALMS1 -/-. Finally, we evaluated the SMAD3 and SMAD2 phosphorylation and cell migration capacity in both models. Results: Depletion of ALMS1 generated apoptosis resistance to thapsigargin (THAP) and C2-Ceramide (C2-C), and G2/M cell cycle arrest in HeLa cells. For mitochondrial activity, results did not show significant differences between ALMS1 +/+ and ALMS1 -/-. Proteomic results showed inhibition of downstream pathways regulated by TGF-β. The protein-coding genes (PCG) were associated with processes like focal adhesion or cell-substrate adherens junction in HeLa. SNAI1 showed an opposite pattern to what would be expected when activating the EMT in HeLa and BJ-5ta. Finally, in BJ-5ta model a reduced activation of SMAD3 but not SMAD2 were also observed. In HeLa model no alterations in the canonical TGF-β pathway were observed but both cell lines showed a reduction in migration capacity. Conclusion: ALMS1 has a role in controlling the cell cycle and the apoptosis processes. Moreover, the depletion of ALMS1 affects the signal transduction through the TGF-β and other processes like the cell migration and adhesion capacity.
Collapse
Affiliation(s)
- Brais Bea-Mascato
- CINBIO, Universidad de Vigo, Vigo, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Elena Neira-Goyanes
- CINBIO, Universidad de Vigo, Vigo, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Antía Iglesias-Rodríguez
- CINBIO, Universidad de Vigo, Vigo, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Diana Valverde
- CINBIO, Universidad de Vigo, Vigo, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| |
Collapse
|
8
|
Primary Cilia: A Cellular Regulator of Articular Cartilage Degeneration. Stem Cells Int 2022; 2022:2560441. [PMID: 36193252 PMCID: PMC9525753 DOI: 10.1155/2022/2560441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/29/2022] [Accepted: 09/02/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease that can cause pain and disability in adults. The main pathological characteristic of OA is cartilage degeneration, which is caused by chondrocyte apoptosis, cartilage matrix degradation, and inflammatory factor destruction. The current treatment for patients with OA focuses on delaying its progression, such as oral anti-inflammatory analgesics or injection of sodium gluconate into the joint cavity. Primary cilia are an important structure involved in cellular signal transduction. Thus, they are very sensitive to mechanical and physicochemical stimuli. It is reported that the primary cilia may play an important role in the development of OA. Here, we review the correlation between the morphology (location, length, incidence, and orientation) of chondrocyte primary cilia and OA and summarize the relevant signaling pathways in chondrocytes that could regulate the OA process through primary cilia, including Hedgehog, Wnt, and inflammation-related signaling pathways. These data provide new ideas for OA treatment.
Collapse
|
9
|
Expression Pattern of α-Tubulin, Inversin and Its Target Dishevelled-1 and Morphology of Primary Cilia in Normal Human Kidney Development and Diseases. Int J Mol Sci 2021; 22:ijms22073500. [PMID: 33800671 PMCID: PMC8037028 DOI: 10.3390/ijms22073500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
The spatiotemporal expression of α-tubulin, inversin and dishevelled-1 (DVL-1) proteins associated with the Wnt-signaling pathway, and primary cilia morphology were analyzed in developing kidneys (14th–38th developmental weeks), healthy postnatal (1.5- and 7-years old) and pathologically changed human kidneys, including multicystic dysplastic kidneys (MCDK), focal segmental glomerulosclerosis (FSGS) and nephrotic syndrome of the Finnish type (CNF). The analysis was performed by double immunofluorescence, electron microscopy, semiquantitative and statistical methods. Cytoplasmic co-expression of α-tubulin, inversin and DVL-1 was observed in the proximal convoluted tubules (pct), distal convoluted tubules (dct) and glomeruli (g) of analyzed tissues. During kidney development, the overall expression of α-tubulin, inversin and DVL-1 decreased, while in the postnatal period slightly increased. The highest expressions of α-tubulin and inversin characterized dct and g, while high DVL-1 characterized pct. α-tubulin, inversin and DVL-1 expression pattern in MCDK, FSGS and CNF kidneys significantly differed from the healthy control. Compared to healthy kidneys, pathologically changed kidneys had dysmorphic primary cilia. Different expression dynamics of α-tubulin, inversin and DVL-1 during kidney development could indicate that switch between the canonical and noncanonical Wnt-signaling is essential for normal kidney morphogenesis. In contrast, their disturbed expression in pathological kidneys might be associated with abnormal primary cilia, leading to chronic kidney diseases.
Collapse
|
10
|
Hu J, Qiao J, Yu Q, Liu B, Zhen J, Liu Y, Ma Q, Li Y, Wang Q, Wang C, Lv Z. Role of SIK1 in the transition of acute kidney injury into chronic kidney disease. J Transl Med 2021; 19:69. [PMID: 33588892 PMCID: PMC7885408 DOI: 10.1186/s12967-021-02717-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/23/2021] [Indexed: 12/20/2022] Open
Abstract
Background Acute kidney injury (AKI), with a high morbidity and mortality, is recognized as a risk factor for chronic kidney disease (CKD). AKI-CKD transition has been regarded as one of the most pressing unmet needs in renal diseases. Recently, studies have showed that salt inducible kinase 1 (SIK1) plays a role in epithelial-mesenchymal transition (EMT) and inflammation, which are the hallmarks of AKI-CKD transition. However, whether SIK1 is involved in AKI-CKD transition and by what mechanism it regulates AKI-CKD transition remains unknown. Methods We firstly detected the expression of SIK1 in kidney tissues of AKI patients and AKI mice by immunohistochemistry staining, and then we established Aristolochic acid (AA)-induced AKI-CKD transition model in C57BL/6 mice and HK2 cells. Subsequently, we performed immunohistochemistry staining, ELISA, real-time PCR, Western blot, immunofluorescence staining and Transwell assay to explore the role and underlying mechanism of SIK1 on AKI-CKD transition. Results The expression of SIK1 was down-regulated in AKI patients, AKI mice, AA-induced AKI-CKD transition mice, and HK2 cells. Functional analysis revealed that overexpression of SIK1 alleviated AA-induced AKI-CKD transition and HK2 cells injury in vivo and in vitro. Mechanistically, we demonstrated that SIK1 mediated AA-induced AKI-CKD transition by regulating WNT/β-catenin signaling, the canonical pathway involved in EMT, inflammation and renal fibrosis. In addition, we discovered that inhibition of WNT/β-catenin pathway and its downstream transcription factor Twist1 ameliorated HK2 cells injury, delaying the progression of AKI-CKD transition. Conclusions Our study demonstrated, for the first time, a protective role of SIK1 in AKI-CKD transition by regulating WNT/β-catenin signaling pathway and its downstream transcription factor Twist1, which will provide novel insights into the prevention and treatment AKI-CKD transition in the future.
Collapse
Affiliation(s)
- Jinxiu Hu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Jiao Qiao
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Qun Yu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Bing Liu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Junhui Zhen
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Yue Liu
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Qiqi Ma
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanmei Li
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Qianhui Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Cheng Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
11
|
Nlandu-Khodo S, Osaki Y, Scarfe L, Yang H, Phillips-Mignemi M, Tonello J, Saito-Diaz K, Neelisetty S, Ivanova A, Huffstater T, McMahon R, Taketo MM, deCaestecker M, Kasinath B, Harris RC, Lee E, Gewin LS. Tubular β-catenin and FoxO3 interactions protect in chronic kidney disease. JCI Insight 2020; 5:135454. [PMID: 32369448 DOI: 10.1172/jci.insight.135454] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays an important role in renal development and is reexpressed in the injured kidney and other organs. β-Catenin signaling is protective in acute kidney injury (AKI) through actions on the proximal tubule, but the current dogma is that Wnt/β-catenin signaling promotes fibrosis and development of chronic kidney disease (CKD). As the role of proximal tubular β-catenin signaling in CKD remains unclear, we genetically stabilized (i.e., activated) β-catenin specifically in murine proximal tubules. Mice with increased tubular β-catenin signaling were protected in 2 murine models of AKI to CKD progression. Oxidative stress, a common feature of CKD, reduced the conventional T cell factor/lymphoid enhancer factor-dependent β-catenin signaling and augmented FoxO3-dependent activity in proximal tubule cells in vitro and in vivo. The protective effect of proximal tubular β-catenin in renal injury required the presence of FoxO3 in vivo. Furthermore, we identified cystathionine γ-lyase as a potentially novel transcriptional target of β-catenin/FoxO3 interactions in the proximal tubule. Thus, our studies overturned the conventional dogma about β-catenin signaling and CKD by showing a protective effect of proximal tubule β-catenin in CKD and identified a potentially new transcriptional target of β-catenin/FoxO3 signaling that has therapeutic potential for CKD.
Collapse
Affiliation(s)
- Stellor Nlandu-Khodo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Yosuke Osaki
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Lauren Scarfe
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, Tennessee, USA
| | - Melanie Phillips-Mignemi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Jane Tonello
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | | | - Surekha Neelisetty
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Tessa Huffstater
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert McMahon
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mark deCaestecker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Balakuntalam Kasinath
- Department of Medicine, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Medicine, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ethan Lee
- Department of Cell and Developmental Biology and
| | - Leslie S Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Department of Cell and Developmental Biology and.,Department of Medicine, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Li S, Wei Z, Li G, Zhang Q, Niu S, Xu D, Mao N, Chen S, Gao X, Cai W, Zhu Y, Zhang G, Li D, Yi X, Yang F, Xu H. Silica Perturbs Primary Cilia and Causes Myofibroblast Differentiation during Silicosis by Reduction of the KIF3A-Repressor GLI3 Complex. Theranostics 2020; 10:1719-1732. [PMID: 32042332 PMCID: PMC6993221 DOI: 10.7150/thno.37049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
The purpose of this study was to determine the effects of Kinesin family member 3A (KIF3A) on primary cilia and myofibroblast differentiation during silicosis by regulating Sonic hedgehog (SHH) signalling. Methods: Changes in primary cilia during silicosis and myofibroblast differentiation were detected in silicotic patients, experimental silicotic rats, and a myofibroblast differentiation model induced by SiO2. We also explored the mechanisms underlying KIF3A regulation of Glioma-associated oncogene homologs (GLIs) involved in myofibroblast differentiation. Results: Primary cilia (marked by ARL13B and Ac-α-Tub) and ciliary-related proteins (IFT 88 and KIF3A) were increased initially and then decreased as silicosis progressed. Loss and shedding of primary cilia were also found during silicosis. Treatment of MRC-5 fibroblasts with silica and then transfection of KIF3A-siRNA blocked activation of SHH signalling, but increased GLI2FL as a transcriptional activator of SRF, and reduced the inhibitory effect of GLI3R on ACTA2. Conclusion: Our findings indicate that primary cilia are markedly altered during silicosis and the loss of KIF3A may promote myofibroblast differentiation induced by SiO2.
Collapse
|
13
|
Uytingco CR, Green WW, Martens JR. Olfactory Loss and Dysfunction in Ciliopathies: Molecular Mechanisms and Potential Therapies. Curr Med Chem 2019; 26:3103-3119. [PMID: 29303074 DOI: 10.2174/0929867325666180105102447] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/08/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ciliopathies are a class of inherited pleiotropic genetic disorders in which alterations in cilia assembly, maintenance, and/or function exhibit penetrance in the multiple organ systems. Olfactory dysfunction is one such clinical manifestation that has been shown in both patients and model organisms. Existing therapies for ciliopathies are limited to the treatment or management of symptoms. The last decade has seen an increase in potential curative therapeutic options including small molecules and biologics. Recent work in multiciliated olfactory sensory neurons has demonstrated the capacity of targeted gene therapy to restore ciliation in terminally differentiated cells and rescue olfactory function. This review will discuss the current understanding of the penetrance of ciliopathies in the olfactory system. Importantly, it will highlight both pharmacological and biological approaches, and their potential therapeutic value in the olfactory system and other ciliated tissues. METHODS We undertook a structured and comprehensive search of peer-reviewed research literature encompassing in vitro, in vivo, model organism, and clinical studies. From these publications, we describe the olfactory system, and discuss the penetrance of ciliopathies and impact of cilia loss on olfactory function. In addition, we outlined the developing therapies for ciliopathies across different organ and cell culture systems, and discussed their potential therapeutic application to the mammalian olfactory system. RESULTS One-hundred sixty-one manuscripts were included in the review, centering on the understanding of olfactory penetrance of ciliopathies, and discussing the potential therapeutic options for ciliopathies in the context of the mammalian olfactory system. Forty-four manuscripts were used to generate a table listing the known congenital causes of olfactory dysfunction, with the first ten listed are linked to ciliopathies. Twenty-three manuscripts were used to outline the potential of small molecules for the olfactory system. Emphasis was placed on HDAC6 inhibitors and lithium, both of which were shown to stabilize microtubule structures, contributing to ciliogenesis and cilia lengthening. Seventy-five manuscripts were used to describe gene therapy and gene therapeutic strategies. Included were the implementation of adenoviral, adeno-associated virus (AAV), and lentiviral vectors to treat ciliopathies across different organ systems and application toward the olfactory system. Thus far, adenoviral and AAVmeditated ciliary restoration demonstrated successful proof-of-principle preclinical studies. In addition, gene editing, ex vivo gene therapy, and transplantation could serve as alternative therapeutic and long-term approaches. But for all approaches, additional assessment of vector immunogenicity, specificity, and efficacy need further investigation. Currently, ciliopathy treatments are limited to symptomatic management with no curative options. However, the accessibility and amenability of the olfactory system to treatment would facilitate development and advancement of a viable therapy. CONCLUSION The findings of this review highlight the contribution of ciliopathies to a growing list of congenial olfactory dysfunctions. Promising results from other organ systems imply the feasibility of biologics, with results from gene therapies proving to be a viable therapeutic option for ciliopathies and olfactory dysfunction.
Collapse
Affiliation(s)
- Cedric R Uytingco
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Warren W Green
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| |
Collapse
|
14
|
Fu D, Senouthai S, Wang J, You Y. FKN Facilitates HK-2 Cell EMT and Tubulointerstitial Lesions via the Wnt/β-Catenin Pathway in a Murine Model of Lupus Nephritis. Front Immunol 2019; 10:784. [PMID: 31134047 PMCID: PMC6524725 DOI: 10.3389/fimmu.2019.00784] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Fractalkine (FKN), also known as chemokine (C-X3-C motif) ligand 1, constitutes an intriguing chemokine with a documented role in the development of numerous inflammatory diseases including autoimmune disease. Specifically, it has been reported that FKN is involved in the disease progression of lupus nephritis (LN). The epithelial-mesenchymal transition (EMT) plays a significant role in the formation of tubulointerstitial lesions (TIL), which are increasingly recognized as a hallmark of tissue fibrogenesis after injury. However, the correlation between FKN and EMT or TIL in LN has not been determined. To investigate the potential role of FKN in EMT and TIL, MRL lymphoproliferation (MRL/lpr) strain mice were treated with an anti-FKN antibody, recombinant-FKN chemokine domain, or isotype antibody. Our results revealed that treatment with the anti-FKN antibody improved EMT, TIL, and renal function in MRL/lpr mice, along with inhibiting activation of the Wnt/β-catenin signaling pathway. In contrast, administration of the recombinant-FKN chemokine domain had the opposite effect. Furthermore, to further explore the roles of FKN in EMT, we assessed the levels of EMT markers in FKN-depleted or overexpressing human proximal tubule epithelial HK-2 cells. Our results provide the first evidence that the E-cadherin level was upregulated, whereas α-SMA and vimentin expression was downregulated in FKN-depleted HK-2 cells. In contrast, overexpression of FKN in HK-2 cells enhanced EMT. In addition, inhibition of the Wnt/β-catenin pathway by XAV939 negated the effect of FKN overexpression, whereas activation of the Wnt/β-catenin pathway by Ang II impaired the effect of the FKN knockout on EMT in HK-2 cells. Together, our data indicate that FKN plays essential roles in the EMT progression and development of TIL in MRL/lpr mice, most likely through activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Dongdong Fu
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Soulixay Senouthai
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Junjie Wang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yanwu You
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
15
|
Non‑canonical Wnt signaling contributes to ventilator‑induced lung injury through upregulation of WISP1 expression. Int J Mol Med 2019; 43:1217-1228. [PMID: 30664165 PMCID: PMC6365043 DOI: 10.3892/ijmm.2019.4067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022] Open
Abstract
Mechanical ventilation may cause ventilator-induced lung injury (VILI). Canonical Wnt signaling has been reported to serve an important role in the pathogenesis of VILI. Bioinformatics analysis revealed that canonical and non-canonical Wnt signaling pathways were activated in VILI. However, the role of non-canonical Wnt signaling in the pathogenesis of VILI remains unclear. The present study aimed to analyze the potential role of non-canonical Wnt signaling in VILI pathogenesis. Lung injury was assessed via Evans blue albumin permeability and histological scoring, as well as by inflammatory cytokine expression and total protein concentration in bronchoalveolar lavage fluid. The relative protein expression of canonical and non-canonical Wnt signaling pathway components were examined via western blotting and immunohistochemistry. The results demonstrated that 6 h of mechanical ventilation at low tidal volume (LTV; 6 ml/kg) or moderate tidal volume (MTV; 12 ml/kg) induced lung injury in sensitive A/J mice. Ventilation with MTV increased the protein levels of Wnt-induced secreted protein 1 (WISP1), Rho-associated protein kinase 1 (ROCK1), phosphorylated (p)-Ras homolog gene family, member A and p-C-Jun N-terminal kinase (JNK). Inhibition of ROCK1 by Y27632 and JNK by SP600125 attenuated MTV-induced lung injury and decreased the expression of proteins involved in non-canonical Wnt signaling, including WISP1. In conclusion, non-canonical Wnt signaling participates in VILI by modulating WISP1 expression, which has been previously noted as critical for VILI development. Therefore, the non-canonical Wnt signaling pathway may provide a preventive and therapeutic target in VILI.
Collapse
|
16
|
Park KM. Can Tissue Cilia Lengths and Urine Cilia Proteins Be Markers of Kidney Diseases? Chonnam Med J 2018; 54:83-89. [PMID: 29854673 PMCID: PMC5972129 DOI: 10.4068/cmj.2018.54.2.83] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/02/2018] [Accepted: 02/06/2018] [Indexed: 01/22/2023] Open
Abstract
The primary cilium is an organelle which consists of a microtubule in the core and a surrounding cilia membrane, and has long been recognized as a “vestigial organelle”. However, new evidence demonstrates that the primary cilium has a notable effect on signal transduction in the cell and is associated with some genetic and non-genetic diseases. In the kidney, the primary cilium protrudes into the Bowman's space and the tubular lumen from the apical side of epithelial cells. The length of primary cilia is dynamically altered during the normal cell cycle, being shortened by retraction into the cell body at the entry of cell division and elongated at differentiation. Furthermore, the length of primary cilia is also dynamically changed in the cells, as a result and/or cause, during the progression of various kidney diseases including acute kidney injury and chronic kidney disease. Notably, recent data has demonstrated that the shortening of the primary cilium in the cell is associated with fragmentation, apart from retraction into the cell body, in the progression of diseases and that the fragmented primary cilia are released into the urine. This data reveals that the alteration of primary cilia length could be related to the progression of diseases. This review will consider if primary cilia length alteration is associated with the progression of kidney diseases and if the length of tissue primary cilia and the presence or increase of cilia proteins in the urine is indicative of kidney diseases.
Collapse
Affiliation(s)
- Kwon Moo Park
- Department of Anatomy and BK21 Plus, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
17
|
Skalická K, Hrčková G, Vaská A, Baranyaiová Á, Kovács L. Genetic defects in ciliary genes in autosomal dominant polycystic kidney disease. World J Nephrol 2018; 7:65-70. [PMID: 29527510 PMCID: PMC5838416 DOI: 10.5527/wjn.v7.i2.65] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 12/31/2017] [Accepted: 02/05/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the genetic defects of ciliary genes causing the loss of primary cilium in autosomal dominant polycystic kidney disease (ADPKD).
METHODS We analyzed 191 structural and functional genes of the primary cilium using next-generation sequencing analysis. We analyzed the kidney samples, which were obtained from 7 patients with ADPKD who underwent nephrectomy. Each sample contained polycystic kidney tissue and matched normal kidney tissue.
RESULTS In our study, we identified genetic defects in the 5 to 15 genes in each ADPKD sample. The most frequently identified defects were found in genes encoding centrosomal proteins (PCM1, ODF2, HTT and CEP89) and kinesin family member 19 (KIF19), which are important for ciliogenesis. In addition, pathogenic mutations in the PCM1 and KIF19 genes were found in all ADPKD samples. Interestingly, mutations in the genes encoding the intraflagellar transport proteins, which are the basis of animal models of ADPKD, were only rarely detected.
CONCLUSION The results of our study revealed the actual state of structural ciliary genes in human ADPKD tissues and provided valuable indications for further research.
Collapse
Affiliation(s)
- Katarína Skalická
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Gabriela Hrčková
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Anita Vaská
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - Ágnes Baranyaiová
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| | - László Kovács
- Laboratory of Clinical and Molecular Genetics, Department of Paediatrics, Faculty of Medicine, Comenius University and University Children’s Hospital, Bratislava 83340, Slovakia
| |
Collapse
|
18
|
Christensen ST, Morthorst SK, Mogensen JB, Pedersen LB. Primary Cilia and Coordination of Receptor Tyrosine Kinase (RTK) and Transforming Growth Factor β (TGF-β) Signaling. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028167. [PMID: 27638178 DOI: 10.1101/cshperspect.a028167] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Since the beginning of the millennium, research in primary cilia has revolutionized our way of understanding how cells integrate and organize diverse signaling pathways during vertebrate development and in tissue homeostasis. Primary cilia are unique sensory organelles that detect changes in their extracellular environment and integrate and transmit signaling information to the cell to regulate various cellular, developmental, and physiological processes. Many different signaling pathways have now been shown to rely on primary cilia to function properly, and mutations that lead to ciliary dysfunction are at the root of a pleiotropic group of diseases and syndromic disorders called ciliopathies. In this review, we present an overview of primary cilia-mediated regulation of receptor tyrosine kinase (RTK) and transforming growth factor β (TGF-β) signaling. Further, we discuss how defects in the coordination of these pathways may be linked to ciliopathies.
Collapse
Affiliation(s)
- Søren T Christensen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Stine K Morthorst
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Johanne B Mogensen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| | - Lotte B Pedersen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen OE, Denmark
| |
Collapse
|
19
|
|
20
|
Wanka H, Lutze P, Staar D, Peters B, Morch A, Vogel L, Chilukoti RK, Homuth G, Sczodrok J, Bäumgen I, Peters J. (Pro)renin receptor (ATP6AP2) depletion arrests As4.1 cells in the G0/G1 phase thereby increasing formation of primary cilia. J Cell Mol Med 2017; 21:1394-1410. [PMID: 28215051 PMCID: PMC5487920 DOI: 10.1111/jcmm.13069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/24/2016] [Indexed: 01/07/2023] Open
Abstract
The (pro)renin receptor [(P)RR, ATP6AP2] is a multifunctional transmembrane protein that activates local renin-angiotensin systems, but also interacts with Wnt pathways and vacuolar H+ -ATPase (V-ATPase) during organogenesis. The aim of this study was to characterize the role of ATP6AP2 in the cell cycle in more detail. ATP6AP2 down-regulation by siRNA in renal As4.1 cells resulted in a reduction in the rate of proliferation and a G0/G1 phase cell cycle arrest. We identified a number of novel target genes downstream of ATP6AP2 knock-down that were related to the primary cilium (Bbs-1, Bbs-3, Bbs-7, Rabl5, Ttc26, Mks-11, Mks-5, Mks-2, Tctn2, Nme7) and the cell cycle (Pierce1, Clock, Ppif). Accordingly, the number of cells expressing the primary cilium was markedly increased. We found no indication that these effects were dependent of V-ATPase activity, as ATP6AP2 knock-down did not affect lysosomal pH and bafilomycin A neither influenced the ciliary expression pattern nor the percentage of ciliated cells. Furthermore, ATP6AP2 appears to be essential for mitosis. ATP6AP2 translocated from the endoplasmatic reticulum to mitotic spindle poles (pro-, meta- and anaphase) and the central spindle bundle (telophase) and ATP6AP2 knock-down results in markedly deformed spindles. We conclude that ATP6AP2 is necessary for cell division, cell cycle progression and mitosis. ATP6AP2 also inhibits ciliogenesis, thus promoting proliferation and preventing differentiation.
Collapse
Affiliation(s)
- Heike Wanka
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Philipp Lutze
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Doreen Staar
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Barbara Peters
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Anica Morch
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Lukas Vogel
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Ravi Kumar Chilukoti
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt-University Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt-University Greifswald, Greifswald, Germany
| | - Jaroslaw Sczodrok
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Inga Bäumgen
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Jörg Peters
- Department of Physiology, University Medicine Greifswald, Karlsburg, Germany
| |
Collapse
|
21
|
Gewin L, Zent R, Pozzi A. Progression of chronic kidney disease: too much cellular talk causes damage. Kidney Int 2016; 91:552-560. [PMID: 27773427 DOI: 10.1016/j.kint.2016.08.025] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/31/2016] [Accepted: 08/16/2016] [Indexed: 01/10/2023]
Abstract
Tubulointerstitial fibrosis, tubular atrophy, and peritubular capillary rarefaction are major hallmarks of chronic kidney disease. The tubulointerstitium consists of multiple cell components including tubular epithelial, mesenchymal (fibroblasts and pericytes), endothelial, and inflammatory cells. Crosstalk among these cell components is a key component in the pathogenesis of this complex disease. After severe or recurrent injury, the renal tubular epithelial cells undergo changes in structure and cell cycle that are accompanied by altered expression and production of cytokines. These cytokines contribute to the initiation of the fibrotic response by favoring activation of fibroblasts, recruitment of inflammatory cells, and loss of endothelial cells. This review focuses on how augmented growth factor and cytokine production induces epithelial crosstalk with cells in the interstitium to promote progressive tubulointerstitial fibrosis after renal injury.
Collapse
Affiliation(s)
- Leslie Gewin
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA.
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Tampe B, Steinle U, Tampe D, Carstens JL, Korsten P, Zeisberg EM, Müller GA, Kalluri R, Zeisberg M. Low-dose hydralazine prevents fibrosis in a murine model of acute kidney injury-to-chronic kidney disease progression. Kidney Int 2016; 91:157-176. [PMID: 27692563 DOI: 10.1016/j.kint.2016.07.042] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/01/2016] [Accepted: 07/28/2016] [Indexed: 11/18/2022]
Abstract
Acute kidney injury (AKI) and progressive chronic kidney disease (CKD) are intrinsically tied syndromes. In this regard, the acutely injured kidney often does not achieve its full regenerative capacity and AKI directly transitions into progressive CKD associated with tubulointerstitial fibrosis. Underlying mechanisms of such AKI-to-CKD progression are still incompletely understood and specific therapeutic interventions are still elusive. Because epigenetic modifications play a role in maintaining tissue fibrosis, we used a murine model of ischemia-reperfusion injury to determine whether aberrant promoter methylation of RASAL1 contributes causally to the switch between physiological regeneration and tubulointerstitial fibrogenesis, a hallmark of AKI-to-CKD progression. It is known that the antihypertensive drug hydralazine has demethylating activity, and that its optimum demethylating activity occurs at concentrations below blood pressure-lowering doses. Administration of low-dose hydralazine effectively induced expression of hydroxylase TET3, which catalyzed RASAL1 hydroxymethylation and subsequent RASAL1 promoter demethylation. Hydralazine-induced CpG promoter demethylation subsequently attenuated renal fibrosis and preserved excretory renal function independent of its blood pressure-lowering effects. In comparison, RASAL1 demethylation and inhibition of tubulointerstitial fibrosis was not detected upon administration of the angiotensin-converting enzyme inhibitor Ramipril in this model. Thus, RASAL1 promoter methylation and subsequent transcriptional RASAL1 suppression plays a causal role in AKI-to-CKD progression.
Collapse
Affiliation(s)
- Björn Tampe
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Ulrike Steinle
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Désirée Tampe
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Julienne L Carstens
- Department of Cancer Biology and the Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peter Korsten
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Elisabeth M Zeisberg
- Department of Cardiology and Pneumology, Göttingen University Medical Center, Georg August University, Göttingen, Germany; German Center for Cardiovascular Research, Göttingen, Germany
| | - Gerhard A Müller
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Raghu Kalluri
- Department of Cancer Biology and the Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany; German Center for Cardiovascular Research, Göttingen, Germany.
| |
Collapse
|
23
|
Edeling M, Ragi G, Huang S, Pavenstädt H, Susztak K. Developmental signalling pathways in renal fibrosis: the roles of Notch, Wnt and Hedgehog. Nat Rev Nephrol 2016; 12:426-39. [PMID: 27140856 DOI: 10.1038/nrneph.2016.54] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Kidney fibrosis is a common histological manifestation of functional decline in the kidney. Fibrosis is a reactive process that develops in response to excessive epithelial injury and inflammation, leading to myofibroblast activation and an accumulation of extracellular matrix. Here, we describe how three key developmental signalling pathways - Notch, Wnt and Hedgehog (Hh) - are reactivated in response to kidney injury and contribute to the fibrotic response. Although transient activation of these pathways is needed for repair of injured tissue, their sustained activation is thought to promote fibrosis. Excessive Wnt and Notch expression prohibit epithelial differentiation, whereas increased Wnt and Hh expression induce fibroblast proliferation and myofibroblastic transdifferentiation. Notch, Wnt and Hh are fundamentally different signalling pathways, but their choreographed activation seems to be just as important for fibrosis as it is for embryonic kidney development. Decreasing the activity of Notch, Wnt or Hh signalling could potentially provide a new therapeutic strategy to ameliorate the development of fibrosis in chronic kidney disease.
Collapse
Affiliation(s)
- Maria Edeling
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA.,Department of Molecular Nephrology, Internal Medicine D, University Hospital Albert-Schweitzer-Straße 33, Münster 48149, Germany
| | - Grace Ragi
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| | - Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| | - Hermann Pavenstädt
- Department of Molecular Nephrology, Internal Medicine D, University Hospital Albert-Schweitzer-Straße 33, Münster 48149, Germany
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
24
|
Tan X, Xu X, Zeisberg EM, Zeisberg M. High inorganic phosphate causes DNMT1 phosphorylation and subsequent fibrotic fibroblast activation. Biochem Biophys Res Commun 2016; 472:459-64. [DOI: 10.1016/j.bbrc.2016.01.077] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 01/12/2016] [Indexed: 12/20/2022]
|
25
|
Tan X, Xu X, Zeisberg M, Zeisberg EM. DNMT1 and HDAC2 Cooperate to Facilitate Aberrant Promoter Methylation in Inorganic Phosphate-Induced Endothelial-Mesenchymal Transition. PLoS One 2016; 11:e0147816. [PMID: 26815200 PMCID: PMC4729486 DOI: 10.1371/journal.pone.0147816] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/09/2016] [Indexed: 12/17/2022] Open
Abstract
While phosphorus in the form of inorganic or organic phosphate is critically involved in most cellular functions, high plasma levels of inorganic phosphate levels have emerged as independent risk factor for cardiac fibrosis, cardiovascular morbidity and decreased life-expectancy. While the link of high phosphate and cardiovascular disease is commonly explained by direct cellular effects of phospho-regulatory hormones, we here explored the possibility of inorganic phosphate directly eliciting biological responses in cells. We demonstrate that human coronary endothelial cells (HCAEC) undergo an endothelial-mesenchymal transition (EndMT) when exposed to high phosphate. We further demonstrate that such EndMT is initiated by recruitment of aberrantly phosphorylated DNMT1 to the RASAL1 CpG island promoter by HDAC2, causing aberrant promoter methylation and transcriptional suppression, ultimately leading to increased Ras-GTP activity and activation of common EndMT regulators Twist and Snail. Our studies provide a novel aspect for known adverse effects of high phosphate levels, as eukaryotic cells are commonly believed to have lost phosphate-sensing mechanisms of prokaryotes during evolution, rendering them insensitive to extracellular inorganic orthophosphate. In addition, our studies provide novel insights into the mechanisms underlying specific targeting of select genes in context of fibrogenesis.
Collapse
Affiliation(s)
- Xiaoying Tan
- Department of Cardiology and Pneumology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Xingbo Xu
- Department of Cardiology and Pneumology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Elisabeth M. Zeisberg
- Department of Cardiology and Pneumology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Göttingen, Germany
- * E-mail:
| |
Collapse
|
26
|
|