1
|
Bin S, Yoo M, Molinari P, Gentile M, Budge K, Cantarelli C, Khan Y, La Manna G, Baldwin WM, Dvorina N, Cravedi P, Gusella GL. Reduced decay-accelerating factor expression promotes complement-mediated cystogenesis in murine ADPKD. JCI Insight 2024; 9:e175220. [PMID: 38912583 PMCID: PMC11383362 DOI: 10.1172/jci.insight.175220] [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: 08/30/2023] [Accepted: 05/15/2024] [Indexed: 06/25/2024] Open
Abstract
Patients with autosomal dominant polycystic kidney disease (ADPKD), a genetic disease due to mutations of the PKD1 or PKD2 gene, show signs of complement activation in the urine and cystic fluid, but their pathogenic role in cystogenesis is unclear. We tested the causal relationship between complement activation and cyst growth using a Pkd1KO renal tubular cell line and newly generated conditional Pkd1-/- C3-/- mice. Pkd1-deficient tubular cells have increased expression of complement-related genes (C3, C5, CfB, C3ar, and C5ar1), while the gene and protein expression of complement regulators DAF, CD59, and Crry is decreased. Pkd1-/- C3-/- mice are unable to fully activate the complement cascade and are characterized by a significantly slower kidney cystogenesis, preserved renal function, and reduced intrarenal inflammation compared with Pkd1-/- C3+/+ controls. Transgenic expression of the cytoplasmic C-terminal tail of Pkd1 in Pkd1KO cells lowered C5ar1 expression, restored Daf levels, and reduced cell proliferation. Consistently, both DAF overexpression and pharmacological inhibition of C5aR1 (but not C3aR) reduced Pkd1KO cell proliferation. In conclusion, the loss of Pkd1 promotes unleashed activation of locally produced complement by downregulating DAF expression in renal tubular cells. Increased C5a formation and C5aR1 activation in tubular cells promotes cyst growth, offering a new therapeutic target.
Collapse
Affiliation(s)
- Sofia Bin
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero- University of Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Miran Yoo
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paolo Molinari
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Micaela Gentile
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Unità Operativa Nefrologia, Azienda-Ospedaliero University of Parma, Department of Medicine and Syrgery, University of Parma, Italy
| | - Kelly Budge
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chiara Cantarelli
- Unità Operativa Nefrologia, Azienda-Ospedaliero University of Parma, Department of Medicine and Syrgery, University of Parma, Italy
| | - Yaseen Khan
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gaetano La Manna
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero- University of Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paolo Cravedi
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - G Luca Gusella
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
2
|
Yoo M, Haydak JC, Azeloglu EU, Lee K, Gusella GL. cGAS Activation Accelerates the Progression of Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2024; 35:466-482. [PMID: 38247039 PMCID: PMC11000720 DOI: 10.1681/asn.0000000000000305] [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: 02/02/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
SIGNIFICANCE STATEMENT The renal immune infiltrate observed in autosomal polycystic kidney disease contributes to the evolution of the disease. Elucidating the cellular mechanisms underlying the inflammatory response could help devise new therapeutic strategies. Here, we provide evidence for a mechanistic link between the deficiency polycystin-1 and mitochondrial homeostasis and the activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of the interferon genes (STING) pathway. Our data identify cGAS as an important mediator of renal cystogenesis and suggest that its inhibition may be useful to slow down the disease progression. BACKGROUND Immune cells significantly contribute to the progression of autosomal dominant polycystic kidney disease (ADPKD), the most common genetic disorder of the kidney caused by the dysregulation of the Pkd1 or Pkd2 genes. However, the mechanisms triggering the immune cells recruitment and activation are undefined. METHODS Immortalized murine collecting duct cell lines were used to dissect the molecular mechanism of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) activation in the context of genotoxic stress induced by Pkd1 ablation. We used conditional Pkd1 and knockout cGas-/- genetic mouse models to confirm the role of cGAS/stimulator of the interferon genes (STING) pathway activation on the course of renal cystogenesis. RESULTS We show that Pkd1 -deficient renal tubular cells express high levels of cGAS, the main cellular sensor of cytosolic nucleic acid and a potent stimulator of proinflammatory cytokines. Loss of Pkd1 directly affects cGAS expression and nuclear translocation, as well as activation of the cGAS/STING pathway, which is reversed by cGAS knockdown or functional pharmacological inhibition. These events are tightly linked to the loss of mitochondrial structure integrity and genotoxic stress caused by Pkd1 depletion because they can be reverted by the potent antioxidant mitoquinone or by the re-expression of the polycystin-1 carboxyl terminal tail. The genetic inactivation of cGAS in a rapidly progressing ADPKD mouse model significantly reduces cystogenesis and preserves normal organ function. CONCLUSIONS Our findings indicate that the activation of the cGAS/STING pathway contributes to ADPKD cystogenesis through the control of the immune response associated with the loss of Pkd1 and suggest that targeting this pathway may slow disease progression.
Collapse
Affiliation(s)
- Miran Yoo
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | |
Collapse
|
3
|
Shaikh Qureshi WM, Hentges KE. Functions of cilia in cardiac development and disease. Ann Hum Genet 2024; 88:4-26. [PMID: 37872827 PMCID: PMC10952336 DOI: 10.1111/ahg.12534] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023]
Abstract
Errors in embryonic cardiac development are a leading cause of congenital heart defects (CHDs), including morphological abnormalities of the heart that are often detected after birth. In the past few decades, an emerging role for cilia in the pathogenesis of CHD has been identified, but this topic still largely remains an unexplored area. Mouse forward genetic screens and whole exome sequencing analysis of CHD patients have identified enrichment for de novo mutations in ciliary genes or non-ciliary genes, which regulate cilia-related pathways, linking cilia function to aberrant cardiac development. Key events in cardiac morphogenesis, including left-right asymmetric development of the heart, are dependent upon cilia function. Cilia dysfunction during left-right axis formation contributes to CHD as evidenced by the substantial proportion of heterotaxy patients displaying complex CHD. Cilia-transduced signaling also regulates later events during heart development such as cardiac valve formation, outflow tract septation, ventricle development, and atrioventricular septa formation. In this review, we summarize the role of motile and non-motile (primary cilia) in cardiac asymmetry establishment and later events during heart development.
Collapse
Affiliation(s)
- Wasay Mohiuddin Shaikh Qureshi
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
4
|
Waddell SH, Yao Y, Olaizola P, Walker A, Jarman EJ, Gournopanos K, Gradinaru A, Christodoulou E, Gautier P, Boerrigter MM, Cadamuro M, Fabris L, Drenth JPH, Kendall TJ, Banales JM, Khamseh A, Mill P, Boulter L. A TGFβ-ECM-integrin signaling axis drives structural reconfiguration of the bile duct to promote polycystic liver disease. Sci Transl Med 2023; 15:eabq5930. [PMID: 37703354 PMCID: PMC7615241 DOI: 10.1126/scitranslmed.abq5930] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
The formation of multiple cysts in the liver occurs in a number of isolated monogenic diseases or multisystemic syndromes, during which bile ducts develop into fluid-filled biliary cysts. For patients with polycystic liver disease (PCLD), nonsurgical treatments are limited, and managing life-long abdominal swelling, pain, and increasing risk of cyst rupture and infection is common. We demonstrate here that loss of the primary cilium on postnatal biliary epithelial cells (via the deletion of the cilia gene Wdr35) drives ongoing pathological remodeling of the biliary tree, resulting in progressive cyst formation and growth. The development of cystic tissue requires the activation of transforming growth factor-β (TGFβ) signaling, which promotes the expression of a procystic, fibronectin-rich extracellular matrix and which itself is perceived by a changing profile of integrin receptors on the cystic epithelium. This signaling axis is conserved in liver cysts from patients with either autosomal dominant polycystic kidney disease or autosomal dominant polycystic liver disease, indicating that there are common cellular mechanisms for liver cyst growth regardless of the underlying genetic cause. Cyst number and size can be reduced by inhibiting TGFβ signaling or integrin signaling in vivo. We suggest that our findings represent a therapeutic route for patients with polycystic liver disease, most of whom would not be amenable to surgery.
Collapse
Affiliation(s)
- Scott H Waddell
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Yuelin Yao
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
- School of Informatics- University of Edinburgh- Edinburgh- UK, EH8 9AB
| | - Paula Olaizola
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain, 20014
- Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK EH16 4TJ
| | - Alexander Walker
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Edward J Jarman
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Konstantinos Gournopanos
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Andreea Gradinaru
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Ersi Christodoulou
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Philippe Gautier
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Melissa M Boerrigter
- Department of Gastroenterology and Hepatology, Radboud University, Nijmegen Medical Center- 6525 GA Nijmegen- Netherlands
| | | | - Luca Fabris
- Department of Molecular Medicine, University of Padua, 35128 Padua, Italy
- Digestive Disease Section, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joost PH Drenth
- Department of Gastroenterology and Hepatology, Radboud University, Nijmegen Medical Center- 6525 GA Nijmegen- Netherlands
| | - Timothy J Kendall
- Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK EH16 4TJ
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain, 20014
- National Institute for the Study of Liver and Gastrointestinal Diseases, CIBERehd, “Instituto de Salud Carlos III”, 28029 Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ava Khamseh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
- School of Informatics- University of Edinburgh- Edinburgh- UK, EH8 9AB
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh- Edinburgh- UK, EH4 2XU
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, Edinburgh, UK, EH4 2XU
| |
Collapse
|
5
|
Ateshian GA, Spack KA, Hone JC, Azeloglu EU, Gusella GL. Computational study of biomechanical drivers of renal cystogenesis. Biomech Model Mechanobiol 2023; 22:1113-1127. [PMID: 37024601 PMCID: PMC10524738 DOI: 10.1007/s10237-023-01704-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/12/2023] [Indexed: 04/08/2023]
Abstract
Renal cystogenesis is the pathological hallmark of autosomal dominant polycystic kidney disease, caused by PKD1 and PKD2 mutations. The formation of renal cysts is a common manifestation in ciliopathies, a group of syndromic disorders caused by mutation of proteins involved in the assembly and function of the primary cilium. Cystogenesis is caused by the derailment of the renal tubular architecture and tissue deformation that eventually leads to the impairment of kidney function. However, the biomechanical imbalance of cytoskeletal forces that are altered in cells with Pkd1 mutations has never been investigated, and its nature and extent remain unknown. In this computational study, we explored the feasibility of various biomechanical drivers of renal cystogenesis by examining several hypothetical mechanisms that may promote morphogenetic markers of cystogenesis. Our objective was to provide physics-based guidance for our formulation of hypotheses and our design of experimental studies investigating the role of biomechanical disequilibrium in cystogenesis. We employed the finite element method to explore the role of (1) wild-type versus mutant cell elastic modulus; (2) contractile stress magnitude in mutant cells; (3) localization and orientation of contractile stress in mutant cells; and (4) sequence of cell contraction and cell proliferation. Our objective was to identify the factors that produce the characteristic tubular cystic growth. Results showed that cystogenesis occurred only when mutant cells contracted along the apical-basal axis, followed or accompanied by cell proliferation, as long as mutant cells had comparable or lower elastic modulus than wild-type cells, with their contractile stresses being significantly greater than their modulus. Results of these simulations allow us to focus future in vitro and in vivo experimental studies on these factors, helping us formulate physics-based hypotheses for renal tubule cystogenesis.
Collapse
Affiliation(s)
- Gerard A Ateshian
- Department of Mechanical Engineering, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Katherine A Spack
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - James C Hone
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, New York, NY, USA
- Department of Pharmacological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - G Luca Gusella
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Szaraz D, Danek Z, Lipovy B, Krivanek J, Buchtova M, Moldovan Putnova B, Putnova I, Stembirek J, Andrasina T, Divacka P, Izakovicova Holla L, Borilova Linhartova P. Primary cilia and hypoxia-associated signaling in developmental odontogenic cysts in relation to autosomal dominant polycystic kidney disease - A novel insight. Heliyon 2023; 9:e17130. [PMID: 37389068 PMCID: PMC10300219 DOI: 10.1016/j.heliyon.2023.e17130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023] Open
Abstract
Developmental cysts are pathological epithelial-lined cavities arising in various organs as a result of systemic or hereditary diseases. Molecular mechanisms involved in the formation of developmental odontogenic cysts (OCs) are not fully understood yet; the cystogenesis of renal cysts originating from the autosomal dominant polycystic kidney disease (ADPKD) has been, however, explored in much greater detail. This narrative review aimed i) to summarize molecular and cellular processes involved in the formation and growth of developmental OCs, especially dentigerous cysts (DCs) and odontogenic keratocysts (OKCs), ii) to find if there are any similarities in their cystogenesis to ADPKD cysts, and, based on that, iii) to suggest potential factors, candidate molecules, and mechanisms that could be involved in the DC formation, thus proposing further research directions. Here we suggest a possible association of developmental OCs with primary cilia disruption and with hypoxia, which have been previously linked with cyst formation in ADPKD patients. This is illustrated on the imagery of tissues from an ADPKD patient (renal cyst) and from developmental OCs, supporting the similarities in cell proliferation, apoptosis, and primary cilia distribution in DC/OKC/ADPKD tissues. Based on all that, we propose a novel hypothesis of OCs formation suggesting a crucial role of mutations associated with the signaling pathways of primary cilia (in particular, Sonic Hedgehog). These can lead to excessive proliferation and formation of cell agglomerates, which is followed by hypoxia-driven apoptosis in the centers of such agglomerates (controlled by molecules such as Hypoxia-inducible factor-1 alpha), leading to cavity formation and, finally, the OCs development. Based on this, we propose future perspectives in the investigation of OC pathogenesis.
Collapse
Affiliation(s)
- David Szaraz
- Clinic of Maxillofacial Surgery, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Zdenek Danek
- Clinic of Maxillofacial Surgery, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Bretislav Lipovy
- Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
- Department of Burns and Plastic Surgery, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Marcela Buchtova
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveří 97, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Barbora Moldovan Putnova
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveří 97, 602 00 Brno, Czech Republic
- Department of Pathological Morphology and Parasitology, University of Veterinary Sciences, Palackého tř. 1946/1, 61242 Brno-Královo Pole, Czech Republic
| | - Iveta Putnova
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveří 97, 602 00 Brno, Czech Republic
- Department of Anatomy, Histology and Embryology, University of Veterinary and Pharmaceutical Sciences, Palackého tř. 1946/1, 61242 Brno-Královo Pole, Czech Republic
| | - Jan Stembirek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Veveří 97, 602 00 Brno, Czech Republic
- Clinic of Maxillofacial Surgery, University Hospital Ostrava, 17. Listopadu 1790/5, 70800 Ostrava-Poruba, Czech Republic
| | - Tomas Andrasina
- Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
- Department of Radiology and Nuclear Medicine, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
| | - Petra Divacka
- Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
- Department of Internal Medicine and Gastroenterology, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
| | - Lydie Izakovicova Holla
- Clinic of Stomatology, Institution Shared with St. Anne’s University Hospital, Faculty of Medicine, Masaryk University, Pekarska 664/53, 60200 Brno, Czech Republic
| | - Petra Borilova Linhartova
- Clinic of Maxillofacial Surgery, University Hospital Brno, Jihlavska 20, 62500 Brno, Czech Republic
- Clinic of Stomatology, Institution Shared with St. Anne’s University Hospital, Faculty of Medicine, Masaryk University, Pekarska 664/53, 60200 Brno, Czech Republic
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| |
Collapse
|
7
|
Germano DPJ, Johnston ST, Crampin EJ, Osborne JM. Modelling realistic 3D deformations of simple epithelia in dynamic homeostasis. Math Biosci 2022; 352:108895. [PMID: 36037860 DOI: 10.1016/j.mbs.2022.108895] [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: 05/24/2022] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 10/15/2022]
Abstract
The maintenance of tissue and organ structures during dynamic homeostasis is often not well understood. In order for a system to be stable, cell renewal, cell migration and cell death must be finely balanced. Moreover, a tissue's shape must remain relatively unchanged. Simple epithelial tissues occur in various structures throughout the body, such as the endothelium, mesothelium, linings of the lungs, saliva and thyroid glands, and gastrointestinal tract. Despite the prevalence of simple epithelial tissues, there are few models which accurately describe how these tissues maintain a stable structure. Here, we present a novel, 3D, deformable, multilayer, cell-centre model of a simple epithelium. Cell movement is governed by the minimisation of a bending potential across the epithelium, cell-cell adhesion, and viscous effects. We show that the model is capable of maintaining a consistent tissue structure while undergoing self renewal. We also demonstrate the model's robustness under tissue renewal, cell migration and cell removal. The model presented here is a valuable advancement towards the modelling of tissues and organs with complex and generalised structures.
Collapse
Affiliation(s)
- Domenic P J Germano
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria 3010, Australia; Systems Biology Laboratory, School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria 3010, Australia; Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stuart T Johnston
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria 3010, Australia; Systems Biology Laboratory, School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria 3010, Australia; Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Edmund J Crampin
- Systems Biology Laboratory, School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria 3010, Australia; Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia; School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - James M Osborne
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
8
|
He J, Zhang S, Qiu Z, Li X, Huang H, Jin W, Xu Y, Shao G, Wang L, Meng J, Wang S, Geng X, Jia Y, Li M, Yang B, Jenny Lu HA, Zhou H. Inhibiting Focal Adhesion Kinase Ameliorates Cyst Development in Polycystin-1-Deficient Polycystic Kidney Disease in Animal Model. J Am Soc Nephrol 2021; 32:2159-2174. [PMID: 34465607 PMCID: PMC8729842 DOI: 10.1681/asn.2020111560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/07/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is characterized by numerous cysts originating from renal tubules and is associated with significant tubular epithelial cell proliferation. Focal adhesion kinase (FAK) promotes tumor growth by regulating multiple proliferative pathways. METHODS We established the forskolin (FSK)-induced three-dimensional (3D) Madin-Darby Canine Kidney cystogenesis model and 8-bromoadenosine-3`,5`-cyclic monophosphate-stimulated cyst formation in ex vivo embryonic kidney culture. Cultured human renal cyst-lining cells (OX-161) and normal tubular epithelial cells were treated with FAK inhibitors or transfected with green fluorescent protein-tagged FAK mutant plasmids for proliferation study. Furthermore, we examined the role of FAK in two transgenic ADPKD animal models, the kidney-specific Pkd1 knockout and the collecting duct-specific Pkd1 knockout mouse models. RESULTS FAK activity was significantly elevated in OX-161 cells and in two ADPKD mouse models. Inhibiting FAK activity reduced cell proliferation in OX-161 cells and prevented cyst growth in ex vivo and 3D cyst models. In tissue-specific Pkd1 knockout mouse models, FAK inhibitors retarded cyst development and mitigated renal function decline. Mechanically, FSK stimulated FAK activation in tubular epithelial cells, which was blocked by a protein kinase A (PKA) inhibitor. Inhibition of FAK activation by inhibitors or transfected cells with mutant FAK constructs interrupted FSK-mediated Src activation and upregulation of ERK and mTOR pathways. CONCLUSIONS Our study demonstrates the critical involvement of FAK in renal cyst development, suggests that FAK is a potential therapeutic target in treating patients with ADPKD, and highlights the role of FAK in cAMP-PKA-regulated proliferation.
Collapse
Affiliation(s)
- Jinzhao He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Shun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiwei Qiu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaowei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Huihui Huang
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - William Jin
- Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, Massachusetts
| | - Yue Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guangying Shao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jia Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shuyuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoqiang Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingli Jia
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hua A. Jenny Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| |
Collapse
|
9
|
Lee K, Gusella GL, He JC. Epithelial proliferation and cell cycle dysregulation in kidney injury and disease. Kidney Int 2021; 100:67-78. [PMID: 33831367 PMCID: PMC8855879 DOI: 10.1016/j.kint.2021.03.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023]
Abstract
Various cellular insults and injury to renal epithelial cells stimulate repair mechanisms to adapt and restore the organ homeostasis. Renal tubular epithelial cells are endowed with regenerative capacity, which allows for a restoration of nephron function after acute kidney injury. However, recent evidence indicates that the repair is often incomplete, leading to maladaptive responses that promote the progression to chronic kidney disease. The dysregulated cell cycle and proliferation is also a key feature of renal tubular epithelial cells in polycystic kidney disease and HIV-associated nephropathy. Therefore, in this review, we provide an overview of cell cycle regulation and the consequences of dysregulated cell proliferation in acute kidney injury, polycystic kidney disease, and HIV-associated nephropathy. An increased understanding of these processes may help define better targets for kidney repair and combat chronic kidney disease progression.
Collapse
Affiliation(s)
- Kyung Lee
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - G Luca Gusella
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Cijiang He
- Department of Medicine, Nephrology Division, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Renal Program, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA.
| |
Collapse
|
10
|
Chen YC, Su YC, Shieh GS, Su BH, Su WC, Huang PH, Jiang ST, Shiau AL, Wu CL. Prothymosin α promotes STAT3 acetylation to induce cystogenesis in Pkd1-deficient mice. FASEB J 2019; 33:13051-13061. [PMID: 31589480 DOI: 10.1096/fj.201900504r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Polycystic kidney disease (PKD) is characterized by the expansion of fluid-filled cysts in the kidney, which impair the function of kidney and eventually leads to end-stage renal failure. It has been previously demonstrated that transgenic overexpression of prothymosin α (ProT) induces the development of PKD; however, the underlying mechanisms remain unclear. In this study, we used a mouse PKD model that sustains kidney-specific low-expression of Pkd1 to illustrate that aberrant up-regulation of ProT occurs in cyst-lining epithelial cells, and we further developed an in vitro cystogenesis model to demonstrate that the suppression of ProT is sufficient to reduce cyst formation. Next, we found that the expression of ProT was accompanied with prominent augmentation of protein acetylation in PKD, which results in the activation of downstream signal transducer and activator of transcription (STAT) 3. The pathologic role of STAT3 in PKD has been previously reported. We determined that this molecular mechanism of protein acetylation is involved with the interaction between ProT and STAT3; consequently, it causes the deprivation of histone deacetylase 3 from the indicated protein. Conclusively, these results elucidate the significant role of ProT, including protein acetylation and STAT3 activation in PKD, which represent potential for ameliorating the disease progression of PKD.-Chen, Y.-C., Su, Y.-C., Shieh, G.-S., Su, B.-H., Su, W.-C., Huang, P.-H., Jiang, S.-T., Shiau, A.-L., Wu, C.-L. Prothymosin α promotes STAT3 acetylation to induce cystogenesis in Pkd1-deficient mice.
Collapse
Affiliation(s)
- Yi-Cheng Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chu Su
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Gia-Shing Shieh
- Department of Urology, Tainan Hospital, Ministry of Health and Welfare, Executive Yuan, Tainan, Taiwan
| | - Bing-Hua Su
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Cheng Su
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Hsin Huang
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Si-Tse Jiang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
11
|
Weydert C, Decuypere JP, De Smedt H, Janssens P, Vennekens R, Mekahli D. Fundamental insights into autosomal dominant polycystic kidney disease from human-based cell models. Pediatr Nephrol 2019; 34:1697-1715. [PMID: 30215095 DOI: 10.1007/s00467-018-4057-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
Several animal- and human-derived models are used in autosomal dominant polycystic kidney disease (ADPKD) research to gain insight in the disease mechanism. However, a consistent correlation between animal and human ADPKD models is lacking. Therefore, established human-derived models are relevant to affirm research results and translate findings into a clinical set-up. In this review, we give an extensive overview of the existing human-based cell models. We discuss their source (urine, nephrectomy and stem cell), immortalisation procedures, genetic engineering, kidney segmental origin and characterisation with nephron segment markers. We summarise the most studied pathways and lessons learned from these different ADPKD models. Finally, we issue recommendations for the derivation of human-derived cell lines and for experimental set-ups with these cell lines.
Collapse
Affiliation(s)
- Caroline Weydert
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Jean-Paul Decuypere
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Peter Janssens
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
- Department of Nephrology, University Hospitals Brussels, Brussels, Belgium
| | - Rudi Vennekens
- VIB Center for Brain and Disease Research, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium.
- Department of Pediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
12
|
Villalobos E, Criollo A, Schiattarella GG, Altamirano F, French KM, May HI, Jiang N, Nhi Nguyen NU, Romero D, Roa JC, García L, Diaz-Araya G, Morselli E, Ferdous A, Conway SJ, Sadek HA, Gillette TG, Lavandero S, Hill JA. Fibroblast Primary Cilia Are Required for Cardiac Fibrosis. Circulation 2019; 139:2342-2357. [PMID: 30818997 PMCID: PMC6517085 DOI: 10.1161/circulationaha.117.028752] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 02/07/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND The primary cilium is a singular cellular structure that extends from the surface of many cell types and plays crucial roles in vertebrate development, including that of the heart. Whereas ciliated cells have been described in developing heart, a role for primary cilia in adult heart has not been reported. This, coupled with the fact that mutations in genes coding for multiple ciliary proteins underlie polycystic kidney disease, a disorder with numerous cardiovascular manifestations, prompted us to identify cells in adult heart harboring a primary cilium and to determine whether primary cilia play a role in disease-related remodeling. METHODS Histological analysis of cardiac tissues from C57BL/6 mouse embryos, neonatal mice, and adult mice was performed to evaluate for primary cilia. Three injury models (apical resection, ischemia/reperfusion, and myocardial infarction) were used to identify the location and cell type of ciliated cells with the use of antibodies specific for cilia (acetylated tubulin, γ-tubulin, polycystin [PC] 1, PC2, and KIF3A), fibroblasts (vimentin, α-smooth muscle actin, and fibroblast-specific protein-1), and cardiomyocytes (α-actinin and troponin I). A similar approach was used to assess for primary cilia in infarcted human myocardial tissue. We studied mice silenced exclusively in myofibroblasts for PC1 and evaluated the role of PC1 in fibrogenesis in adult rat fibroblasts and myofibroblasts. RESULTS We identified primary cilia in mouse, rat, and human heart, specifically and exclusively in cardiac fibroblasts. Ciliated fibroblasts are enriched in areas of myocardial injury. Transforming growth factor β-1 signaling and SMAD3 activation were impaired in fibroblasts depleted of the primary cilium. Extracellular matrix protein levels and contractile function were also impaired. In vivo, depletion of PC1 in activated fibroblasts after myocardial infarction impaired the remodeling response. CONCLUSIONS Fibroblasts in the neonatal and adult heart harbor a primary cilium. This organelle and its requisite signaling protein, PC1, are required for critical elements of fibrogenesis, including transforming growth factor β-1-SMAD3 activation, production of extracellular matrix proteins, and cell contractility. Together, these findings point to a pivotal role of this organelle, and PC1, in disease-related pathological cardiac remodeling and suggest that some of the cardiovascular manifestations of autosomal dominant polycystic kidney disease derive directly from myocardium-autonomous abnormalities.
Collapse
Affiliation(s)
- Elisa Villalobos
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Alfredo Criollo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago 8380492, Chile
- Research Institute for Odontology Sciences, Faculty of Odontology, University of Chile, Santiago 8380492, Chile
| | - Gabriele G. Schiattarella
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Kristin M. French
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Herman I. May
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Nan Jiang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Ngoc Uyen Nhi Nguyen
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Diego Romero
- Department of Pathology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 7820436, Chile
| | - Juan Carlos Roa
- Department of Pathology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 7820436, Chile
| | - Lorena García
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontifical Catholic University of Chile, Santiago 7820436, Chile
| | - Anwarul Ferdous
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Simon J. Conway
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202-3082 USA
| | - Hesham A. Sadek
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Sergio Lavandero
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| |
Collapse
|
13
|
Peintner L, Borner C. Role of apoptosis in the development of autosomal dominant polycystic kidney disease (ADPKD). Cell Tissue Res 2017; 369:27-39. [PMID: 28560694 DOI: 10.1007/s00441-017-2628-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a widespread genetic disorder in the Western world and is characterized by cystogenesis that often leads to end-stage renal disease (ESRD). Mutations in the pkd1 gene, encoding for polycystin-1 (PC1) and its interaction partner pkd2, encoding for polycystin-2 (PC2), are the main drivers of this disease. PC1 and PC2 form a multiprotein membrane complex at cilia sites of the plasma membrane and at intracellular membranes. This complex mediates calcium influx and stimulates various signaling pathways regulating cell survival, proliferation and differentiation. The molecular consequences of pkd1 and pkd2 mutations are still a matter of debate. In particular, the ways in which the cysts are initially formed and progress throughout the disease are unknown. The mechanisms proposed to play a role include enhanced cell proliferation, increased apoptotic cell death and diminished autophagy. In this review, we summarize our current understanding about the contribution of apoptosis to cystogenesis and ADPKD. We present the animal models and the tools and methods that have been created to analyze this process. We also critically review the data that are in favor or against the involvement of apoptosis in disease generation. We argue that apoptosis is probably not the sole driver of cystogenesis but that a cooperative action of cell death, compensatory cell proliferation and perturbed autophagy gradually establish the disease. Finally, we propose novel strategies for uncovering the mode of action of PC1 and PC2 and suggest means by which their dysfunction or loss of expression lead to cystogenesis and ADPKD development.
Collapse
Affiliation(s)
- Lukas Peintner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University of Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University of Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany. .,Spemann Graduate School of Biology and Medicine, Albert Ludwigs University of Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany.
| |
Collapse
|
14
|
Bondzie PA, Chen HA, Cao MZ, Tomolonis JA, He F, Pollak MR, Henderson JM. Non-muscle myosin-IIA is critical for podocyte f-actin organization, contractility, and attenuation of cell motility. Cytoskeleton (Hoboken) 2016; 73:377-95. [PMID: 27232264 DOI: 10.1002/cm.21313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 05/20/2016] [Accepted: 05/25/2016] [Indexed: 12/12/2022]
Abstract
Several glomerular pathologies resulting from podocyte injury are linked to genetic variation involving the MYH9 gene, which encodes the heavy chain of non-muscle myosin-IIA (NM-IIA). However, the functional role of NM-IIA has not been studied extensively in podocytes. We hypothesized that NM-IIA is critical for maintenance of podocyte structure and mechanical function. To test this hypothesis, we studied murine podocytes in vitro subjected to blebbistatin inhibition of NM-II activity, or RNA interference-mediated, isoform-specific ablation of Myh9 gene and protein (NM-IIA) or its paralog Myh10 gene and protein (NM-IIB). Using quantitative immunofluorescence microscopy, traction force microscopy, and attachment and "wound healing" assays, we found that NM-IIA ablation altered podocyte actin cytoskeletal structure and focal adhesion distribution, decreased cell attachment and contractility, and increased cell motility. Blebbistatin treatment had similar effects. NM-IIB ablation produced cells that exhibited poor attachment, but cytoskeletal structural organization, contractility and motility were maintained. These findings indicate that NM-IIA is essential for maintenance of podocyte cytoskeletal structure and mechanical function in vitro, and NM-IIB does not replace it in this role when NM-IIA expression is altered. We conclude that critical podocyte functions may be affected by MYH9 mutations or disease-associated haplotypes. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Philip A Bondzie
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Hui A Chen
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Mei Zhen Cao
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Julie A Tomolonis
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Fangfang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Martin R Pollak
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
15
|
Ong ACM, Harris PC. A polycystin-centric view of cyst formation and disease: the polycystins revisited. Kidney Int 2015; 88:699-710. [PMID: 26200945 PMCID: PMC4589452 DOI: 10.1038/ki.2015.207] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/30/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
Abstract
It is 20 years since the identification of PKD1, the major gene mutated in autosomal dominant polycystic kidney disease (ADPKD), followed closely by the cloning of PKD2. These major breakthroughs have led in turn to a period of intense investigation into the function of the two proteins encoded, polycystin-1 and polycystin-2, and how defects in either protein lead to cyst formation and nonrenal phenotypes. In this review, we summarize the major findings in this area and present a current model of how the polycystin proteins function in health and disease.
Collapse
Affiliation(s)
- Albert CM Ong
- Kidney Genetics Group, Academic Nephrology Unit, Department of Infection and Immunity, University of Sheffield Medical School, Sheffield, UK
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Lee K, Boctor S, Barisoni LMC, Gusella GL. Inactivation of integrin-β1 prevents the development of polycystic kidney disease after the loss of polycystin-1. J Am Soc Nephrol 2014; 26:888-95. [PMID: 25145933 DOI: 10.1681/asn.2013111179] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Dysregulation of polycystin-1 (PC1) leads to autosomal dominant polycystic kidney disease (ADPKD), a disorder characterized by the formation of multiple bilateral renal cysts, the progressive accumulation of extracellular matrix (ECM), and the development of tubulointerstitial fibrosis. Correspondingly, cystic epithelia express higher levels of integrins (ECM receptors that control various cellular responses, such as cell proliferation, migration, and survival) that are characteristically altered in cystic cells. To determine whether the altered expression of ECM and integrins could establish a pathologic autostimulatory loop, we tested the role of integrin-β1 in vitro and on the cystic development of ADPKD in vivo. Compared with wild-type cells, PC1-depleted immortalized renal collecting duct cells had higher levels of integrin-β1 and fibronectin and displayed increased integrin-mediated signaling in the presence of Mn(2+). In mice, conditional inactivation of integrin-β1 in collecting ducts resulted in a dramatic inhibition of Pkd1-dependent cystogenesis with a concomitant suppression of fibrosis and preservation of normal renal function. Our data provide genetic evidence that a functional integrin-β1 is required for the early events leading to renal cystogenesis in ADPKD and suggest that the integrin signaling pathway may be an effective therapeutic target for slowing disease progression.
Collapse
Affiliation(s)
- Kyung Lee
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | - Sylvia Boctor
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | | | - G Luca Gusella
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and
| |
Collapse
|
17
|
Parallel analysis of mRNA and microRNA microarray profiles to explore functional regulatory patterns in polycystic kidney disease: using PKD/Mhm rat model. PLoS One 2013; 8:e53780. [PMID: 23326503 PMCID: PMC3542345 DOI: 10.1371/journal.pone.0053780] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/03/2012] [Indexed: 01/08/2023] Open
Abstract
Autosomal polycystic kidney disease (ADPKD) is a frequent monogenic renal disease, characterised by fluid-filled cysts that are thought to result from multiple deregulated pathways such as cell proliferation and apoptosis. MicroRNAs (miRNAs) are small non-coding RNAs that regulate the expression of many genes associated with such biological processes and human pathologies. To explore the possible regulatory role of miRNAs in PKD, the PKD/Mhm (cy/+) rat, served as a model to study human ADPKD. A parallel microarray-based approach was conducted to profile the expression changes of mRNAs and miRNAs in PKD/Mhm rats. 1,573 up- and 1,760 down-regulated genes were differentially expressed in PKD/Mhm. These genes are associated with 17 pathways (such as focal adhesion, cell cycle, ECM-receptor interaction, DNA replication and metabolic pathways) and 47 (e.g., cell proliferation, Wnt and Tgfβ signaling) Gene Ontologies. Furthermore, we found the similar expression patterns of deregulated genes between PKD/Mhm (cy/+) rat and human ADPKD, PKD1L3/L3, PKD1−/−, Hnf1α-deficient, and Glis2lacZ/lacZ models. Additionally, several differentially regulated genes were noted to be target hubs for miRNAs. We also obtained 8 significantly up-regulated miRNAs (rno-miR-199a-5p, −214, −146b, −21, −34a, −132, −31 and −503) in diseased kidneys of PKD/Mhm rats. Additionally, the binding site overrepresentation and pathway enrichment analyses were accomplished on the putative targets of these 8 miRNAs. 7 out of these 8 miRNAs and their possible interactions have not been previously described in ADPKD. We have shown a strong overlap of functional patterns (pathways) between deregulated miRNAs and mRNAs in the PKD/Mhm (cy/+) rat model. Our findings suggest that several miRNAs may be associated in regulating pathways in ADPKD. We further describe novel miRNAs and their possible targets in ADPKD, which will open new avenues to understand the pathogenesis of human ADPKD. Furthermore they could serve as a useful resource for anti-fibrotic therapeutics.
Collapse
|
18
|
Patel V, Hajarnis S, Williams D, Hunter R, Huynh D, Igarashi P. MicroRNAs regulate renal tubule maturation through modulation of Pkd1. J Am Soc Nephrol 2012; 23:1941-8. [PMID: 23138483 DOI: 10.1681/asn.2012030321] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) contribute to the regulation of early kidney development, but their role during later stages of renal tubule maturation is not well understood. Here, we found that ablation of the miRNA-processing enzyme Dicer from maturing renal tubules produces tubular and glomerular cysts in mice. Inactivation of Dicer is associated with downregulation of miR-200, a kidney-enriched miRNA family, and upregulation of the polycystic kidney disease gene Pkd1. Inhibition of miR-200 in cultured renal epithelial cells disrupted tubulogenesis and led to upregulation of Pkd1. Using bioinformatic and in vitro approaches, we found that miR-200b/c/429 induce post-transcriptional repression of Pkd1 through two conserved binding sites in the 3'-Untranslated regions of Pkd1. Overexpression of PKD1 in renal epithelial cells was sufficient to disrupt tubulogenesis and produce cyst-like structures. In conclusion, miRNAs are essential for the maturation of renal tubules, and Pkd1 is a target of miR-200. These results also suggest that miRNAs may modulate PKD1 gene dosage and play a role in the initiation of cystogenesis.
Collapse
Affiliation(s)
- Vishal Patel
- Department of Internal Medicine/Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8856, Dallas, TX 75390, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Subramanian B, Ko WC, Yadav V, DesRochers TM, Perrone RD, Zhou J, Kaplan DL. The regulation of cystogenesis in a tissue engineered kidney disease system by abnormal matrix interactions. Biomaterials 2012; 33:8383-94. [PMID: 22940218 DOI: 10.1016/j.biomaterials.2012.08.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 08/10/2012] [Indexed: 11/17/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) remains a major health care concern affecting several million patients worldwide and for which there is no specific treatment. We have employed a 3D tissue engineered disease-like system to emulate cystic structures in vitro and analyzed the extracellular matrix (ECM) interactions in it. The tissue system was developed by culturing normal or polycystin-1 silenced mouse Inner Medullary Collecting Duct (mIMCD) cells in ECM infused into 3D porous silk protein biomaterial scaffolds. In this system, the silk scaffolds provide slow degradation, biocompatibility, and maintain structure and transport for the 3D system, while the ECM molecules retain biological signaling. Using this 3D tissue system we provide evidence for an autocrine signaling loop involving abnormal matrix deposition (collagen type IV and laminin) and its integrin receptor subunit protein (Integrin-β1) in Pkd1 silenced mIMCD cells. In addition, we report that abnormal pericystic ECM interactions between matrix molecules and integrin subunit proteins regulate the rate of cystogenesis in the disease system. Molecular signaling showed abnormalities in cyclin proteins and cell-cycle progression upon Pkd1 knockdown. Importantly, disruption of the abnormal matrix interactions by an additional knockdown (double-silencing) of integrin-β1 in Pkd1 silenced cells reversed the abnormalities and reduced the rate of cystogenesis. Together, these findings indicate that abnormal matrix deposition and altered integrin profile distribution as observed in ADPKD and are critical in cystogenesis and should be considered a target for the development of therapeutics.
Collapse
|
20
|
Seeger-Nukpezah T, Golemis EA. The extracellular matrix and ciliary signaling. Curr Opin Cell Biol 2012; 24:652-61. [PMID: 22819513 DOI: 10.1016/j.ceb.2012.06.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/29/2012] [Accepted: 06/11/2012] [Indexed: 12/24/2022]
Abstract
The primary cilium protrudes like an antenna from the cell surface, sensing mechanical and chemical cues provided in the cellular environment. In some tissue types, ciliary orientation to lumens allows response to fluid flow; in others, such as bone, ciliary protrusion into the extracellular matrix allows response to compression forces. The ciliary membrane contains receptors for Hedgehog, Wnt, Notch, and other potent growth factors, and in some instances also harbors integrin and cadherin family members, allowing receipt of a robust range of signals. A growing list of ciliopathies, arising from deficient formation or function of cilia, includes both developmental defects and chronic, progressive disorders such as polycystic kidney disease (PKD); changes in ciliary function have been proposed to support cancer progression. Recent findings have revealed extensive signaling dialog between cilia and extracellular matrix (ECM), with defects in cilia associated with fibrosis in multiple contexts. Further, a growing number of proteins have been determined to possess multiple roles in control of cilia and focal adhesion interactions with the ECM, further coordinating functionality. We summarize and discuss these recent findings.
Collapse
Affiliation(s)
- Tamina Seeger-Nukpezah
- Program in Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | |
Collapse
|
21
|
Parsi S, Soltani BM, Hosseini E, Tousi SE, Mowla SJ. Experimental verification of a predicted intronic microRNA in human NGFR gene with a potential pro-apoptotic function. PLoS One 2012; 7:e35561. [PMID: 22558167 PMCID: PMC3338703 DOI: 10.1371/journal.pone.0035561] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 03/20/2012] [Indexed: 12/12/2022] Open
Abstract
Neurotrophins (NTs) are a family of secreted growth factor proteins primarily involved in the regulation of survival and appropriate development of neural cells, functioning by binding to their specific (TrkA, TtkB, and TrkC) and/or common NGFR receptor. NGFR is the common receptor of NTs, binding with low-affinity to all members of the family. Among different functions assigned to NGFR, it is also involved in apoptosis induction and tumorigenesis processes. Interestingly, some of the functions of NGFR appear to be ligand-independent, suggesting a probable involvement of non-coding RNA residing within the sequence of the gene. Here, we are reporting the existence of a conserved putative microRNA, named Hsa-mir-6165 [EBI accession#: FR873488]. Transfection of a DNA segment corresponding to the pre-mir-6165 sequence in Hela cell line caused the generation of mature exogenous mir-6165 (a ∼200,000 fold overexpression). Furthermore, using specific primers, we succeeded to detect the endogenous expression of mir-6165 in several glioma cell lines and glioma primary tumors known to express NGFR. Similar to the pro-apoptotic role of NGFR in some cell types, overexpression of pre-mir-6165 in U87 cell line resulted in an elevated rate of apoptosis. Moreover, coordinated with the increased level of mir-6165 in the transfected U87 cell line, two of its predicted target genes (Pkd1 and DAGLA) were significantly down-regulated. The latter findings suggest that some of the previously attributed functions of NGFR could be explained indirectly by co-transcription of mir-6165 in the cells.
Collapse
Affiliation(s)
- Sepideh Parsi
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M. Soltani
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ebrahim Hosseini
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samaneh E. Tousi
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed J. Mowla
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
22
|
Zhou H, Gao J, Zhou L, Li X, Li W, Li X, Xia Y, Yang B. Ginkgolide B inhibits renal cyst development in in vitro and in vivo cyst models. Am J Physiol Renal Physiol 2012; 302:F1234-42. [PMID: 22338085 DOI: 10.1152/ajprenal.00356.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common inherited disease characterized by massive enlargement of fluid-filled cysts in the kidney. However, there is no effective therapy yet for this disease. To examine whether ginkgolide B, a natural compound, inhibits cyst development, a Madin-Darby canine kidney (MDCK) cyst model, an embryonic kidney cyst model, and a PKD mouse model were used. Interestingly, ginkgolide B significantly inhibited MDCK cyst formation dose dependently, with up to 69% reduction by 2 μM ginkgolide B. Ginkgolide B also significantly inhibited cyst enlargement in the MDCK cyst model, embryonic kidney cyst model, and PKD mouse model. To determine the underlying mechanisms, the effect of ginkgolide B on MDCK cell viability, proliferation, apoptosis, chloride transporter CFTR activity, and intracellular signaling pathways were also studied. Ginkgolide B did not affect cell viability, proliferation, and expression and activity of the chloride transporter CFTR that mediates cyst fluid secretion. Ginkgolide B induced cyst cell differentiation and altered the Ras/MAPK signaling pathway. Taken together, our results demonstrate that ginkgolide B inhibits renal cyst formation and enlargement, suggesting that ginkgolide B might be developed into a novel candidate drug for ADPKD.
Collapse
Affiliation(s)
- Hong Zhou
- Dept. of Pharmacology. School of Basic Medical Sciences, Peking Univ., 38 Xueyuan Lu, Haidian District, Beijing 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Apico-basal polarity in polycystic kidney disease epithelia. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1239-48. [DOI: 10.1016/j.bbadis.2011.05.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/19/2011] [Accepted: 05/24/2011] [Indexed: 12/29/2022]
|
24
|
Lee K, Battini L, Gusella GL. Cilium, centrosome and cell cycle regulation in polycystic kidney disease. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1812:1263-71. [PMID: 21376807 PMCID: PMC3138898 DOI: 10.1016/j.bbadis.2011.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 01/10/2011] [Accepted: 02/16/2011] [Indexed: 12/19/2022]
Abstract
Polycystic kidney disease is the defining condition of a group of common life-threatening genetic disorders characterized by the bilateral formation and progressive expansion of renal cysts that lead to end stage kidney disease. Although a large body of information has been acquired in the past years about the cellular functions that characterize the cystic cells, the mechanisms triggering the cystogenic conversion are just starting to emerge. Recent findings link defects in ciliary functions, planar cell polarity pathway, and centrosome integrity in early cystic development. Many of the signals dysregulated during cystogenesis may converge on the centrosome for its central function as a structural support for cilia formation and a coordinator of protein trafficking, polarity, and cell division. Here, we will discuss the contribution of proliferation, cilium and planar cell polarity to the cystic signal and will analyze in particular the possible role that the basal bodies/centrosome may play in the cystogenetic mechanisms. This article is part of a Special Issue entitled: Polycystic Kidney Disease.
Collapse
Affiliation(s)
- Kyung Lee
- Department of Medicine, The Mount Sinai School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
25
|
Elliott J, Zheleznova NN, Wilson PD. c-Src inactivation reduces renal epithelial cell-matrix adhesion, proliferation, and cyst formation. Am J Physiol Cell Physiol 2011; 301:C522-9. [PMID: 21508333 DOI: 10.1152/ajpcell.00163.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
c-Src is a non-receptor tyrosine kinase whose activity is induced by phosphorylation at Y418 and translocation from the cytoplasm to the cell membrane. Increased activity of c-Src has been associated with cell proliferation, matrix adhesion, motility, and apoptosis in tumors. Immunohistochemistry suggested that activated (pY(418))-Src activity is increased in cyst-lining autosomal dominant polycystic kidney disease (ADPKD) epithelial cells in human and mouse ADPKD. Western blot analysis showed that SKI-606 (Wyeth) is a specific inhibitor of pY(418)-Src without demonstrable effects on epidermal growth factor receptor or ErbB2 activity in renal epithelia. In vitro studies on mouse inner medullary collecting duct (mIMCD) cells and human ADPKD cyst-lining epithelial cells showed that SKI-606 inhibited epithelial cell proliferation over a 24-h time frame. In addition, SKI-606 treatment caused a striking statistically significant decrease in adhesion of mIMCD and human ADPKD to extracellular collagen matrix. Retained viability of unattached cells was consistent with a primary effect on epithelial cell anchorage dependence mediated by the loss of extracellular matrix (ECM)-attachment due to α(2)β(1)-integrin function. SKI-606-mediated attenuation of the human ADPKD hyperproliferative and hyper-ECM-adhesive epithelial cell phenotype in vitro was paralleled by retardation of the renal cystic phenotype of Pkd1 orthologous ADPKD heterozygous mice in vivo. This suggests that SKI-606 has dual effects on cystic epithelial cell proliferation and ECM adhesion and may have therapeutic potential for ADPKD patients.
Collapse
Affiliation(s)
- Justine Elliott
- Division of Nephrology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
26
|
Primary cilia dynamics instruct tissue patterning and repair of corneal endothelium. Proc Natl Acad Sci U S A 2011; 108:2819-24. [PMID: 21285373 DOI: 10.1073/pnas.1016702108] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Primary cilia are required for several signaling pathways, but their function in cellular morphogenesis is poorly understood. Here we show that emergence of an hexagonal cellular pattern during development of the corneal endothelium (CE), a monolayer of neural crest-derived cells that maintains corneal transparency, depends on a precise temporal control of assembly of primary cilia that subsequently disassemble in adult corneal endothelial cells (CECs). However, cilia reassembly occurs rapidly in response to an in vivo mechanical injury and precedes basal body polarization and cellular elongation in mature CECs neighboring the wound. In contrast, CE from hypomorphic IFT88 mutants (Tg737(orpk)) or following in vivo lentiviral-mediated IFT88 knockdown display dysfunctional cilia and show disorganized patterning, mislocalization of junctional markers, and accumulation of cytoplasmic acetylated tubulin. Our results indicate an active role of cilia in orchestrating coordinated morphogenesis of CECs during development and repair and define the murine CE as a powerful in vivo system to study ciliary-based cellular dynamics.
Collapse
|
27
|
Gao J, Zhou H, Lei T, Zhou L, Li W, Li X, Yang B. Curcumin inhibits renal cyst formation and enlargement in vitro by regulating intracellular signaling pathways. Eur J Pharmacol 2010; 654:92-9. [PMID: 21187084 DOI: 10.1016/j.ejphar.2010.12.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2010] [Revised: 12/08/2010] [Accepted: 12/11/2010] [Indexed: 02/02/2023]
Abstract
Autosomal dominant polycystic kidney disease, a common inherited disease affecting about 1/1000 and 1/400 live births, is characterized by massive enlargement of fluid-filled cysts and eventually causes renal failure. The purpose of this study is to identify the inhibitory effect of curcumin on renal cyst development and to investigate the inhibitory mechanism. Madin-Darby canine kidney (MDCK) cyst model and murine embryonic kidney cyst model were used to evaluate inhibitory activity. Cell viability, proliferation, apoptosis, CFTR function and expression, and signaling pathways in MDCK cells were determined to explore the mechanism of cyst inhibition. Curcumin was found to significantly inhibit MDCK cyst development. At maximum dose curcumin caused 62% inhibition of the cyst formation (IC(50) was 0.12 μM). Curcumin slowed cyst enlargement in both MDCK cyst model and embryonic kidney cyst model with dose-response relationship. Curcumin neither induced cytotoxicity nor apoptosis in MDCK cells at <100 μM. Curcumin failed to affect the chloride transporter CFTR expression and function. Interestingly, curcumin inhibited forskolin-promoted cell proliferation and promoted the tubule formation in MDCK cells, which indicates curcumin promotes MDCK cell differentiation. Furthermore, curcumin reduced the intracellular signaling proteins Ras, B-raf, p-MEK, p-ERK, c-fos, Egr-1, but increased Raf-1 and NAB2 in MDCK cells exposed to forskolin. These results define that curcumin inhibits renal cyst formation and enlargement and suggest that curcumin might be developed as a candidate drug for polycystic kidney disease.
Collapse
Affiliation(s)
- Jinsheng Gao
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Flores D, Battini L, Gusella GL, Rohatgi R. Fluid shear stress induces renal epithelial gene expression through polycystin-2-dependent trafficking of extracellular regulated kinase. Nephron Clin Pract 2010; 117:p27-36. [PMID: 21109758 DOI: 10.1159/000321640] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 09/20/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The cilium and cilial proteins have emerged as principal mechanosensors of renal epithelial cells responsible for translating mechanical forces into intracellular signals. Polycystin-2 (PC-2), a cilial protein, regulates flow/shear-induced changes in intracellular Ca(2+) ([Ca(2+)](i)) and recently has been implicated in the regulation of mitogen-activated protein (MAP) kinases. We hypothesize that fluid shear stress (FSS) activates PC-2 which regulates MAP kinase and, in turn, induces MAP kinase-dependent gene expression, specifically, monocyte chemoattractant protein-1 (MCP-1). METHODS To test this, PC-2 expression was constitutively reduced in a murine inner medullary collecting duct (IMCD3) cell line, and the expression of FSS-induced MCP-1 expression and MAP kinase signaling compared between the parental (PC-2-expressing) and PC-2-deficient IMCD3 cells. RESULTS FSS induces MAP kinase signaling and downstream MCP-1 mRNA expression in wild-type IMCD3 cells, while inhibitors of MAP kinase prevented the FSS-induced MCP-1 mRNA response. In contradistinction, FSS did not induce MCP-1 mRNA expression in PC-2-deficient cells, but did increase activation of the upstream MAP kinases. Wild-type cells exposed to FSS augmented the nuclear abundance of activated MAP kinase while PC-2-deficient cells did not. CONCLUSIONS PC-2 regulates FSS-induced MAP kinase trafficking into the nucleus of CD cells.
Collapse
Affiliation(s)
- Daniel Flores
- Department of Medicine, The Mount Sinai School of Medicine, New York, NY 10000209, USA
| | | | | | | |
Collapse
|
29
|
Kong T, Xu D, Tran M, Denker BM. Regulation of integrin expression by Gα12: An additional potential mechanism modulating cell attachment. Cell Adh Migr 2010; 4:372-6. [PMID: 20424510 DOI: 10.4161/cam.4.3.11639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Integrins regulate cell attachment and migration through interactions with specific proteins in the extra-cellular matrix. Heterotrimeric G proteins are essential signal transduction proteins that intersect with integrin signaling to regulate fundamental cellular behaviors. Although integrin and G protein signaling often act in concert, how these mechanisms interact in epithelial cells has not been extensively studied. We recently reported Gα12 regulation of epithelial cell attachment and migration on collagen-I through α2β1 integrins (Kong et al. Mol Biol Cell 2009). Activated Gα12 inhibited α2β1 integrin functions through an inside-out signaling mechanism that involved Rho, Src and protein phosphatases without affecting α2 or β1 expression. Activated Gα12 prevented tubulogenesis in 3D-MDCK cell cultures and promoted the formation of cystic structures. Herein, we extend these findings to show Gα12-stimulated transcriptional changes in integrin expression that affect MDCK cell attachment. Based on results from a microarray with MDCK cells expressing constitutively active Gα12 (QLα12), we confirmed with real time PCR that expressing QLα12 led to a 4-fold inhibition of α6 mRNA expression. Cell surface expression and total α6 protein was reduced by FACS and immunofluorescence. QLα12 expressing MDCK cells also revealed less attachment to laminin-5, an α6 integrin ligand. Taken together, G proteins regulate integrins through canonical signaling pathways and potentially regulate integrin expression levels to modulate cellular responses in a variety of pathophysiologic conditions including polycystic kidney disease.
Collapse
Affiliation(s)
- Tianqing Kong
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
30
|
Yu W, Kong T, Beaudry S, Tran M, Negoro H, Yanamadala V, Denker BM. Polycystin-1 protein level determines activity of the Galpha12/JNK apoptosis pathway. J Biol Chem 2010; 285:10243-51. [PMID: 20106977 DOI: 10.1074/jbc.m109.070821] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mutations in PKD1 are the most common cause of autosomal dominant polycystic kidney disease (ADPKD). The protein product of PKD1 (polycystin-1 (PC1)) is a large transmembrane protein with a short intracellular C terminus that interacts with numerous signaling molecules, including Galpha(12). Cyst formation in ADPKD results from numerous cellular defects, including abnormal cilia, changes in polarity, and dysregulated apoptosis and proliferation. Recently, we reported increased apoptosis in Madin-Darby canine kidney (MDCK) cells through Galpha(12) stimulation of JNK and degradation of the anti-apoptotic protein Bcl-2 (Yanamadala, V., Negoro, H., Gunaratnam, L., Kong, T., and Denker, B. M. (2007) J. Biol. Chem. 282, 24352-24363). Herein, we confirm this pathway in Galpha(12)-silenced MDCK cells and utilize MDCK cell lines harboring either overexpressed or silenced PC1 to demonstrate that PC1 expression levels determine activity of the JNK/Bcl-2 apoptosis pathway. PC1-overexpressing MDCK cells were resistant to thrombin/Galpha(12)-stimulated apoptosis, JNK activation, and Bcl-2 degradation. In contrast, PC1-silenced MDCK cells displayed enhanced thrombin-induced apoptosis, JNK activity, and Bcl-2 degradation. In pulldown experiments, PC1 bound to Galpha(12), but not the related Galpha(13) subunit, and thrombin-stimulated MDCK cells led to increased interaction of Galpha(12) with the PC1 C terminus. In transient transfection assays, a PC1 C-terminal mutant lacking the G protein-binding domain was uncoupled from PC1-inhibited apoptosis. PC1 expression levels may be increased or decreased in ADPKD, and these findings suggest a mechanism in which levels of PC1 expression modulate Galpha(12)/JNK-stimulated apoptosis. Taken together, these findings are consistent with a set point model in which PC1 expression levels regulate specific G protein signaling pathways important to cyst development.
Collapse
Affiliation(s)
- Wanfeng Yu
- Renal Division, Brigham and Women's Hospital, Harvard Institutes of Medicine, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Israeli S, Amsler K, Zheleznova N, Wilson PD. Abnormalities in focal adhesion complex formation, regulation, and function in human autosomal recessive polycystic kidney disease epithelial cells. Am J Physiol Cell Physiol 2009; 298:C831-46. [PMID: 19923420 DOI: 10.1152/ajpcell.00032.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Integrin-associated focal adhesion complex formation and turnover plays an essential role in directing interactions between epithelial cells and the extracellular matrix during organogenesis, leading to appropriate cell spreading, cell-matrix adhesion, and migration. Autosomal recessive polycystic kidney disease (ARPKD) is associated with loss of function of PKHD1-encoded protein fibrocystin-1 and is characterized by cystic dilation of renal collecting tubules (CT) in utero and loss of renal function in patients if they survive the perinatal period. Normal polycystin-1 (PC-1)/focal adhesion complex function is required for control of CT diameter during renal development, and abnormalities in these complexes have been demonstrated in human PC-1 mutant cystic cells. To determine whether loss of fibrocystin-1 was associated with focal adhesion abnormalities, ARPKD cells or normal age-matched human fetal (HF)CT cells in which fibrocystin-1 had been decreased by 85% by small interfering RNA inhibition were compared with normal HFCT. Accelerated attachment and spreading on collagen matrix and decreased motility of fibrocystin-1-deficient cells were associated with longer paxillin-containing focal adhesions, more complex actin-cytoskeletal rearrangements, and increased levels of total beta(1)-integrin, c-Src, and paxillin. Immunoblot analysis of adhesive cells using site-specific phospho-antibodies demonstrated ARPKD-associated loss of activation of focal adhesion kinase (FAK) by phosphorylation at its autophosphorylation site (Y397); accelerated FAK inhibition by phosphorylation at Y407, S843, and S910; as well as increased activation of c-Src at pY418. Paxillin coimmunoprecipitation analyses suggested that fibrocystin-1 was a component of the normal focal adhesion complex and that actin and fibrocystin-1 were lost from ARPKD complexes.
Collapse
Affiliation(s)
- Sharon Israeli
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | |
Collapse
|
32
|
Kong T, Xu D, Yu W, Takakura A, Boucher I, Tran M, Kreidberg JA, Shah J, Zhou J, Denker BM. G alpha 12 inhibits alpha2 beta1 integrin-mediated Madin-Darby canine kidney cell attachment and migration on collagen-I and blocks tubulogenesis. Mol Biol Cell 2009; 20:4596-610. [PMID: 19776354 DOI: 10.1091/mbc.e09-03-0220] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Regulation of epithelial cell attachment and migration are essential for normal development and maintenance of numerous tissues. G proteins and integrins are critical signaling proteins regulating these processes, yet in polarized cells little is known about the interaction of these pathways. Herein, we demonstrate that G alpha 12 inhibits interaction of MDCK cells with collagen-I, the major ligand for alpha2 beta1 integrin. Activating G alpha 12 (QL point mutation or stimulating endogenous G alpha 12 with thrombin) inhibited focal adhesions and lamellipodia formation and led to impaired cell migration. Consistent with G alpha 12-regulated attachment to collagen-I, G alpha 12-silenced MDCK cells revealed a more adherent phenotype. Inhibiting Rho kinase completely restored normal attachment in G alpha 12-activated cells, and there was partial recovery with inhibition of Src and protein phosphatase pathways. G alpha 12 activation led to decreased phosphorylation of focal adhesion kinase and paxillin with displacement of alpha2 integrin from the focal adhesion protein complex. Using the MDCK cell 3D-tubulogenesis assay, activated G alpha 12 inhibited tubulogenesis and led to the formation of cyst-like structures. Furthermore, G alpha 12-silenced MDCK cells were resistant to thrombin-stimulated cyst development. Taken together, these studies provide direct evidence for G alpha 12-integrin regulation of epithelial cell spreading and migration necessary for normal tubulogenesis.
Collapse
Affiliation(s)
- Tianqing Kong
- Renal Division, Brigham and Women's Hospital, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ossipova O, Ezan J, Sokol SY. PAR-1 phosphorylates Mind bomb to promote vertebrate neurogenesis. Dev Cell 2009; 17:222-33. [PMID: 19686683 PMCID: PMC2849776 DOI: 10.1016/j.devcel.2009.06.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 04/04/2009] [Accepted: 06/17/2009] [Indexed: 12/25/2022]
Abstract
Generation of neurons in the vertebrate central nervous system requires a complex transcriptional regulatory network and signaling processes in polarized neuroepithelial progenitor cells. Here we demonstrate that neurogenesis in the Xenopus neural plate in vivo and mammalian neural progenitors in vitro involves intrinsic antagonistic activities of the polarity proteins PAR-1 and aPKC. Furthermore, we show that Mind bomb (Mib), a ubiquitin ligase that promotes Notch ligand trafficking and activity, is a crucial molecular substrate for PAR-1. The phosphorylation of Mib by PAR-1 results in Mib degradation, repression of Notch signaling, and stimulation of neuronal differentiation. These observations suggest a conserved mechanism for neuronal fate determination that might operate during asymmetric divisions of polarized neural progenitor cells.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
34
|
Li X, Hyink DP, Radbill B, Sudol M, Zhang H, Zheleznova NN, Wilson PD. Protein kinase-X interacts with Pin-1 and Polycystin-1 during mouse kidney development. Kidney Int 2009; 76:54-62. [DOI: 10.1038/ki.2009.95] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
35
|
Abstract
Autosomal dominant polycystic kidney disease is the most prevalent, potentially lethal monogenic disorder. It has large inter- and intra-familial variability explained to a large extent by its genetic heterogeneity and modifier genes. An increased understanding of its underlying genetic, molecular, and cellular mechanisms and a better appreciation of its progression and systemic manifestations have laid out the foundation for the development of clinical trials and potentially effective therapies. The purpose of this review is to update the core of knowledge in this area with recent publications that have appeared during 2006-2009.
Collapse
Affiliation(s)
- Vicente E Torres
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | |
Collapse
|
36
|
Battini L, Macip S, Fedorova E, Dikman S, Somlo S, Montagna C, Gusella GL. Loss of polycystin-1 causes centrosome amplification and genomic instability. Hum Mol Genet 2008; 17:2819-33. [PMID: 18566106 PMCID: PMC2722891 DOI: 10.1093/hmg/ddn180] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2008] [Accepted: 06/16/2008] [Indexed: 02/06/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenetic disease predominantly caused by alteration or dysregulation of the PKD1 gene, which encodes polycystin-1 (PC1). The disease is characterized by the progressive expansion of bilateral fluid-filled renal cysts that ultimately lead to renal failure. Individual cysts, even within patients with germline mutations, are genetically heterogeneous, displaying diverse chromosomal abnormalities. To date, the molecular mechanisms responsible for this genetic heterogeneity remain unknown. Using a lentiviral-mediated siRNA expression model of Pkd1 hypomorphism, we show that loss of PC1 function is sufficient to produce centrosome amplification and multipolar spindle formation. These events lead to genomic instability characterized by gross polyploidism and mitotic catastrophe. Following these dramatic early changes, the cell population rapidly converges toward a stable ploidy in which centrosome amplification is significantly decreased, though cytological abnormalities such as micronucleation, chromatin bridges and aneuploidy remain common. In agreement with our in vitro findings, we provide the first in vivo evidence that significant centrosome amplification occurs in kidneys from conditional Pkd1 knockout mice at early and late time during the disease progression as well as in human ADPKD patients. These findings establish a novel function of PC1 in ADPKD pathogenesis and a genetic mechanism that may underlie the intrafamilial variability of ADPKD progression.
Collapse
Affiliation(s)
| | | | | | - Steven Dikman
- Department of Pathology, Mount Sinai School of Medicine, One Gustave Levy Place, Box 1243New York, NY 10029, USA
| | - Stefan Somlo
- Department of Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Cristina Montagna
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY,USA
| | | |
Collapse
|
37
|
Zeltner R, Hilgers KF, Schmieder RE, Porst M, Schulze BD, Hartner A. A promoter polymorphism of the alpha 8 integrin gene and the progression of autosomal-dominant polycystic kidney disease. Nephron Clin Pract 2008; 108:c169-75. [PMID: 18277079 DOI: 10.1159/000116887] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Accepted: 10/15/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Dysregulation of integrins is a feature of tissue remodeling in autosomal-dominant polycystic kidney disease (ADPKD). The alpha 8 beta 1 integrin (alpha8beta1) affects kidney development and the susceptibility to renal injury in mice. We investigated whether the -414 T/C polymorphism in the promoter region of the alpha 8 integrin chain gene (ITGA8) is associated with the progression of renal disease in ADPKD. METHODS Genotyping for the -414 T/C polymorphism was performed by allelic separation using RT-PCR in 294 patients with ADPKD. Alpha 8 integrin expression was detected by RT-PCR and immunohistochemistry. RESULTS 41% of the study population reached end stage renal disease at a mean age of 51 +/- 12 years. The frequency of the -414 C allele was 0.194 in ADPKD. C allele carriers (CC and TC genotypes) were compared with patients homozygous for the T allele (TT genotype). Kaplan-Meier analysis revealed that end-stage renal failure occurred at a significantly younger age in TT homozygotes (median age, 47 years; 95% CI, 46-49 years) than in C allele carriers (median age, 51 years; 95% CI, 49-53 years; p = 0.046 by the log-rank test). When parameters of ADPKD patients were compared between genotype by analysis of variance, only age at onset of end-stage renal failure was significantly different (p = 0.026) whereas age at onset of hypertension, body surface area, 24-hour systolic and diastolic blood pressure did not differ. In kidneys of ADPKD, expression of alpha 8 integrin is increased and found de novo in cystic epithelia. CONCLUSION A polymorphism of the ITGA8 promoter modifies the progression of renal failure in ADPKD.
Collapse
Affiliation(s)
- Raoul Zeltner
- Department of Nephrology and Hypertension, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Polycystic kidney disease (PKD) is a diverse group of human monogenic lethal conditions inherited as autosomal dominant (AD) or recessive (AR) traits. Recent development of genetically engineered mouse models of ADPKD, ARPKD, and nephronophthisis/medullary cystic disease (NPHP) are providing additional insights into the molecular mechanisms governing of these disease processes as well as the developmental differentiation of the normal kidney. Genotypic and phenotypic mouse models are discussed and provide evidence for the fundamental involvement of cell-matrix, cell-cell, and primary cilia-lumen interactions, as well as epithelial proliferation, apoptosis, and polarization. Structure/function relationships between the PKD1, PKD2, PKHD1, and NPHP genes and proteins support the notion of a regulatory multiprotein cystic complex with a mechanosensory function that integrates signals from the extracellular environment. The plethora of intracellular signaling cascades that can impact renal cystic development suggest an exquisitely sensitive requirement for integrated downstream transduction and provide potential targets for therapeutic intervention. Appropriate genocopy models that faithfully recapitulate the phenotypic characteristics of the disease will be invaluable tools to analyze the effects of modifier genes and small molecule inhibitor therapies.
Collapse
|
39
|
Chang MY, Ong ACM. Autosomal dominant polycystic kidney disease: recent advances in pathogenesis and treatment. Nephron Clin Pract 2007; 108:p1-7. [PMID: 18075279 DOI: 10.1159/000112495] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Accepted: 09/23/2007] [Indexed: 12/11/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder affecting 1 in 1,000 people in the general population and accounts for up to 10% of all patients on renal replacement therapy. Numerous fluid-filled epithelial cysts arise from different nephron segments as spherical dilatations or small out-pouchings, enlarge progressively and eventually become disconnected from the rest of the renal tubule. The development of cysts is accompanied by destruction of the renal parenchyma, interstitial fibrosis, cellular infiltration and loss of functional nephrons. ADPKD is not only a kidney disease but also a systemic disorder associated with intracranial arterial aneurysms, cardiac valvular defects, colonic diverticulosis and cyst formation in other organs such as the liver, spleen and pancreas. The identification of PKD1 and PKD2 together with the drive to elucidate the functions of their encoded proteins, polycystin-1 (PC1) and polycystin-2 (PC2), has led to an explosion of clinical and scientific interest in this common disorder. The aim of this review is to highlight recent advances in our understanding of ADPKD pathogenesis which are leading to exciting new treatment strategies.
Collapse
Affiliation(s)
- Ming-Yang Chang
- Academic Nephrology Unit, Sheffield Kidney Institute, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, UK
| | | |
Collapse
|