1
|
Becker K. Retrospective analysis on the occurrence of kidney cysts in mice in a central animal facility in the years 2009-2019. Lab Anim 2024:236772241242538. [PMID: 39102530 DOI: 10.1177/00236772241242538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Kidney cysts in humans are mainly caused by inheritable polycystic kidney disease. Although they are a regular finding in laboratory mice, their occurrence upon dissection has not been systematically investigated, yet. Therefore, the aim of this report was to investigate on prevalence, phenotype and aetiology of spontaneously occurring kidney cysts in mice by retrospectively analysing the laboratory-receipt tables of the in-house laboratory of a central animal facility in North Rhine-Westphalia, Germany, years 2009-2019. A percentage of 0.4% of dissected mice displayed kidney cysts, with more male than female animals affected and average age equal to that of all dissected animals. Preliminary report in half of the cases was distended abdomen, and a few individuals displayed additional pathologic alterations of kidneys, most commonly dilated renal pelvis, or extrarenal comorbidities. Kidney cysts occurred independently of a renal phenotype of the transgenic strain or presence of infectious agents in health monitoring. To conclude, kidney cysts were characterized as harmless for affected mice but, as inheritability is suggested according with the literature, affected animals should be excluded from breeding.
Collapse
Affiliation(s)
- Katrin Becker
- Cardiovascular Research Laboratory, Division of Cardiology, Pulmonary Diseases and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
- Institute for Cardiovascular Sciences, University Hospital Bonn, University Bonn, Germany
| |
Collapse
|
2
|
Djaziri N, Burel C, Abbad L, Bakey Z, Piedagnel R, Lelongt B. Cleavage of periostin by MMP9 protects mice from kidney cystic disease. PLoS One 2023; 18:e0294922. [PMID: 38039285 PMCID: PMC10691688 DOI: 10.1371/journal.pone.0294922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/12/2023] [Indexed: 12/03/2023] Open
Abstract
The matrix metalloproteinase MMP9 influences cellular morphology and function, and plays important roles in organogenesis and disease. It exerts both protective and deleterious effects in renal pathology, depending upon its specific substrates. To explore new functions for MMP9 in kidney cysts formation and disease progression, we generated a mouse model by breeding juvenile cystic kidney (jck) mice with MMP9 deficient mice. Specifically, we provide evidence that MMP9 is overexpressed in cystic tissue where its enzymatic activity is increased 7-fold. MMP9 deficiency in cystic kidney worsen cystic kidney diseases by decreasing renal function, favoring cyst expansion and fibrosis. In addition, we find that periostin is a new critical substrate for MMP9 and in its absence periostin accumulates in cystic lining cells. As periostin promotes renal cyst growth and interstitial fibrosis in polycystic kidney diseases, we propose that the control of periostin by MMP9 and its associated intracellular signaling pathways including integrins, integrin-linked kinase and focal adhesion kinase confers to MMP9 a protective effect on the severity of the disease.
Collapse
Affiliation(s)
- Nabila Djaziri
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Cindy Burel
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Lilia Abbad
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Zeineb Bakey
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Rémi Piedagnel
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Brigitte Lelongt
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| |
Collapse
|
3
|
Yanda MK, Ciobanu C, Guggino WB, Cebotaru L. CFTR and PC2, partners in the primary cilia in autosomal dominant polycystic kidney disease. Am J Physiol Cell Physiol 2023; 325:C682-C693. [PMID: 37519231 PMCID: PMC10635646 DOI: 10.1152/ajpcell.00197.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Defects in the primary cilium are associated with autosomal dominant polycystic kidney disease (ADPKD). We used a combination of animal models, Western blotting, and confocal microscopy and discovered that CFTR and polycystin 2 (PC2) are both colocalized to the cilium in normal kidneys, with the levels of both being decreased in cystic epithelia. Cilia were longer in CFTR-null mice and in cystic cells in our ADPKD animal models. We examined septin 2, known to play a role in cilia length, to act as a diffusion barrier and to serve as an enhancer of proliferation. We found that septin 2 protein levels were upregulated and colocalized strongly with CFTR in cystic cells. Application of VX-809, the CFTR corrector, restored CFTR and PC2 toward normal in the cilia, decreased the protein levels of septin 2, and drastically reduced septin 2 colocalization with CFTR. Our data suggest that CFTR is present in the cilia and plays a role there, perhaps through its conductance of Cl-. We also postulate that septin 2 is important for localizing CFTR to the apical membrane in cystic epithelia.NEW & NOTEWORTHY CFTR is present in the primary cilia together with polycystin 2 (PC2). Ablation of CFTR makes cilia longer suggesting that CFTR plays a role there, perhaps through its conductance of Cl.
Collapse
Affiliation(s)
- Murali K Yanda
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Cristian Ciobanu
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - William B Guggino
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Liudmila Cebotaru
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Panda DK, Bai X, Zhang Y, Stylianesis NA, Koromilas AE, Lipman ML, Karaplis AC. SCF-SKP2 E3 ubiquitin ligase links mTORC1/ER stress/ISR with YAP activation in murine renal cystogenesis. J Clin Invest 2022; 132:153943. [PMID: 36326820 PMCID: PMC9754004 DOI: 10.1172/jci153943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
The Hippo pathway nuclear effector Yes-associated protein (YAP) potentiates the progression of polycystic kidney disease (PKD) arising from ciliopathies. The mechanisms underlying the increase in YAP expression and transcriptional activity in PKD remain obscure. We observed that in kidneys from mice with juvenile cystic kidney (jck) ciliopathy, the aberrant hyperactivity of mechanistic target of rapamycin complex 1 (mTORC1), driven by ERK1/2 and PI3K/AKT cascades, induced ER proteotoxic stress. To reduce this stress by reprogramming translation, the protein kinase R-like ER kinase-eukaryotic initiation factor 2α (PERK/eIF2α) arm of the integrated stress response (ISR) was activated. PERK-mediated phosphorylation of eIF2α drove the selective translation of activating transcription factor 4 (ATF4), potentiating YAP expression. In parallel, YAP underwent K63-linked polyubiquitination by SCF S-phase kinase-associated protein 2 (SKP2) E3 ubiquitin ligase, a Hippo-independent, nonproteolytic ubiquitination that enhances YAP nuclear trafficking and transcriptional activity in cancer cells. Defective ISR cellular adaptation to ER stress in eIF2α phosphorylation-deficient jck mice further augmented YAP-mediated transcriptional activity and renal cyst growth. Conversely, pharmacological tuning down of ER stress/ISR activity and SKP2 expression in jck mice by administration of tauroursodeoxycholic acid (TUDCA) or tolvaptan impeded these processes. Restoring ER homeostasis and/or interfering with the SKP2-YAP interaction represent potential therapeutic avenues for stemming the progression of renal cystogenesis.
Collapse
Affiliation(s)
- Dibyendu K. Panda
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital,,Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Xiuying Bai
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Yan Zhang
- Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | | | - Antonis E. Koromilas
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Mark L. Lipman
- Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Andrew C. Karaplis
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| |
Collapse
|
5
|
Swenson-Fields KI, Ward CJ, Lopez ME, Fross S, Heimes Dillon AL, Meisenheimer JD, Rabbani AJ, Wedlock E, Basu MK, Jansson KP, Rowe PS, Stubbs JR, Wallace DP, Vitek MP, Fields TA. Caspase-1 and the inflammasome promote polycystic kidney disease progression. Front Mol Biosci 2022; 9:971219. [PMID: 36523654 PMCID: PMC9745047 DOI: 10.3389/fmolb.2022.971219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/15/2022] [Indexed: 05/03/2024] Open
Abstract
We and others have previously shown that the presence of renal innate immune cells can promote polycystic kidney disease (PKD) progression. In this study, we examined the influence of the inflammasome, a key part of the innate immune system, on PKD. The inflammasome is a system of molecular sensors, receptors, and scaffolds that responds to stimuli like cellular damage or microbes by activating Caspase-1, and generating critical mediators of the inflammatory milieu, including IL-1β and IL-18. We provide evidence that the inflammasome is primed in PKD, as multiple inflammasome sensors were upregulated in cystic kidneys from human ADPKD patients, as well as in kidneys from both orthologous (PKD1 RC/RC or RC/RC) and non-orthologous (jck) mouse models of PKD. Further, we demonstrate that the inflammasome is activated in female RC/RC mice kidneys, and this activation occurs in renal leukocytes, primarily in CD11c+ cells. Knock-out of Casp1, the gene encoding Caspase-1, in the RC/RC mice significantly restrained cystic disease progression in female mice, implying sex-specific differences in the renal immune environment. RNAseq analysis implicated the promotion of MYC/YAP pathways as a mechanism underlying the pro-cystic effects of the Caspase-1/inflammasome in females. Finally, treatment of RC/RC mice with hydroxychloroquine, a widely used immunomodulatory drug that has been shown to inhibit the inflammasome, protected renal function specifically in females and restrained cyst enlargement in both male and female RC/RC mice. Collectively, these results provide evidence for the first time that the activated Caspase-1/inflammasome promotes cyst expansion and disease progression in PKD, particularly in females. Moreover, the data suggest that this innate immune pathway may be a relevant target for therapy in PKD.
Collapse
Affiliation(s)
- Katherine I. Swenson-Fields
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Christopher J. Ward
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, United States
| | - Micaila E. Lopez
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shaneann Fross
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Anna L. Heimes Dillon
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - James D. Meisenheimer
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Adib J. Rabbani
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Emily Wedlock
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Malay K. Basu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Kyle P. Jansson
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, United States
| | - Peter S. Rowe
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jason R. Stubbs
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, United States
| | - Darren P. Wallace
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, United States
| | - Michael P. Vitek
- Duke University Medical Center, Durham, NC, United States
- Resilio Therapeutics LLC, Durham, NC, United States
| | - Timothy A. Fields
- The Jared J. Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
6
|
In Mitosis You Are Not: The NIMA Family of Kinases in Aspergillus, Yeast, and Mammals. Int J Mol Sci 2022; 23:ijms23074041. [PMID: 35409400 PMCID: PMC8999480 DOI: 10.3390/ijms23074041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
The Never in mitosis gene A (NIMA) family of serine/threonine kinases is a diverse group of protein kinases implicated in a wide variety of cellular processes, including cilia regulation, microtubule dynamics, mitotic processes, cell growth, and DNA damage response. The founding member of this family was initially identified in Aspergillus and was found to play important roles in mitosis and cell division. The yeast family has one member each, Fin1p in fission yeast and Kin3p in budding yeast, also with functions in mitotic processes, but, overall, these are poorly studied kinases. The mammalian family, the main focus of this review, consists of 11 members named Nek1 to Nek11. With the exception of a few members, the functions of the mammalian Neks are poorly understood but appear to be quite diverse. Like the prototypical NIMA, many members appear to play important roles in mitosis and meiosis, but their functions in the cell go well beyond these well-established activities. In this review, we explore the roles of fungal and mammalian NIMA kinases and highlight the most recent findings in the field.
Collapse
|
7
|
Schirrer L, Marín-García PJ, Llobat L. Feline Polycystic Kidney Disease: An Update. Vet Sci 2021; 8:269. [PMID: 34822642 PMCID: PMC8625840 DOI: 10.3390/vetsci8110269] [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: 09/20/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Polycystic kidney disease (PKD) is a disease that affects felines and other mammals, such as humans. The common name is autosomal dominant polycystic kidney disease (ADPKD) and causes a progressive development of fluid-filled cysts in the kidney and sometimes in other organs as the liver and pancreas. The formation and growth of cysts progress slowly, causing deterioration of kidney tissue and a gradual decrease in kidney function, leading to irreversible kidney failure. Feline PKD or ADPKD in humans are hereditary pathologies of autosomal dominant transmission. ADPKD is one of the genetic diseases with the highest prevalence in humans. In cats, this disease also has a high prevalence, mainly in the Persian breed, being one of the most common feline genetic diseases. Imaging tests seem to be the most reliable method for diagnosis of the disease, although more genetic tests are being developed to detect the presence of the responsible mutation. In this review, we summarize the current knowledge about feline PKD to guide future research related to an adequate diagnosis and early detection of causal mutations. It can allow the establishment of selection programs to reduce or eliminate this pathology in feline breeds.
Collapse
Affiliation(s)
| | - Pablo Jesús Marín-García
- Department of Animal Production and Health, Veterinary Public Health and Food Sciences and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain;
| | - Lola Llobat
- Department of Animal Production and Health, Veterinary Public Health and Food Sciences and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain;
| |
Collapse
|
8
|
Schwarz H, Popp B, Airik R, Torabi N, Knaup KX, Stoeckert J, Wiech T, Amann K, Reis A, Schiffer M, Wiesener MS, Schueler M. Biallelic ANKS6 mutations cause late onset ciliopathy with chronic kidney disease through YAP dysregulation. Hum Mol Genet 2021; 31:1357-1369. [PMID: 34740236 DOI: 10.1093/hmg/ddab322] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Nephronophthisis-related ciliopathies (NPHP-RC) comprises a group of inherited kidney diseases, caused by mutations in genes encoding proteins localizing to primary cilia. NPHP-RC represent the one of the most frequent monogenic causes of renal failure within the first three decades of life, but its molecular disease mechanisms remains unclear. Here, we identified biallelic ANKS6 mutations in two affected siblings with late onset chronic kidney disease by whole exome sequencing. We employed patient derived fibroblasts generating an in vitro model to study the precise biological impact of distinct human ANKS6 mutations, completed by immunohistochemistry studies on renal biopsy samples. Functional studies using patient derived cells showed an impaired integrity of the ciliary Inversin compartment with reduced cilia length. Further analyses demonstrated that ANKS6 deficiency leads to a dysregulation of Hippo-signaling through nuclear YAP imbalance, and disrupted ciliary localization of YAP. Additionally an altered transcriptional activity of canonical Wnt target genes and altered expression of non-phosphorylated (active) β-catenin and phosphorylated GSK3β were observed. Upon ciliation ANKS6 deficiency revealed a deranged subcellular localization and expression of components of the endocytic recycling compartment. Our results demonstrate that ANKS6 plays a key role in regulating the Hippo pathway and ANKS6 deficiency is linked to dysregulation of signaling pathways. Our study provides molecular clues in understanding pathophysiological mechanisms of NPHP-RC and may offer new therapeutic targets.
Collapse
Affiliation(s)
- Hannah Schwarz
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Bernt Popp
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig, 04103, Germany.,Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Rannar Airik
- Department of Pediatrics, Division of Nephrology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA
| | - Nasrin Torabi
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Karl X Knaup
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Johanna Stoeckert
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Thorsten Wiech
- Institute of Pathology, Nephropathology Section, University Hospital Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Michael S Wiesener
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Markus Schueler
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
| |
Collapse
|
9
|
Gupta S, Ozimek-Kulik JE, Phillips JK. Nephronophthisis-Pathobiology and Molecular Pathogenesis of a Rare Kidney Genetic Disease. Genes (Basel) 2021; 12:genes12111762. [PMID: 34828368 PMCID: PMC8623546 DOI: 10.3390/genes12111762] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
The exponential rise in our understanding of the aetiology and pathophysiology of genetic cystic kidney diseases can be attributed to the identification of cystogenic genes over the last three decades. The foundation of this was laid by positional cloning strategies which gradually shifted towards next-generation sequencing (NGS) based screenings. This shift has enabled the discovery of novel cystogenic genes at an accelerated pace unlike ever before and, most notably, the past decade has seen the largest increase in identification of the genes which cause nephronophthisis (NPHP). NPHP is a monogenic autosomal recessive cystic kidney disease caused by mutations in a diverse clade of over 26 identified genes and is the most common genetic cause of renal failure in children. NPHP gene types present with some common pathophysiological features alongside a diverse range of extra-renal phenotypes associated with specific syndromic presentations. This review provides a timely update on our knowledge of this disease, including epidemiology, pathophysiology, anatomical and molecular features. We delve into the diversity of the NPHP causing genes and discuss known molecular mechanisms and biochemical pathways that may have possible points of intersection with polycystic kidney disease (the most studied renal cystic pathology). We delineate the pathologies arising from extra-renal complications and co-morbidities and their impact on quality of life. Finally, we discuss the current diagnostic and therapeutic modalities available for disease management, outlining possible avenues of research to improve the prognosis for NPHP patients.
Collapse
Affiliation(s)
- Shabarni Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- Correspondence:
| | - Justyna E. Ozimek-Kulik
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW 2031, Australia
- Department of Paediatric Nephrology, Sydney Children’s Hospital Network, Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Jacqueline Kathleen Phillips
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
| |
Collapse
|
10
|
Cordido A, Vizoso-Gonzalez M, Garcia-Gonzalez MA. Molecular Pathophysiology of Autosomal Recessive Polycystic Kidney Disease. Int J Mol Sci 2021; 22:6523. [PMID: 34204582 PMCID: PMC8235086 DOI: 10.3390/ijms22126523] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is a rare disorder and one of the most severe forms of polycystic kidney disease, leading to end-stage renal disease (ESRD) in childhood. PKHD1 is the gene that is responsible for the vast majority of ARPKD. However, some cases have been related to a new gene that was recently identified (DZIP1L gene), as well as several ciliary genes that can mimic a ARPKD-like phenotypic spectrum. In addition, a number of molecular pathways involved in the ARPKD pathogenesis and progression were elucidated using cellular and animal models. However, the function of the ARPKD proteins and the molecular mechanism of the disease currently remain incompletely understood. Here, we review the clinics, treatment, genetics, and molecular basis of ARPKD, highlighting the most recent findings in the field.
Collapse
Affiliation(s)
- Adrian Cordido
- Grupo de Xenética e Bioloxía do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxía (No. 11), Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.)
- Grupo de Medicina Xenómica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
| | - Marta Vizoso-Gonzalez
- Grupo de Xenética e Bioloxía do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxía (No. 11), Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.)
- Grupo de Medicina Xenómica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
| | - Miguel A. Garcia-Gonzalez
- Grupo de Xenética e Bioloxía do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxía (No. 11), Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.)
- Grupo de Medicina Xenómica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
- Fundación Publica Galega de Medicina Xenómica-SERGAS, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
11
|
Stokman MF, Saunier S, Benmerah A. Renal Ciliopathies: Sorting Out Therapeutic Approaches for Nephronophthisis. Front Cell Dev Biol 2021; 9:653138. [PMID: 34055783 PMCID: PMC8155538 DOI: 10.3389/fcell.2021.653138] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Nephronophthisis (NPH) is an autosomal recessive ciliopathy and a major cause of end-stage renal disease in children. The main forms, juvenile and adult NPH, are characterized by tubulointerstitial fibrosis whereas the infantile form is more severe and characterized by cysts. NPH is caused by mutations in over 20 different genes, most of which encode components of the primary cilium, an organelle in which important cellular signaling pathways converge. Ciliary signal transduction plays a critical role in kidney development and tissue homeostasis, and disruption of ciliary signaling has been associated with cyst formation, epithelial cell dedifferentiation and kidney function decline. Drugs have been identified that target specific signaling pathways (for example cAMP/PKA, Hedgehog, and mTOR pathways) and rescue NPH phenotypes in in vitro and/or in vivo models. Despite identification of numerous candidate drugs in rodent models, there has been a lack of clinical trials and there is currently no therapy that halts disease progression in NPH patients. This review covers the most important findings of therapeutic approaches in NPH model systems to date, including hypothesis-driven therapies and untargeted drug screens, approached from the pathophysiology of NPH. Importantly, most animal models used in these studies represent the cystic infantile form of NPH, which is less prevalent than the juvenile form. It appears therefore important to develop new models relevant for juvenile/adult NPH. Alternative non-orthologous animal models and developments in patient-based in vitro model systems are discussed, as well as future directions in personalized therapy for NPH.
Collapse
Affiliation(s)
- Marijn F Stokman
- Department of Genetics, University Medical Center Utrecht, Utrecht, Netherlands
- Université de Paris, Imagine Institute, Laboratory of Inherited Kidney Diseases, INSERM UMR 1163, Paris, France
| | - Sophie Saunier
- Université de Paris, Imagine Institute, Laboratory of Inherited Kidney Diseases, INSERM UMR 1163, Paris, France
| | - Alexandre Benmerah
- Université de Paris, Imagine Institute, Laboratory of Inherited Kidney Diseases, INSERM UMR 1163, Paris, France
| |
Collapse
|
12
|
Pavan ICB, Peres de Oliveira A, Dias PRF, Basei FL, Issayama LK, Ferezin CDC, Silva FR, Rodrigues de Oliveira AL, Alves dos Reis Moura L, Martins MB, Simabuco FM, Kobarg J. On Broken Ne(c)ks and Broken DNA: The Role of Human NEKs in the DNA Damage Response. Cells 2021; 10:507. [PMID: 33673578 PMCID: PMC7997185 DOI: 10.3390/cells10030507] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
NIMA-related kinases, or NEKs, are a family of Ser/Thr protein kinases involved in cell cycle and mitosis, centrosome disjunction, primary cilia functions, and DNA damage responses among other biological functional contexts in vertebrate cells. In human cells, there are 11 members, termed NEK1 to 11, and the research has mainly focused on exploring the more predominant roles of NEKs in mitosis regulation and cell cycle. A possible important role of NEKs in DNA damage response (DDR) first emerged for NEK1, but recent studies for most NEKs showed participation in DDR. A detailed analysis of the protein interactions, phosphorylation events, and studies of functional aspects of NEKs from the literature led us to propose a more general role of NEKs in DDR. In this review, we express that NEK1 is an activator of ataxia telangiectasia and Rad3-related (ATR), and its activation results in cell cycle arrest, guaranteeing DNA repair while activating specific repair pathways such as homology repair (HR) and DNA double-strand break (DSB) repair. For NEK2, 6, 8, 9, and 11, we found a role downstream of ATR and ataxia telangiectasia mutated (ATM) that results in cell cycle arrest, but details of possible activated repair pathways are still being investigated. NEK4 shows a connection to the regulation of the nonhomologous end-joining (NHEJ) repair of DNA DSBs, through recruitment of DNA-PK to DNA damage foci. NEK5 interacts with topoisomerase IIβ, and its knockdown results in the accumulation of damaged DNA. NEK7 has a regulatory role in the detection of oxidative damage to telomeric DNA. Finally, NEK10 has recently been shown to phosphorylate p53 at Y327, promoting cell cycle arrest after exposure to DNA damaging agents. In summary, this review highlights important discoveries of the ever-growing involvement of NEK kinases in the DDR pathways. A better understanding of these roles may open new diagnostic possibilities or pharmaceutical interventions regarding the chemo-sensitizing inhibition of NEKs in various forms of cancer and other diseases.
Collapse
Affiliation(s)
- Isadora Carolina Betim Pavan
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Andressa Peres de Oliveira
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Pedro Rafael Firmino Dias
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Fernanda Luisa Basei
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Luidy Kazuo Issayama
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Camila de Castro Ferezin
- Graduate Program in “Biologia Funcional e Molecular”, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-857, Brazil;
| | - Fernando Riback Silva
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Ana Luisa Rodrigues de Oliveira
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Lívia Alves dos Reis Moura
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
| | - Mariana Bonjiorno Martins
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
- Graduate Program in “Biologia Funcional e Molecular”, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-857, Brazil;
| | | | - Jörg Kobarg
- Graduate Program in “Ciências Farmacêuticas”, School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), R. Cândido Portinari 200, Prédio 2, Campinas CEP 13083-871, Brazil; (I.C.B.P.); (A.P.d.O.); (P.R.F.D.); (F.L.B.); (L.K.I.); (F.R.S.); (A.L.R.d.O.); (L.A.d.R.M.); (M.B.M.)
- Graduate Program in “Biologia Funcional e Molecular”, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-857, Brazil;
| |
Collapse
|
13
|
Blanc T, Goudin N, Zaidan M, Traore MG, Bienaime F, Turinsky L, Garbay S, Nguyen C, Burtin M, Friedlander G, Terzi F, Pontoglio M. Three-dimensional architecture of nephrons in the normal and cystic kidney. Kidney Int 2020; 99:632-645. [PMID: 33137337 DOI: 10.1016/j.kint.2020.09.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022]
Abstract
Kidney function is crucially dependent on the complex three-dimensional structure of nephrons. Any distortion of their shape may lead to kidney dysfunction. Traditional histological methods present major limitations for three-dimensional tissue reconstruction. Here, we combined tissue clearing, multi-photon microscopy and digital tracing for the reconstruction of single nephrons under physiological and pathological conditions. Sets of nephrons differing in location, shape and size according to their function were identified. Interestingly, nephrons tend to lie in planes. When this technique was applied to a model of cystic kidney disease, cysts were found to develop only in specific nephron segments. Along the same segment, cysts are contiguous within normal non-dilated tubules. Moreover, the shapes of cysts varied according to the nephron segment. Thus, our findings provide a valuable strategy for visualizing the complex structure of kidneys at the single nephron level and, more importantly, provide a basis for understanding pathological processes such as cystogenesis.
Collapse
Affiliation(s)
- Thomas Blanc
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France; Service de Chirurgie Viscérale et Urologie Pédiatrique, AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, US24-UMS3633, Paris, France
| | - Mohamad Zaidan
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France; Service de Néphrologie-Transplantation, AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | | | - Frank Bienaime
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France; Service d'Explorations Fonctionnelles, AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Lisa Turinsky
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France
| | - Serge Garbay
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France
| | - Clément Nguyen
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France
| | - Martine Burtin
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France
| | - Gérard Friedlander
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France; Service d'Explorations Fonctionnelles, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Fabiola Terzi
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France.
| | - Marco Pontoglio
- Institut National de la Santé et de la Recherche Médicale U1151, Centre National de la Recherche Scientifique UMR8253, Université de Paris, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Paris, France.
| |
Collapse
|
14
|
An Overview of In Vivo and In Vitro Models for Autosomal Dominant Polycystic Kidney Disease: A Journey from 3D-Cysts to Mini-Pigs. Int J Mol Sci 2020; 21:ijms21124537. [PMID: 32630605 PMCID: PMC7352572 DOI: 10.3390/ijms21124537] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inheritable cause of end stage renal disease and, as of today, only a single moderately effective treatment is available for patients. Even though ADPKD research has made huge progress over the last decades, the precise disease mechanisms remain elusive. However, a wide variety of cellular and animal models have been developed to decipher the pathophysiological mechanisms and related pathways underlying the disease. As none of these models perfectly recapitulates the complexity of the human disease, the aim of this review is to give an overview of the main tools currently available to ADPKD researchers, as well as their main advantages and limitations.
Collapse
|
15
|
Molinari E, Sayer JA. Disease Modeling To Understand the Pathomechanisms of Human Genetic Kidney Disorders. Clin J Am Soc Nephrol 2020; 15:855-872. [PMID: 32139361 PMCID: PMC7274277 DOI: 10.2215/cjn.08890719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The class of human genetic kidney diseases is extremely broad and heterogeneous. Accordingly, the range of associated disease phenotypes is highly variable. Many children and adults affected by inherited kidney disease will progress to ESKD at some point in life. Extensive research has been performed on various different disease models to investigate the underlying causes of genetic kidney disease and to identify disease mechanisms that are amenable to therapy. We review some of the research highlights that, by modeling inherited kidney disease, contributed to a better understanding of the underlying pathomechanisms, leading to the identification of novel genetic causes, new therapeutic targets, and to the development of new treatments. We also discuss how the implementation of more efficient genome-editing techniques and tissue-culture methods for kidney research is providing us with personalized models for a precision-medicine approach that takes into account the specificities of the patient and the underlying disease. We focus on the most common model systems used in kidney research and discuss how, according to their specific features, they can differentially contribute to biomedical research. Unfortunately, no definitive treatment exists for most inherited kidney disorders, warranting further exploitation of the existing disease models, as well as the implementation of novel, complex, human patient-specific models to deliver research breakthroughs.
Collapse
Affiliation(s)
- Elisa Molinari
- Faculty of Medical Sciences, Translational and Clinical Research Institute, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A. Sayer
- Faculty of Medical Sciences, Translational and Clinical Research Institute, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
- Renal Services, Newcastle Upon Tyne Hospitals National Health Service Trust, Newcastle upon Tyne, United Kingdom
- National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
16
|
Al Mutairi F, Alkhalaf R, Alkhorayyef A, Alroqi F, Yusra A, Umair M, Nouf F, Khan A, Meshael A, Hamad A, Monira A, Asiri A, Alhamoudi KM, Alfadhel M. Homozygous truncating NEK10 mutation, associated with primary ciliary dyskinesia: a case report. BMC Pulm Med 2020; 20:141. [PMID: 32414360 PMCID: PMC7229615 DOI: 10.1186/s12890-020-1175-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/29/2020] [Indexed: 11/10/2022] Open
Abstract
Background Primary Ciliary Dyskinesia (PCD) is also known as immotile-cilia syndrome, an autosomal recessive disorder of ciliary function, leading to mucus retention in the respiratory system in childhood. Our knowledge in the pathophysiological aspect of this devastating disorder is increasing with the advancement of genetic and molecular testing. Case presentation Here in, we report two siblings with a classical clinical and radiological presentation of PCD. Using whole exome sequencing we identified a homozygous truncating variant (c.3402 T > A); p.(Tyr1134*) in the NEK10 gene. Western bolt analysis revealed a decrease in the expression of NEK10 protein in the patient cells. Conclusions NEK10 plays a central role in the post-mitotic process of cilia assembly, regulating ciliary length and functions during physiological and pathological status. This study highlights the challenges of identifying disease-causing variants for a highly heterogeneous disorder and reports on the identification of a novel variant in NEK10 which recently associated with PCD.
Collapse
Affiliation(s)
- Fuad Al Mutairi
- Medical Genetics Division, Department of Pediatrics, King Abdullah specialized Children's Hospital, King Abdulaziz Medical City, P. O Box 22490, Riyadh, 11426, Saudi Arabia. .,Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.
| | - Randa Alkhalaf
- Medical Genetics Division, Department of Pediatrics, King Abdullah specialized Children's Hospital, King Abdulaziz Medical City, P. O Box 22490, Riyadh, 11426, Saudi Arabia
| | - Abdullah Alkhorayyef
- Pulmonary Division, Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Fayhan Alroqi
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,Immunology Division, Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Alyafee Yusra
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Fetaini Nouf
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Amjad Khan
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Alharbi Meshael
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Aleidi Hamad
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Alaujan Monira
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Abdulaziz Asiri
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Kheloud M Alhamoudi
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| | - Majid Alfadhel
- Medical Genetics Division, Department of Pediatrics, King Abdullah specialized Children's Hospital, King Abdulaziz Medical City, P. O Box 22490, Riyadh, 11426, Saudi Arabia.,Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Zhang JQJ, Burgess J, Stepanova D, Saravanabavan S, Wong ATY, Kaldis P, Rangan GK. Role of cyclin-dependent kinase 2 in the progression of mouse juvenile cystic kidney disease. J Transl Med 2020; 100:696-711. [PMID: 31915367 DOI: 10.1038/s41374-019-0360-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
A hallmark of polycystic kidney diseases (PKDs) is aberrant proliferation, which leads to the formation and growth of renal cysts. Proliferation is mediated by cyclin-dependent kinases (Cdks), and the administration of roscovitine (a pan-Cdk inhibitor) attenuates renal cystic disease in juvenile cystic kidney (jck) mice. Cdk2 is a key regulator of cell proliferation, but its specific role in PKD remains unknown. The aim of this study was to test the hypothesis that Cdk2 deficiency reduces renal cyst growth in PKD. Three studies were undertaken: (i) a time course (days 28, 56, and 84) of cyclin and Cdk activity was examined in jck mice and compared with wild-type mice; (ii) the progression was compared in jck mice with or without Cdk2 ablation from birth; and (iii) the effect of sirolimus (an antiproliferative agent) on Cdk2 activity in jck mice was investigated. Renal disease in jck mice was characterized by diffuse tubular cyst growth, interstitial inflammation and fibrosis, and renal impairment, peaking on day 84. Renal cell proliferation peaked during earlier stages of disease (days 28-56), whereas the expression of Cdk2-cyclin partners (A and E) and Cdk1 and 2 activity, was maximal in the later stages of disease (days 56-84). Cdk2 ablation did not attenuate renal disease progression and was associated with persistent Cdk1 activity. In contrast, the postnatal treatment of jck mice with sirolimus reduced both Cdk2 and Cdk1 activity and reduced renal cyst growth. In conclusion, (i) the kinetics of Cdk2 and Cdk2-cyclin partners did not correlate with proliferation in jck mice; and (ii) the absence of Cdk2 did not alter renal cyst growth, most likely due to compensation by Cdk1. Taken together, these data suggest that Cdk2 is dispensable for the proliferation of cystic epithelial cells and progression of PKD.
Collapse
Affiliation(s)
- Jennifer Qin Jing Zhang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia. .,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia.
| | - Jane Burgess
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Daria Stepanova
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Sayanthooran Saravanabavan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Annette T Y Wong
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science Technology and Research), Singapore, 138673, Republic of Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, 117597, Republic of Singapore
| | - Gopala K Rangan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Road, PO Box 412, Westmead, NSW, 2145, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, NSW, 2145, Australia
| |
Collapse
|
18
|
Melo-Hanchuk TD, Martins MB, Cunha LL, Soares FA, Ward LS, Vassallo J, Kobarg J. Expression of the NEK family in normal and cancer tissue: an immunohistochemical study. BMC Cancer 2020; 20:23. [PMID: 31906878 PMCID: PMC6945616 DOI: 10.1186/s12885-019-6408-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The NEK serine/threonine protein kinases are involved in cell cycle checkpoints, DNA damage repair, and apoptosis. Alterations in these pathways are frequently associated with cell malignant cellular transformations. Thyroid cancer is the most common malignant tumour in the endocrine system. Despite good treatment methods, the number of cases has increased significantly in recent years. Here, we studied the expression of NEK1, NEK2, NEK3, and NEK5 in different types of normal and malignant tissues, using tissue microarray analysis, and identified NEKs as potential markers in thyroid malignancy. METHODS The studied cases comprised multiple cancer tissue microarrays, including breast, colon, esophagus, kidney, lung, pancreas, prostate, stomach, thyroid and uterine cervix, as well as 281 patients who underwent thyroid resection for thyroid cancer or thyroid nodules. The expression of NEK1, NEK2, NEK3, and NEK5 was analyzed by immunohistochemistry. The expression pattern was evaluated in terms of intensity by two methods, semiquantitative and quantitative, and was compared between normal and cancer tissue. RESULTS We analysed the expression of each member of the NEK family in a tissue-dependent manner. Compared to normal tissue, most of the evaluated proteins showed lower expression in lung tumour. However, in the thyroid, the expression was higher in malignant tissue, especially for NEK 1, NEK3 and NEK5. Concerning characteristics of the thyroid tumour, such as aggressiveness, NEK1 expression was higher in tumours with multifocality and in patients with lymph node metastasis. NEK3 expression was stronger in patients with stage II, that involved metastasis. NEK5, on the other hand, showed high expression in patients with invasion and metastasis and in patients with tumour size > 4 cm. Furthermore, this work, demonstrated for the first time a high specificity and sensitivity of over-expression of NEK1 in classical and follicular variants of papillary thyroid cancer and NEK3 in tall-cell papillary thyroid cancer. CONCLUSION Taken together, the NEK protein kinases emerge as important proteins in thyroid cancer development and may help to identify malignancy and aggressiveness features during diagnosis. TRIAL REGISTRATION This study was retrospectively registered. www.accamargo.org.br/cientistas-pesquisadores/comite-de-etica-em-pequisa-cep.
Collapse
Affiliation(s)
- Talita Diniz Melo-Hanchuk
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Mariana Bonjiorno Martins
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Lucas Leite Cunha
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | - Laura Sterian Ward
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - José Vassallo
- Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Jörg Kobarg
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil. .,Faculdade de Ciências Farmacêuticas-UNICAMP, Universidade Estadual de Campinas, Campinas, Inst. de Biologia, Dep. Bioquímica e Biologia Tecidual, Rua Monteiro Lobato 255, CEP 13083-862, Campinas-SP, Brazil.
| |
Collapse
|
19
|
Rothé B, Gagnieux C, Leal-Esteban LC, Constam DB. Role of the RNA-binding protein Bicaudal-C1 and interacting factors in cystic kidney diseases. Cell Signal 2019; 68:109499. [PMID: 31838063 DOI: 10.1016/j.cellsig.2019.109499] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/03/2023]
Abstract
Polycystic kidneys frequently associate with mutations in individual components of cilia, basal bodies or centriolar satellites that perturb complex protein networks. In this review, we focus on the RNA-binding protein Bicaudal-C1 (BICC1) which was found mutated in renal cystic dysplasia, and on its interactions with the ankyrin repeat and sterile α motif (SAM)-containing proteins ANKS3 and ANKS6 and associated kinases and their partially overlapping ciliopathy phenotypes. After reviewing BICC1 homologs in model organisms and their functions in mRNA and cell metabolism during development and in renal tubules, we discuss recent insights from cell-based assays and from structure analysis of the SAM domains, and how SAM domain oligomerization might influence multivalent higher order complexes that are implicated in ciliary signal transduction.
Collapse
Affiliation(s)
- Benjamin Rothé
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland
| | - Céline Gagnieux
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland
| | - Lucia Carolina Leal-Esteban
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland; Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
20
|
Jiang YY, Maier W, Baumeister R, Minevich G, Joachimiak E, Wloga D, Ruan Z, Kannan N, Bocarro S, Bahraini A, Vasudevan KK, Lechtreck K, Orias E, Gaertig J. LF4/MOK and a CDK-related kinase regulate the number and length of cilia in Tetrahymena. PLoS Genet 2019; 15:e1008099. [PMID: 31339880 PMCID: PMC6682161 DOI: 10.1371/journal.pgen.1008099] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/05/2019] [Accepted: 06/13/2019] [Indexed: 11/18/2022] Open
Abstract
The length of cilia is controlled by a poorly understood mechanism that involves members of the conserved RCK kinase group, and among them, the LF4/MOK kinases. The multiciliated protist model, Tetrahymena, carries two types of cilia (oral and locomotory) and the length of the locomotory cilia is dependent on their position with the cell. In Tetrahymena, loss of an LF4/MOK ortholog, LF4A, lengthened the locomotory cilia, but also reduced their number. Without LF4A, cilia assembled faster and showed signs of increased intraflagellar transport (IFT). Consistently, overproduced LF4A shortened cilia and downregulated IFT. GFP-tagged LF4A, expressed in the native locus and imaged by total internal reflection microscopy, was enriched at the basal bodies and distributed along the shafts of cilia. Within cilia, most LF4A-GFP particles were immobile and a few either diffused or moved by IFT. We suggest that the distribution of LF4/MOK along the cilium delivers a uniform dose of inhibition to IFT trains that travel from the base to the tip. In a longer cilium, the IFT machinery may experience a higher cumulative dose of inhibition by LF4/MOK. Thus, LF4/MOK activity could be a readout of cilium length that helps to balance the rate of IFT-driven assembly with the rate of disassembly at steady state. We used a forward genetic screen to identify a CDK-related kinase, CDKR1, whose loss-of-function suppressed the shortening of cilia caused by overexpression of LF4A, by reducing its kinase activity. Loss of CDKR1 alone lengthened both the locomotory and oral cilia. CDKR1 resembles other known ciliary CDK-related kinases: LF2 of Chlamydomonas, mammalian CCRK and DYF-18 of C. elegans, in lacking the cyclin-binding motif and acting upstream of RCKs. The new genetic tools we developed here for Tetrahymena have potential for further dissection of the principles of cilia length regulation in multiciliated cells.
Collapse
Affiliation(s)
- Yu-Yang Jiang
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Wolfgang Maier
- Bio 3/Bioinformatics and Molecular Genetics, Faculty of Biology and ZBMZ, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Ralf Baumeister
- Bio 3/Bioinformatics and Molecular Genetics, Faculty of Biology and ZBMZ, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Gregory Minevich
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York, United States of America
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Zheng Ruan
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Stephen Bocarro
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Anoosh Bahraini
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Krishna Kumar Vasudevan
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Karl Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Eduardo Orias
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
21
|
Outtandy P, Russell C, Kleta R, Bockenhauer D. Zebrafish as a model for kidney function and disease. Pediatr Nephrol 2019; 34:751-762. [PMID: 29502161 PMCID: PMC6424945 DOI: 10.1007/s00467-018-3921-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/31/2022]
Abstract
Kidney disease is a global problem with around three million people diagnosed in the UK alone and the incidence is rising. Research is critical to develop better treatments. Animal models can help to better understand the pathophysiology behind the various kidney diseases and to screen for therapeutic compounds, but the use especially of mammalian models should be minimised in the interest of animal welfare. Zebrafish are increasingly used, as they are genetically tractable and have a basic renal anatomy comparable to mammalian kidneys with glomerular filtration and tubular filtration processing. Here, we discuss how zebrafish have advanced the study of nephrology and the mechanisms underlying kidney disease.
Collapse
Affiliation(s)
- Priya Outtandy
- Centre for Nephrology, Royal Free Hospital/Medical School, University College London, 1. Floor, Room 1.7007, Rowland Hill Street, London, NW3 2PF, UK
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Claire Russell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Robert Kleta
- Centre for Nephrology, Royal Free Hospital/Medical School, University College London, 1. Floor, Room 1.7007, Rowland Hill Street, London, NW3 2PF, UK.
| | - Detlef Bockenhauer
- Centre for Nephrology, Royal Free Hospital/Medical School, University College London, 1. Floor, Room 1.7007, Rowland Hill Street, London, NW3 2PF, UK
| |
Collapse
|
22
|
Lenhard SC, McAlexander A, Virtue A, Fieles W, Skedzielewski T, Rambo M, Trinh H, Cheng SH, Hong H, Isidro-Llobet A, Nadin A, Geske R, Klein JL, Lee D, Jucker BM, Hu E. In Vivo Imaging of Small Molecular Weight Peptides for Targeted Renal Drug Delivery: A Study in Normal and Polycystic Kidney Diseased Mice. J Pharmacol Exp Ther 2019; 370:786-795. [PMID: 30936291 DOI: 10.1124/jpet.119.257022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Stephen C Lenhard
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Allen McAlexander
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Anthony Virtue
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - William Fieles
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Tina Skedzielewski
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Mary Rambo
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Han Trinh
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Shih-Hsun Cheng
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Hyundae Hong
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Albert Isidro-Llobet
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Alan Nadin
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Robert Geske
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Jean-Louis Klein
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Dennis Lee
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Beat M Jucker
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| | - Erding Hu
- Bioimaging (S.C.L., T.S., M.R., S.-H.C., H.H., B.M.J.), Renal Discovery Group, Future Pipeline Discovery (A.V, E.H.), Experimental Cell and Tissue Biology, Target and Pathway Validation (W.F., H.T., R.G., J.-L.K.), Drug Delivery (A.M., D.L.), and Drug Design and Selection (A.I.-L., A.N.), GlaxoSmithKline plc, Collegeville, Pennsylvania
| |
Collapse
|
23
|
Miksiunas R, Mobasheri A, Bironaite D. Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac Development, Degeneration and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:155-178. [PMID: 30945165 DOI: 10.1007/5584_2019_349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the most common cause of human death in the developing world. Extensive evidence indicates that various toxic environmental factors and unhealthy lifestyle choices contribute to the risk, incidence and severity of cardiovascular diseases. Alterations in the genetic level of myocardium affects normal heart development and initiates pathological processes leading to various types of cardiac diseases. Homeobox genes are a large and highly specialized family of closely related genes that direct the formation of body structure, including cardiac development. Homeobox genes encode homeodomain proteins that function as transcription factors with characteristic structures that allow them to bind to DNA, regulate gene expression and subsequently control the proper physiological function of cells, tissues and organs. Mutations in homeobox genes are rare and usually lethal with evident alterations in cardiac function at or soon after the birth. Our understanding of homeobox gene family expression and function has expanded significantly during the recent years. However, the involvement of homeobox genes in the development of human and animal cardiac tissue requires further investigation. The phenotype of human congenital heart defects unveils only some aspects of human heart development. Therefore, mouse models are often used to gain a better understanding of human heart function, pathology and regeneration. In this review, we have focused on the role of homeobox genes in the development and pathology of human heart as potential tools for the future development of targeted regenerative strategies for various heart malfunctions.
Collapse
Affiliation(s)
- Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
24
|
Asymmetry in Mechanosensitive Gene Expression during Aortic Arch Morphogenesis. Sci Rep 2018; 8:16948. [PMID: 30446764 PMCID: PMC6240117 DOI: 10.1038/s41598-018-35127-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/30/2018] [Indexed: 11/09/2022] Open
Abstract
Embryonic aortic arches (AA) are initially bilaterally paired, transitional vessels and failures in remodeling based on hemodynamic and growth-related adaptations cause a spectrum of congenital heart disease (CHD) anatomies. Identifying regulatory mechanisms and cross-talk between the genetic elements of these vessels are critical to understand the ethiology of CHD and refine predictive computational models. This study aims to screen expression profiles of fundamental biological pathways in AA at early stages of chick embryo morphogenesis and correlate them with our current understanding of growth and mechanical loading. Reverse transcription-quantitative PCR (RT-qPCR) was followed by correlation and novel peak expression analyses to compare the behaviour and activation period of the genes. Available protein networks were also integrated to investigate the interactions between molecules and highlight major hierarchies. Only wall shear stress (WSS) and growth-correlated expression patterns were investigated. Effect of WSS was seen directly on angiogenesis as well on structural and apoptosis-related genes. Our time-resolved network suggested that WSS-correlated genes coordinate the activity of critical growth factors. Moreover, differential gene expression of left and right AA might be an indicator of subsequent asymmetric morphogenesis. These findings may further our understanding of the complex processes of cardiac morphogenesis and errors resulting in CHD.
Collapse
|
25
|
Impact of prenatal and postnatal maternal environment on nephron endowment, renal function and blood pressure in the Lewis polycystic kidney rat. J Dev Orig Health Dis 2018; 10:154-163. [PMID: 30274564 DOI: 10.1017/s2040174418000673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Maternal insufficiency during fetal development can have long-lasting effects on the offspring, most notably on nephron endowment. In polycystic kidney disease (PKD), variability in severity of disease is observed and maternal environment may be a modifying factor. In this study, we first established that in a rodent model of PKD, the Lewis polycystic kidney (LPK) rat's nephron numbers are 25% lower compared with wildtype animals. We then investigated the effects of prenatal and postnatal maternal environment on phenotype and nephron number. LPK pups born from and raised by homozygous LPK dams (control) were compared with LPK pups cross-fostered onto heterozygous LPK dams to improve postnatal environment; with LPK pups born from and raised by heterozygous LPK dams to improve both prenatal and postnatal environment and with LPK pups born from and raised by Wistar Kyoto-LPK heterozygous dams to improve both prenatal and postnatal environment on a different genetic background. Improvement in both prenatal and postnatal environment improved postnatal growth, renal function and reduced blood pressure, most notably in animals with different genetic background. Animals with improved postnatal environment only showed improved growth and blood pressure, but to a lesser extent. All intervention groups showed increased nephron number compared with control LPK. In summary, prenatal and postnatal environment had significant effect in delaying progression and reducing severity of PKD, including nephron endowment.
Collapse
|
26
|
Hoff S, Epting D, Falk N, Schroda S, Braun DA, Halbritter J, Hildebrandt F, Kramer-Zucker A, Bergmann C, Walz G, Lienkamp SS. The nucleoside-diphosphate kinase NME3 associates with nephronophthisis proteins and is required for ciliary function during renal development. J Biol Chem 2018; 293:15243-15255. [PMID: 30111592 PMCID: PMC6166740 DOI: 10.1074/jbc.ra117.000847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 08/07/2018] [Indexed: 01/12/2023] Open
Abstract
Nephronophthisis (NPH) is an autosomal recessive renal disease leading to kidney failure in children and young adults. The protein products of the corresponding genes (NPHPs) are localized in primary cilia or their appendages. Only about 70% of affected individuals have a mutation in one of 100 renal ciliopathy genes, and no unifying pathogenic mechanism has been identified. Recently, some NPHPs, including NIMA-related kinase 8 (NEK8) and centrosomal protein 164 (CEP164), have been found to act in the DNA-damage response pathway and to contribute to genome stability. Here, we show that NME/NM23 nucleoside-diphosphate kinase 3 (NME3) that has recently been found to facilitate DNA-repair mechanisms binds to several NPHPs, including NEK8, CEP164, and ankyrin repeat and sterile α motif domain-containing 6 (ANKS6). Depletion of nme3 in zebrafish and Xenopus resulted in typical ciliopathy-associated phenotypes, such as renal malformations and left-right asymmetry defects. We further found that endogenous NME3 localizes to the basal body and that it associates also with centrosomal proteins, such as NEK6, which regulates cell cycle arrest after DNA damage. The ciliopathy-typical manifestations of NME3 depletion in two vertebrate in vivo models, the biochemical association of NME3 with validated NPHPs, and its localization to the basal body reveal a role for NME3 in ciliary function. We conclude that mutations in the NME3 gene may aggravate the ciliopathy phenotypes observed in humans.
Collapse
Affiliation(s)
- Sylvia Hoff
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daniel Epting
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nathalie Falk
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Schroda
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jan Halbritter
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Albrecht Kramer-Zucker
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Carsten Bergmann
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany, and
| | - Gerd Walz
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Biological Signaling Studies (BIOSS), 79104 Freiburg, Germany
| | - Soeren S Lienkamp
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany,
- Center for Biological Signaling Studies (BIOSS), 79104 Freiburg, Germany
| |
Collapse
|
27
|
Versacci P, Pugnaloni F, Digilio MC, Putotto C, Unolt M, Calcagni G, Baban A, Marino B. Some Isolated Cardiac Malformations Can Be Related to Laterality Defects. J Cardiovasc Dev Dis 2018; 5:jcdd5020024. [PMID: 29724030 PMCID: PMC6023464 DOI: 10.3390/jcdd5020024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/22/2022] Open
Abstract
Human beings are characterized by a left–right asymmetric arrangement of their internal organs, and the heart is the first organ to break symmetry in the developing embryo. Aberrations in normal left–right axis determination during embryogenesis lead to a wide spectrum of abnormal internal laterality phenotypes, including situs inversus and heterotaxy. In more than 90% of instances, the latter condition is accompanied by complex and severe cardiovascular malformations. Atrioventricular canal defect and transposition of the great arteries—which are particularly frequent in the setting of heterotaxy—are commonly found in situs solitus with or without genetic syndromes. Here, we review current data on morphogenesis of the heart in human beings and animal models, familial recurrence, and upstream genetic pathways of left–right determination in order to highlight how some isolated congenital heart diseases, very common in heterotaxy, even in the setting of situs solitus, may actually be considered in the pathogenetic field of laterality defects.
Collapse
Affiliation(s)
- Paolo Versacci
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Flaminia Pugnaloni
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Carolina Putotto
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marta Unolt
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Giulio Calcagni
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Anwar Baban
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Bruno Marino
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
28
|
Panda DK, Bai X, Sabbagh Y, Zhang Y, Zaun HC, Karellis A, Koromilas AE, Lipman ML, Karaplis AC. Defective interplay between mTORC1 activity and endoplasmic reticulum stress-unfolded protein response in uremic vascular calcification. Am J Physiol Renal Physiol 2018; 314:F1046-F1061. [PMID: 29357413 DOI: 10.1152/ajprenal.00350.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular calcification increases the risk of cardiovascular disease and death in patients with chronic kidney disease (CKD). Increased activity of mammalian target of rapamycin complex 1 (mTORC1) and endoplasmic reticulum (ER) stress-unfolded protein response (UPR) are independently reported to partake in the pathogenesis of vascular calcification in CKD. However, the association between mTORC1 activity and ER stress-UPR remains unknown. We report here that components of the uremic state [activation of the receptor for advanced glycation end products (RAGE) and hyperphosphatemia] potentiate vascular smooth muscle cell (VSMC) calcification by inducing persistent and exaggerated activity of mTORC1. This gives rise to prolonged and excessive ER stress-UPR as well as attenuated levels of sestrin 1 ( Sesn1) and Sesn3 feeding back to inhibit mTORC1 activity. Activating transcription factor 4 arising from the UPR mediates cell death via expression of CCAAT/enhancer-binding protein (c/EBP) homologous protein (CHOP), impairs the generation of pyrophosphate, a potent inhibitor of mineralization, and potentiates VSMC transdifferentiation to the osteochondrocytic phenotype. Short-term treatment of CKD mice with rapamycin, an inhibitor of mTORC1, or tauroursodeoxycholic acid, a bile acid that restores ER homeostasis, normalized mTORC1 activity, molecular markers of UPR, and calcium content of aortas. Collectively, these data highlight that increased and/or protracted mTORC1 activity arising from the uremic state leads to dysregulated ER stress-UPR and VSMC calcification. Manipulation of the mTORC1-ER stress-UPR pathway opens up new therapeutic strategies for the prevention and treatment of vascular calcification in CKD.
Collapse
Affiliation(s)
- Dibyendu K Panda
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Xiuying Bai
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Yves Sabbagh
- Rare Disease, Sanofi Genzyme, Framingham, Massachusetts
| | - Yan Zhang
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Hans-Christian Zaun
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Angeliki Karellis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Antonis E Koromilas
- Department of Oncology and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Mark L Lipman
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Andrew C Karaplis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| |
Collapse
|
29
|
Gehrig J, Pandey G, Westhoff JH. Zebrafish as a Model for Drug Screening in Genetic Kidney Diseases. Front Pediatr 2018; 6:183. [PMID: 30003073 PMCID: PMC6031734 DOI: 10.3389/fped.2018.00183] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
Genetic disorders account for a wide range of renal diseases emerging during childhood and adolescence. Due to the utilization of modern biochemical and biomedical techniques, the number of identified disease-associated genes is increasing rapidly. Modeling of congenital human disease in animals is key to our understanding of the biological mechanism underlying pathological processes and thus developing novel potential treatment options. The zebrafish (Danio rerio) has been established as a versatile small vertebrate organism that is widely used for studying human inherited diseases. Genetic accessibility in combination with elegant experimental methods in zebrafish permit modeling of human genetic diseases and dissecting the perturbation of underlying cellular networks and physiological processes. Beyond its utility for genetic analysis and pathophysiological and mechanistic studies, zebrafish embryos, and larvae are amenable for phenotypic screening approaches employing high-content and high-throughput experiments using automated microscopy. This includes large-scale chemical screening experiments using genetic models for searching for disease-modulating compounds. Phenotype-based approaches of drug discovery have been successfully performed in diverse zebrafish-based screening applications with various phenotypic readouts. As a result, these can lead to the identification of candidate substances that are further examined in preclinical and clinical trials. In this review, we discuss zebrafish models for inherited kidney disease as well as requirements and considerations for the technical realization of drug screening experiments in zebrafish.
Collapse
Affiliation(s)
- Jochen Gehrig
- Acquifer is a Division of Ditabis, Digital Biomedical Imaging Systems AG, Pforzheim, Germany
| | - Gunjan Pandey
- Acquifer is a Division of Ditabis, Digital Biomedical Imaging Systems AG, Pforzheim, Germany.,Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Jens H Westhoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
Fry AM, Bayliss R, Roig J. Mitotic Regulation by NEK Kinase Networks. Front Cell Dev Biol 2017; 5:102. [PMID: 29250521 PMCID: PMC5716973 DOI: 10.3389/fcell.2017.00102] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 12/24/2022] Open
Abstract
Genetic studies in yeast and Drosophila led to identification of cyclin-dependent kinases (CDKs), Polo-like kinases (PLKs) and Aurora kinases as essential regulators of mitosis. These enzymes have since been found in the majority of eukaryotes and their cell cycle-related functions characterized in great detail. However, genetic studies in another fungal species, Aspergillus nidulans, identified a distinct family of protein kinases, the NEKs, that are also widely conserved and have key roles in the cell cycle, but which remain less well studied. Nevertheless, it is now clear that multiple NEK family members act in networks to regulate specific events of mitosis, including centrosome separation, spindle assembly and cytokinesis. Here, we describe our current understanding of how the NEK kinases contribute to these processes, particularly through targeted phosphorylation of proteins associated with the microtubule cytoskeleton. We also present the latest findings on molecular events that control the activation state of the NEKs and how these are revealing novel modes of enzymatic regulation relevant not only to other kinases but also to pathological mechanisms of disease.
Collapse
Affiliation(s)
- Andrew M. Fry
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Richard Bayliss
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Joan Roig
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| |
Collapse
|
31
|
Abstract
The primary cilium is a microtubule-based organelle that is considered to be a cellular antennae, because proteins related to multiple signaling pathways such as Wnt, PDGFRα, Hh, and mechanosignaling are localized to the membrane of the primary cilium. In the kidney, primary cilia extend from the cell membrane to the lumen of renal tubules to respond to fluidic stress. Recent studies have indicated that the disruption of ciliary proteins including polycystin-1 (PC1), polycystin-2 (PC2), and members of the intraflagellar transport (IFT) family induce the development of polycystic kidney disease (PKD), suggesting that the malformation or absence of primary cilia is a driving force of the onset of PKD. Therefore, in this chapter, the renal cystogenesis mechanism induced by cilia defects and pathogenic ciliary proteins associated with PKD development will be described.
Collapse
Affiliation(s)
- Je Yeong Ko
- Molecular Medicine Laboratory, Department of Life systems, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul, 04310, South Korea.
| |
Collapse
|
32
|
Lažetić V, Fay DS. Conserved Ankyrin Repeat Proteins and Their NIMA Kinase Partners Regulate Extracellular Matrix Remodeling and Intracellular Trafficking in Caenorhabditis elegans. Genetics 2017; 205:273-293. [PMID: 27799278 PMCID: PMC5223508 DOI: 10.1534/genetics.116.194464] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/28/2016] [Indexed: 12/27/2022] Open
Abstract
Molting is an essential developmental process in nematodes during which the epidermal apical extracellular matrix, the cuticle, is remodeled to accommodate further growth. Using genetic approaches, we identified a requirement for three conserved ankyrin repeat-rich proteins, MLT-2/ANKS6, MLT-3/ANKS3, and MLT-4/INVS, in Caenorhabditis elegans molting. Loss of mlt function resulted in severe defects in the ability of larvae to shed old cuticle and led to developmental arrest. Genetic analyses demonstrated that MLT proteins functionally cooperate with the conserved NIMA kinase family members NEKL-2/NEK8 and NEKL-3/NEK6/NEK7 to promote cuticle shedding. MLT and NEKL proteins were specifically required within the hyp7 epidermal syncytium, and fluorescently tagged mlt and nekl alleles were expressed in puncta within this tissue. Expression studies further showed that NEKL-2-MLT-2-MLT-4 and NEKL-3-MLT-3 colocalize within largely distinct assemblies of apical foci. MLT-2 and MLT-4 were required for the normal accumulation of NEKL-2 at the hyp7-seam cell boundary, and loss of mlt-2 caused abnormal nuclear accumulation of NEKL-2 Correspondingly, MLT-3, which bound directly to NEKL-3, prevented NEKL-3 nuclear localization, supporting the model that MLT proteins may serve as molecular scaffolds for NEKL kinases. Our studies additionally showed that the NEKL-MLT network regulates early steps in clathrin-mediated endocytosis at the apical surface of hyp7, which may in part account for molting defects observed in nekl and mlt mutants. This study has thus identified a conserved NEKL-MLT protein network that regulates remodeling of the apical extracellular matrix and intracellular trafficking, functions that may be conserved across species.
Collapse
Affiliation(s)
- Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming 82071
| | - David S Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming 82071
| |
Collapse
|
33
|
Abeyta A, Castella M, Jacquemont C, Taniguchi T. NEK8 regulates DNA damage-induced RAD51 foci formation and replication fork protection. Cell Cycle 2016; 16:335-347. [PMID: 27892797 PMCID: PMC5324754 DOI: 10.1080/15384101.2016.1259038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Proteins essential for homologous recombination play a pivotal role in the repair of DNA double strand breaks, DNA inter-strand crosslinks and replication fork stability. Defects in homologous recombination also play a critical role in the development of cancer and the sensitivity of these cancers to chemotherapy. RAD51, an essential factor for homologous recombination and replication fork protection, accumulates and forms immunocytochemically detectable nuclear foci at sites of DNA damage. To identify kinases that may regulate RAD51 localization to sites of DNA damage, we performed a human kinome siRNA library screen, using DNA damage-induced RAD51 foci formation as readout. We found that NEK8, a NIMA family kinase member, is required for efficient DNA damage-induced RAD51 foci formation. Interestingly, knockout of Nek8 in murine embryonic fibroblasts led to cellular sensitivity to the replication inhibitor, hydroxyurea, and inhibition of the ATR kinase. Furthermore, NEK8 was required for proper replication fork protection following replication stall with hydroxyurea. Loading of RAD51 to chromatin was decreased in NEK8-depleted cells and Nek8-knockout cells. Single-molecule DNA fiber analyses revealed that nascent DNA tracts were degraded in the absence of NEK8 following treatment with hydroxyurea. Consistent with this, Nek8-knockout cells showed increased chromosome breaks following treatment with hydroxyurea. Thus, NEK8 plays a critical role in replication fork stability through its regulation of the DNA repair and replication fork protection protein RAD51.
Collapse
Affiliation(s)
- Antonio Abeyta
- a Divisions of Human Biology and Public Health Sciences , Fred Hutchinson Cancer Research Center , Seattle , WA , USA.,b Molecular & Cellular Biology Graduate Program , University of Washington , Seattle , WA , USA
| | - Maria Castella
- a Divisions of Human Biology and Public Health Sciences , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| | - Celine Jacquemont
- a Divisions of Human Biology and Public Health Sciences , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| | - Toshiyasu Taniguchi
- a Divisions of Human Biology and Public Health Sciences , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| |
Collapse
|
34
|
High-resolution genetic localization of a modifying locus affecting disease severity in the juvenile cystic kidneys (jck) mouse model of polycystic kidney disease. Mamm Genome 2016; 27:191-9. [PMID: 27114383 DOI: 10.1007/s00335-016-9633-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/07/2016] [Indexed: 12/31/2022]
Abstract
We have previously demonstrated that a locus on proximal Chr 4 modifies disease severity in the juvenile cystic kidney (jck) mouse, a model of polycystic kidney disease (PKD) that carries a mutation of the Nek8 serine-threonine kinase. In this study, we used QTL analysis of independently constructed B6.D2 congenic lines to confirm this and showed that this locus has a highly significant effect. We constructed sub-congenic lines to more specifically localize the modifier and have determined it resides in a 3.2 Mb interval containing 28 genes. These include Invs and Anks6, which are both excellent candidates for the modifier as mutations in these genes result in PKD and both genes are known to genetically and physically interact with Nek8. However, examination of strain-specific DNA sequence and kidney expression did not reveal clear differences that might implicate either gene as a modifier of PKD severity. The fact that our high-resolution analysis did not yield an unambiguous result highlights the challenge of establishing the causality of strain-specific variants as genetic modifiers, and suggests that alternative strategies be considered.
Collapse
|
35
|
Husson H, Moreno S, Smith LA, Smith MM, Russo RJ, Pitstick R, Sergeev M, Ledbetter SR, Bukanov NO, Lane M, Zhang K, Billot K, Carlson G, Shah J, Meijer L, Beier DR, Ibraghimov-Beskrovnaya O. Reduction of ciliary length through pharmacologic or genetic inhibition of CDK5 attenuates polycystic kidney disease in a model of nephronophthisis. Hum Mol Genet 2016; 25:2245-2255. [PMID: 27053712 PMCID: PMC5081056 DOI: 10.1093/hmg/ddw093] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
Polycystic kidney diseases (PKDs) comprise a subgroup of ciliopathies characterized by the formation of fluid-filled kidney cysts and progression to end-stage renal disease. A mechanistic understanding of cystogenesis is crucial for the development of viable therapeutic options. Here, we identify CDK5, a kinase active in post mitotic cells, as a new and important mediator of PKD progression. We show that long-lasting attenuation of PKD in the juvenile cystic kidneys (jck) mouse model of nephronophthisis by pharmacological inhibition of CDK5 using either R-roscovitine or S-CR8 is accompanied by sustained shortening of cilia and a more normal epithelial phenotype, suggesting this treatment results in a reprogramming of cellular differentiation. Also, a knock down of Cdk5 in jck cells using small interfering RNA results in significant shortening of ciliary length, similar to what we observed with R-roscovitine. Finally, conditional inactivation of Cdk5 in the jck mice significantly attenuates cystic disease progression and is associated with shortening of ciliary length as well as restoration of cellular differentiation. Our results suggest that CDK5 may regulate ciliary length by affecting tubulin dynamics via its substrate collapsin response mediator protein 2. Taken together, our data support therapeutic approaches aimed at restoration of ciliogenesis and cellular differentiation as a promising strategy for the treatment of renal cystic diseases.
Collapse
Affiliation(s)
- Hervé Husson
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Sarah Moreno
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Laurie A Smith
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Mandy M Smith
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Ryan J Russo
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Rose Pitstick
- McLaughlin Research Institute, 1520 23rd Street South, Great Falls, Montana 59405, USA
| | - Mikhail Sergeev
- Harvard Institutes of Medicine, 4 Blackfan Circle HIM568, Boston, MA 02115, USA
| | - Steven R Ledbetter
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Nikolay O Bukanov
- Department of Rare Diseases, Sanofi-Genzyme R&D Center, 49 New York Avenue, Framingham, MA 01701, USA
| | - Monica Lane
- Department of Biological Mass Spectrometry & Biomarker Research, Sanofi-Genzyme R&D Center, 1 Mountain Road, Framingham, MA 01701, USA
| | - Kate Zhang
- Department of Biological Mass Spectrometry & Biomarker Research, Sanofi-Genzyme R&D Center, 1 Mountain Road, Framingham, MA 01701, USA
| | - Katy Billot
- ManRos Therapeutics, Hotel de Recherche-Centre de Perharidy, 29680 Roscoff, France
| | - George Carlson
- McLaughlin Research Institute, 1520 23rd Street South, Great Falls, Montana 59405, USA
| | - Jagesh Shah
- Harvard Institutes of Medicine, 4 Blackfan Circle HIM568, Boston, MA 02115, USA
| | - Laurent Meijer
- ManRos Therapeutics, Hotel de Recherche-Centre de Perharidy, 29680 Roscoff, France
| | - David R Beier
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
| | | |
Collapse
|
36
|
Grampa V, Delous M, Zaidan M, Odye G, Thomas S, Elkhartoufi N, Filhol E, Niel O, Silbermann F, Lebreton C, Collardeau-Frachon S, Rouvet I, Alessandri JL, Devisme L, Dieux-Coeslier A, Cordier MP, Capri Y, Khung-Savatovsky S, Sigaudy S, Salomon R, Antignac C, Gubler MC, Benmerah A, Terzi F, Attié-Bitach T, Jeanpierre C, Saunier S. Novel NEK8 Mutations Cause Severe Syndromic Renal Cystic Dysplasia through YAP Dysregulation. PLoS Genet 2016; 12:e1005894. [PMID: 26967905 PMCID: PMC4788435 DOI: 10.1371/journal.pgen.1005894] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/02/2016] [Indexed: 01/01/2023] Open
Abstract
Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, >500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of >1200 ciliary genes. NEK8/NPHP9 mutations were identified in five cases with severe overlapping phenotypes including renal cystic dysplasia/hypodysplasia, situs inversus, cardiopathy with hypertrophic septum and bile duct paucity. These cases highlight a genotype-phenotype correlation, with missense and nonsense mutations associated with hypodysplasia and enlarged cystic organs, respectively. Functional analyses of NEK8 mutations in patient fibroblasts and mIMCD3 cells showed that these mutations differentially affect ciliogenesis, proliferation/apoptosis/DNA damage response, as well as epithelial morphogenesis. Notably, missense mutations exacerbated some of the defects due to NEK8 loss of function, highlighting their likely gain-of-function effect. We also showed that NEK8 missense and loss-of-function mutations differentially affect the regulation of the main Hippo signaling effector, YAP, as well as the expression of its target genes in patient fibroblasts and renal cells. YAP imbalance was also observed in enlarged spheroids of Nek8-invalidated renal epithelial cells grown in 3D culture, as well as in cystic kidneys of Jck mice. Moreover, co-injection of nek8 MO with WT or mutated NEK8-GFP RNA in zebrafish embryos led to shortened dorsally curved body axis, similar to embryos injected with human YAP RNA. Finally, treatment with Verteporfin, an inhibitor of YAP transcriptional activity, partially rescued the 3D spheroid defects of Nek8-invalidated cells and the abnormalities of NEK8-overexpressing zebrafish embryos. Altogether, our study demonstrates that NEK8 human mutations cause major organ developmental defects due to altered ciliogenesis and cell differentiation/proliferation through deregulation of the Hippo pathway. Genes mutated in ciliopathies encode proteins with various localizations and functions at the primary cilium. Here we report novel NEK8 mutations in patients with renal cystic hypodysplasia and associated ciliopathy defects. NEK8 belongs to a protein complex defining the Inversin compartment of the cilium. It is also a negative regulator of the Hippo signaling pathway that controls organ growth. We report genotype-phenotype correlation in the patients. We functionally demonstrate that the two types of mutations (missense versus nonsense) differentially affect ciliogenesis, cell apoptosis and epithelialisation. We also show that all the mutations lead to dysregulation of the Hippo pathway through nuclear YAP imbalance but that the nature of this imbalance is different according to the type of mutation. We confirm alteration of the Hippo pathway associated with Nek8 mutation in vivo in Jck mice. Remarkably, we show that morphogenesis defects observed in Nek8 knockdown epithelial cells or zebrafish embryos are rescued by Verteporfin, a specific inhibitor of YAP transcriptional activity, demonstrating the causative role of YAP dysregulation in the occurrence of these defects. Altogether, this study links NEK8 mutations to dysregulation of the Hippo pathway and provide molecular clues to understand the variability of the multiorgan defects in the patients.
Collapse
Affiliation(s)
- Valentina Grampa
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Marion Delous
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Mohamad Zaidan
- INSERM U1151, CNRS UMR8253, Paris Descartes—Sorbonne Paris Cité University, Necker-Enfants Malades Institute, Mechanisms and Therapeutic Strategies of Chronic Kidney Diseases, Necker Hospital, Paris, France
| | - Gweltas Odye
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Sophie Thomas
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris, France
| | - Nadia Elkhartoufi
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris, France
- Department of Genetics, AP-HP, Necker Hospital, Paris, France
| | - Emilie Filhol
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Olivier Niel
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Department of Pediatric Nephrology, AP-HP, Robert Debré Hospital, Paris, France
| | - Flora Silbermann
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Corinne Lebreton
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Intestinal Immunity, Paris, France
| | | | - Isabelle Rouvet
- Cellular Biotechnology Department and Biobank, Hospices Civils de Lyon, CHU de Lyon, Lyon, France
| | | | - Louise Devisme
- Anatomopathological Department, CHRU Lille, University Hospital, Lille, France
| | | | - Marie-Pierre Cordier
- Department of Genetics, Femme Mère-Enfant Hospital, University of Lyon 1, Bron, France
| | - Yline Capri
- Department of Genetics, CHU Robert-Debré, Paris, France
| | | | - Sabine Sigaudy
- Multidisciplinary Department of Prenatal Diagnosis, La Timone Children’s Hospital, Marseille, France
| | - Rémi Salomon
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Department of Pediatric Nephrology, AP-HP, Necker Hospital, Paris, France
| | - Corinne Antignac
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Department of Genetics, AP-HP, Necker Hospital, Paris, France
| | - Marie-Claire Gubler
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Alexandre Benmerah
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Fabiola Terzi
- INSERM U1151, CNRS UMR8253, Paris Descartes—Sorbonne Paris Cité University, Necker-Enfants Malades Institute, Mechanisms and Therapeutic Strategies of Chronic Kidney Diseases, Necker Hospital, Paris, France
| | - Tania Attié-Bitach
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris, France
- Department of Histology-Embryology and Cytogenetics, AP-HP, Necker Hospital, Paris, France
| | - Cécile Jeanpierre
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Sophie Saunier
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France
- Paris Descartes—Sorbonne Paris Cité University, Imagine Institute, Paris, France
- * E-mail:
| |
Collapse
|
37
|
Al-Hamed MH, Kurdi W, Alsahan N, Alabdullah Z, Abudraz R, Tulbah M, Alnemer M, Khan R, Al-Jurayb H, Alahmed A, Tahir AI, Khalil D, Edwards N, Al Abdulaziz B, Binhumaid FS, Majid S, Faquih T, El-Kalioby M, Abouelhoda M, Altassan N, Monies D, Meyer B, Sayer JA, Albaqumi M. Genetic spectrum of Saudi Arabian patients with antenatal cystic kidney disease and ciliopathy phenotypes using a targeted renal gene panel. J Med Genet 2016; 53:338-47. [PMID: 26862157 PMCID: PMC4853542 DOI: 10.1136/jmedgenet-2015-103469] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/04/2016] [Indexed: 12/24/2022]
Abstract
Background Inherited cystic kidney disorders are a common cause of end-stage renal disease. Over 50 ciliopathy genes, which encode proteins that influence the structure and function of the primary cilia, are implicated in cystic kidney disease. Methods To define the phenotype and genotype of cystic kidney disease in fetuses and neonates, we correlated antenatal ultrasound examination and postnatal renal ultrasound examination with targeted exon sequencing, using a renal gene panel. A cohort of 44 families in whom antenatal renal ultrasound scanning findings in affected cases included bilateral cystic kidney disease, echogenic kidneys or enlarged kidneys was investigated. Results In this cohort, disease phenotypes were severe with 36 cases of stillbirth or perinatal death. Extra renal malformations, including encephalocele, polydactyly and heart malformations, consistent with ciliopathy phenotypes, were frequently detected. Renal gene panel testing identified causative mutations in 21 out of 34 families (62%), where patient and parental DNA was available. In the remaining 10 families, where only parental DNA was available, 7 inferred causative mutations were found. Together, mutations were found in 12 different genes with a total of 13 novel pathogenic variants, including an inferred novel variant in NEK8. Mutations in CC2D2A were the most common cause of an antenatal cystic kidney disease and a suspected ciliopathy in our cohort. Conclusions In families with ciliopathy phenotypes, mutational analysis using a targeted renal gene panel allows a rapid molecular diagnosis and provides important information for patients, parents and their physicians.
Collapse
Affiliation(s)
- Mohamed H Al-Hamed
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Wesam Kurdi
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Nada Alsahan
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Zainab Alabdullah
- Obstetrics & Gynecology Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Rania Abudraz
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Maha Tulbah
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Maha Alnemer
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Rubina Khan
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Haya Al-Jurayb
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ahmed Alahmed
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Asma I Tahir
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dania Khalil
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Noel Edwards
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Basma Al Abdulaziz
- Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Faisal S Binhumaid
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Salma Majid
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Tariq Faquih
- Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Mohamed El-Kalioby
- Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Mohamed Abouelhoda
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Nada Altassan
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Dorota Monies
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Brian Meyer
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia Saudi Human Genome Project, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - John A Sayer
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Mamdouh Albaqumi
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Quek KJ, Boyd R, Ameer OZ, Zangerl B, Butlin M, Murphy TV, Avolio AP, Phillips JK. Progressive vascular remodelling, endothelial dysfunction and stiffness in mesenteric resistance arteries in a rodent model of chronic kidney disease. Vascul Pharmacol 2016; 81:42-52. [PMID: 26771067 DOI: 10.1016/j.vph.2015.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/02/2015] [Accepted: 12/31/2015] [Indexed: 12/25/2022]
Abstract
Chronic kidney disease (CKD) and hypertension are co-morbid conditions both associated with altered resistance artery structure, biomechanics and function. We examined these characteristics in mesenteric artery together with renal function and systolic blood pressure (SBP) changes in the Lewis polycystic kidney (LPK) rat model of CKD. Animals were studied at early (6-weeks), intermediate (12-weeks), and late (18-weeks) time-points (n=21), relative to age-matched Lewis controls (n=29). At 12 and 18-weeks, LPK arteries exhibited eutrophic and hypertrophic inward remodelling characterised by thickened medial smooth muscle, decreased lumen diameter, and unchanged or increased media cross-sectional area, respectively. At these later time points, endothelium-dependent vasorelaxation was also compromised, associated with impaired endothelium-dependent hyperpolarisation and reduced nitric oxide synthase activity. Stiffness, elastic-modulus/stress slopes and collagen/elastin ratios were increased in 6 and 18-week-old-LPK, in contrast to greater arterial compliance at 12weeks. Multiple linear regression analysis highlighted SBP as the main predictor of wall-lumen ratio (r=0.536, P<0.001 n=46 pairs). Concentration-response curves revealed increased sensitivity to phenylephrine but not potassium chloride in 18-week-LPK. Our results indicate that impairment in LPK resistance vasculature is evident at 6weeks, and worsens with hypertension and progression of renal disease.
Collapse
Affiliation(s)
- K J Quek
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - R Boyd
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - O Z Ameer
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - B Zangerl
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia; Centre for Eye Health, University of New South Wales, Sydney, Australia.
| | - M Butlin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - T V Murphy
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia.
| | - A P Avolio
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - J K Phillips
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
39
|
Validation of Effective Therapeutic Targets for ADPKD Using Animal Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 933:71-84. [DOI: 10.1007/978-981-10-2041-4_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Rajagopalan R, Grochowski CM, Gilbert MA, Falsey AM, Coleman K, Romero R, Loomes KM, Piccoli DA, Devoto M, Spinner NB. Compound heterozygous mutations in NEK8 in siblings with end-stage renal disease with hepatic and cardiac anomalies. Am J Med Genet A 2015; 170:750-3. [PMID: 26697755 DOI: 10.1002/ajmg.a.37512] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/29/2015] [Indexed: 11/10/2022]
Abstract
We studied two brothers who presented in the newborn period with cardiac, renal, and hepatic anomalies that were initially suggestive of ALGS, although no mutations in JAG1 or NOTCH2 were identified. Exome sequencing demonstrated compound heterozygous mutations in the NEK8 gene (Never in mitosis A-related Kinase 8), a ciliary kinase indispensable for cardiac and renal development based on murine studies. The mutations included a c.2069_2070insC variant (p.Ter693LeufsTer86), and a c.1043C>T variant (p.Thr348Met) in the highly conserved RCC1 (Regulation of Chromosome Condensation 1) domain. The RCC1 domain is crucial for localization of the NEK8 protein to the centrosomes and cilia. Mutations in NEK8 have been previously reported in three fetuses (from a single family) with renal-hepatic-pancreatic dysplasia 2 (RHPD2), similar to Ivemark syndrome, and in three individuals with nephronophthisis (NPHP9). This is the third report of disease-causing mutations in the NEK8 gene in humans and only the second describing multi-organ involvement. The clinical features we describe differ from those in the previously published report in that (1) a pancreatic phenotype was not observed in the individuals reported here, (2) there were more prominent cardiac findings, (consistent with observations in murine models), and (3) we observed bile duct hypoplasia rather than ductal plate malformation. The patients reported here expand our understanding of the NEK8-associated phenotype. Our findings highlight the variable phenotypic expressivity and the spectrum of clinical manifestations due to mutations in the NEK8 gene.
Collapse
Affiliation(s)
- Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher M Grochowski
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melissa A Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexandra M Falsey
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Karlene Coleman
- Marcus Autism Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Rene Romero
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Kathleen M Loomes
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Piccoli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marcella Devoto
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Nancy B Spinner
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Jeewandara TM, Ameer OZ, Boyd R, Wyse BF, Underwood CF, Phillips JK. Protective cardiorenal effects of spironolactone in a rodent model of polycystic kidney disease. Clin Exp Pharmacol Physiol 2015; 42:353-60. [PMID: 25676668 DOI: 10.1111/1440-1681.12372] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/08/2015] [Accepted: 01/22/2015] [Indexed: 11/28/2022]
Abstract
Studies were performed to examine the contribution of aldosterone to the pathogenesis of cardiovascular and renal disease in a rodent model of genetic kidney disease. Spironolactone (20 mg/kg per day) was administered in water to mixed sex Lewis Polycystic Kidney (LPK) rats (n = 20) and control Lewis rats (n = 27) from 4 to 12 weeks of age. At 12 weeks of age, hypertension was reduced in female LPK rats; systolic blood pressure declined from 226.4 ± 26.8 mmHg in untreated rats and to 179.2 ± 3.2 mmHg in treated rats (P = 0.018). No similar effect on male or control rats was found. Water consumption and urine volume were significantly greater in LPK animals than in Lewis rats, and treatment reduced both variables by ~30% in LPK animals (P < 0.05). Proteinuria and the urinary protein-to-creatinine ratio were normalized in treated LPK relative to Lewis controls, and plasma creatinine levels were significantly reduced by treatment in LPK rats. Spironolactone did not alter kidney morphology in LPK rats (fibrosis or cyst size). Aortic vascular responses to noradrenaline and acetylcholine were sensitized and impaired in the LPK (P < 0.01). Aldosterone antagonism did not alter these responses or indicators of aortic structural remodelling. There was no treatment effect on left ventricular hypertrophy or elevated cardiac messenger RNA for β-myosin-heavy chain and brain natriuretic peptide in the LPK rats. However, perivascular fibrosis and messenger RNA for α-cardiac actin were normalized by spironolactone in LPK animals relative to Lewis controls. In conclusion, we have shown an important blood pressure independent effect whereby inhibition of aldosterone via spironolactone was able to retard both renal and cardiac disease progression in a rodent model of polycystic kidney disease.
Collapse
Affiliation(s)
- Thamarasee M Jeewandara
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Rahmati-Holasoo H, Masoudifard M, Ebrahimzadeh Mousavi H, Shokrpoor S, Tavakkoli A, Farazandemehr MS. Cystic lesions in the kidney of flower horn fish, hybrid cichlid. JOURNAL OF FISH DISEASES 2015; 38:833-838. [PMID: 25130925 DOI: 10.1111/jfd.12292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/21/2014] [Accepted: 06/22/2014] [Indexed: 06/03/2023]
Affiliation(s)
- H Rahmati-Holasoo
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - M Masoudifard
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - H Ebrahimzadeh Mousavi
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - S Shokrpoor
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - A Tavakkoli
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - M S Farazandemehr
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
43
|
Cantero MDR, Velázquez IF, Streets AJ, Ong ACM, Cantiello HF. The cAMP Signaling Pathway and Direct Protein Kinase A Phosphorylation Regulate Polycystin-2 (TRPP2) Channel Function. J Biol Chem 2015; 290:23888-96. [PMID: 26269590 DOI: 10.1074/jbc.m115.661082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Indexed: 11/06/2022] Open
Abstract
Polycystin-2 (PC2) is a TRP-type, Ca(2+)-permeable non-selective cation channel that plays an important role in Ca(2+) signaling in renal and non-renal cells. The effect(s) of the cAMP pathway and kinase mediated phosphorylation of PC2 seem to be relevant to PC2 trafficking and its interaction with polycystin-1. However, the role of PC2 phosphorylation in channel function is still poorly defined. Here we reconstituted apical membranes of term human syncytiotrophoblast (hST), containing endogenous PC2 (PC2hst), and in vitro translated channel protein (PC2iv). Addition of the catalytic subunit of PKA increased by 566% the spontaneous PC2hst channel activity in the presence of ATP. Interestingly, 8-Br-cAMP also stimulated spontaneous PC2hst channel activity in the absence of the exogenous kinase. Either stimulation was inhibited by addition of alkaline phosphatase, which in turn, was reversed by the phosphatase inhibitor vanadate. Neither maneuver modified the single channel conductance but instead increased channel mean open time. PKA directly phosphorylated PC2, which increased the mean open time but not the single channel conductance of the channel. PKA phosphorylation did not modify either R742X truncated or S829A-mutant PC2iv channel function. The data indicate that the cAMP pathway regulates PC2-mediated cation transport in the hST. The relevant PKA site for PC2 channel regulation centers on a single residue serine 829, in the carboxyl terminus.
Collapse
Affiliation(s)
- María del Rocío Cantero
- From the Cátedra de Biofísica, Facultad de Odontología, Universidad de Buenos Aires, C1122AAH Buenos Aires, Argentina and
| | - Irina F Velázquez
- From the Cátedra de Biofísica, Facultad de Odontología, Universidad de Buenos Aires, C1122AAH Buenos Aires, Argentina and
| | - Andrew J Streets
- Kidney Genetics Group, Academic Nephrology Unit, The Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX, United Kingdom
| | - Albert C M Ong
- Kidney Genetics Group, Academic Nephrology Unit, The Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX, United Kingdom
| | - Horacio F Cantiello
- From the Cátedra de Biofísica, Facultad de Odontología, Universidad de Buenos Aires, C1122AAH Buenos Aires, Argentina and
| |
Collapse
|
44
|
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
|
45
|
Bakey Z, Bihoreau MT, Piedagnel R, Delestré L, Arnould C, de Villiers AD, Devuyst O, Hoffmann S, Ronco P, Gauguier D, Lelongt B. The SAM domain of ANKS6 has different interacting partners and mutations can induce different cystic phenotypes. Kidney Int 2015; 88:299-310. [PMID: 26039630 DOI: 10.1038/ki.2015.122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/12/2015] [Accepted: 03/05/2015] [Indexed: 01/18/2023]
Abstract
The ankyrin repeat and sterile α motif (SAM) domain-containing six gene (Anks6) is a candidate for polycystic kidney disease (PKD). Originally identified in the PKD/Mhm(cy/+) rat model of PKD, the disease is caused by a mutation (R823W) in the SAM domain of the encoded protein. Recent studies support the etiological role of the ANKS6 SAM domain in human cystic diseases, but its function in kidney remains unknown. To investigate the role of ANKS6 in cyst formation, we screened an archive of N-ethyl-N-nitrosourea-treated mice and derived a strain carrying a missense mutation (I747N) within the SAM domain of ANKS6. This mutation is only six amino acids away from the PKD-causing mutation (R823W) in cy/+ rats. Evidence of renal cysts in these mice confirmed the crucial role of the SAM domain of ANKS6 in kidney function. Comparative phenotype analysis in cy/+ rats and our Anks6(I747N) mice further showed that the two models display noticeably different PKD phenotypes and that there is a defective interaction between ANKS6 with ANKS3 in the rat and between ANKS6 and BICC1 (bicaudal C homolog 1) in the mouse. Thus, our data demonstrate the importance of ANKS6 for kidney structure integrity and the essential mediating role of its SAM domain in the formation of protein complexes.
Collapse
Affiliation(s)
- Zeineb Bakey
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France
| | | | - Rémi Piedagnel
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France
| | - Laure Delestré
- 1] UPD University of Paris 05, Paris, France [2] INSERM, UMR_S1138, CRC, Paris, France
| | - Catherine Arnould
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France
| | - Alexandre d'Hotman de Villiers
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France
| | - Olivier Devuyst
- 1] UCL Medical School, Brussels, Belgium [2] University of Zurich, Zürich, Switzerland
| | - Sigrid Hoffmann
- Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Pierre Ronco
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France [3] AP-HP, Hôpital Tenon, Paris, France
| | - Dominique Gauguier
- 1] UPD University of Paris 05, Paris, France [2] INSERM, UMR_S1138, CRC, Paris, France [3] Institute of Cardiometabolism and Nutrition, University Pierre & Marie Curie, Hospital Pitié Salpetrière, Paris, France
| | - Brigitte Lelongt
- 1] Sorbonne Universités, UPMC Univ Paris 06, UMR_S1155, Paris, France [2] INSERM, UMR_S1155, Hôpital Tenon, Paris, France
| |
Collapse
|
46
|
Yochem J, Lažetić V, Bell L, Chen L, Fay D. C. elegans NIMA-related kinases NEKL-2 and NEKL-3 are required for the completion of molting. Dev Biol 2015; 398:255-66. [PMID: 25523392 PMCID: PMC4314388 DOI: 10.1016/j.ydbio.2014.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/29/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
Caenorhabditis elegans molting is a process during which the apical extracellular matrix of the epidermis, the cuticle, is remodeled through a process of degradation and re-synthesis. Using a genetic approach, we identified nekl-3 as essential for the completion of molting. NEKL-3 is highly similar to the mammalian NEK kinase family members NEK6 and NEK7. Animals homozygous for a hypomorphic mutation in nekl-3, sv3, had a novel molting defect in which the central body region, but not the head or tail, was unable to shed the old cuticle. In contrast, a null mutation in nekl-3, gk506, led to complete enclosure within the old cuticle. nekl-2, which is most similar to mammalian NEK8, was also essential for molting. Mosaic analyses demonstrated that NEKL-2 and NEKL-3 were specifically required within the large epidermal syncytium, hyp7, to facilitate molting. Consistent with this, NEKL-2 and NEKL-3 were expressed at the apical surface of hyp7 where they localized to small spheres or tubular structures. Inhibition of nekl-2, but not nekl-3, led to the mislocalization of LRP-1/megalin, a cell surface receptor for low-density lipoprotein (LDL)-binding proteins. In addition, nekl-2 inhibition led to the mislocalization of several other endosome-associated proteins. Notably, LRP-1 acts within hyp7 to facilitate completion of molting, suggesting at least one mechanism by which NEKL-2 may influence molting. Notably, our studies failed to reveal a requirement for NEKL-2 or NEKL-3 in cell division, a function reported for several mammalian NEKs including NEK6 and NEK7. Our findings provide the first genetic and in vivo evidence for a role of NEK family members in endocytosis, which may be evolutionarily conserved.
Collapse
Affiliation(s)
- John Yochem
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States; Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - Leslie Bell
- Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - Lihsia Chen
- Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - David Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States.
| |
Collapse
|
47
|
Sinigaglia C, Busengdal H, Lerner A, Oliveri P, Rentzsch F. Molecular characterization of the apical organ of the anthozoan Nematostella vectensis. Dev Biol 2015; 398:120-33. [PMID: 25478911 PMCID: PMC4300403 DOI: 10.1016/j.ydbio.2014.11.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/16/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
Abstract
Apical organs are sensory structures present in many marine invertebrate larvae where they are considered to be involved in their settlement, metamorphosis and locomotion. In bilaterians they are characterised by a tuft of long cilia and receptor cells and they are associated with groups of neurons, but their relatively low morphological complexity and dispersed phylogenetic distribution have left their evolutionary relationship unresolved. Moreover, since apical organs are not present in the standard model organisms, their development and function are not well understood. To provide a foundation for a better understanding of this structure we have characterised the molecular composition of the apical organ of the sea anemone Nematostella vectensis. In a microarray-based comparison of the gene expression profiles of planulae with either a wildtype or an experimentally expanded apical organ, we identified 78 evolutionarily conserved genes, which are predominantly or specifically expressed in the apical organ of Nematostella. This gene set comprises signalling molecules, transcription factors, structural and metabolic genes. The majority of these genes, including several conserved, but previously uncharacterized ones, are potentially involved in different aspects of the development or function of the long cilia of the apical organ. To demonstrate the utility of this gene set for comparative analyses, we further analysed the expression of a subset of previously uncharacterized putative orthologs in sea urchin larvae and detected expression for twelve out of eighteen of them in the apical domain. Our study provides a molecular characterization of the apical organ of Nematostella and represents an informative tool for future studies addressing the development, function and evolutionary history of apical organ cells.
Collapse
Affiliation(s)
- Chiara Sinigaglia
- Sars Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgt 55, 5008 Bergen, Norway
| | - Henriette Busengdal
- Sars Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgt 55, 5008 Bergen, Norway
| | - Avi Lerner
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Paola Oliveri
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Fabian Rentzsch
- Sars Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgt 55, 5008 Bergen, Norway.
| |
Collapse
|
48
|
Czarnecki PG, Gabriel GC, Manning DK, Sergeev M, Lemke K, Klena NT, Liu X, Chen Y, Li Y, San Agustin JT, Garnaas MK, Francis RJ, Tobita K, Goessling W, Pazour GJ, Lo CW, Beier DR, Shah JV. ANKS6 is the critical activator of NEK8 kinase in embryonic situs determination and organ patterning. Nat Commun 2015; 6:6023. [PMID: 25599650 PMCID: PMC4361001 DOI: 10.1038/ncomms7023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 12/02/2014] [Indexed: 11/09/2022] Open
Abstract
The ciliary kinase NEK8 plays a critical role in situs determination and cystic kidney disease, yet its exact function remains unknown. In this study, we identify ANKS6 as a target and activator of NEK8. ANKS6 requires NEK8 for localizing to the ciliary inversin compartment (IC) and activates NEK8 by binding to its kinase domain. Here we demonstrate the functional importance of this interaction through the analysis of two novel mouse mutations, Anks6(Streaker) and Nek8(Roc). Both display heterotaxy, cardiopulmonary malformations and cystic kidneys, a syndrome also characteristic of mutations in Invs and Nphp3, the other known components of the IC. The Anks6(Strkr) mutation decreases ANKS6 interaction with NEK8, precluding NEK8 activation. The Nek8(Roc) mutation inactivates NEK8 kinase function while preserving ANKS6 localization to the IC. Together, these data reveal the crucial role of NEK8 kinase activation within the IC, promoting proper left-right patterning, cardiopulmonary development and renal morphogenesis.
Collapse
Affiliation(s)
- Peter G Czarnecki
- 1] Department of Systems Biology, Harvard Medical School, 4 Blackfan Circle, HIM 568, Boston, Massachussetts 02115, USA [2] Renal Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA [3] Renal Division, Beth Israel Deaconess Medical Center, Boston, Massachussetts 02215, USA
| | - George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Danielle K Manning
- Genetics Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA
| | - Mikhail Sergeev
- 1] Department of Systems Biology, Harvard Medical School, 4 Blackfan Circle, HIM 568, Boston, Massachussetts 02115, USA [2] Renal Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA
| | - Kristi Lemke
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Nikolai T Klena
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Xiaoqin Liu
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Yu Chen
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - You Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Jovenal T San Agustin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachussetts 01655, USA
| | - Maija K Garnaas
- Genetics Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA
| | - Richard J Francis
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Kimimasa Tobita
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Wolfram Goessling
- Genetics Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachussetts 01655, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - David R Beier
- 1] Genetics Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA [2] Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | - Jagesh V Shah
- 1] Department of Systems Biology, Harvard Medical School, 4 Blackfan Circle, HIM 568, Boston, Massachussetts 02115, USA [2] Renal Division, Brigham and Women's Hospital, Boston, Massachussetts 02115, USA
| |
Collapse
|
49
|
Moraes EC, Meirelles GV, Honorato RV, de Souza TDACB, de Souza EE, Murakami MT, de Oliveira PSL, Kobarg J. Kinase inhibitor profile for human nek1, nek6, and nek7 and analysis of the structural basis for inhibitor specificity. Molecules 2015; 20:1176-91. [PMID: 25591119 PMCID: PMC6272266 DOI: 10.3390/molecules20011176] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/05/2015] [Indexed: 12/11/2022] Open
Abstract
Human Neks are a conserved protein kinase family related to cell cycle progression and cell division and are considered potential drug targets for the treatment of cancer and other pathologies. We screened the activation loop mutant kinases hNek1 and hNek2, wild-type hNek7, and five hNek6 variants in different activation/phosphorylation statesand compared them against 85 compounds using thermal shift denaturation. We identified three compounds with significant Tm shifts: JNK Inhibitor II for hNek1(Δ262-1258)-(T162A), Isogranulatimide for hNek6(S206A), andGSK-3 Inhibitor XIII for hNek7wt. Each one of these compounds was also validated by reducing the kinases activity by at least 25%. The binding sites for these compounds were identified by in silico docking at the ATP-binding site of the respective hNeks. Potential inhibitors were first screened by thermal shift assays, had their efficiency tested by a kinase assay, and were finally analyzed by molecular docking. Our findings corroborate the idea of ATP-competitive inhibition for hNek1 and hNek6 and suggest a novel non-competitive inhibition for hNek7 in regard to GSK-3 Inhibitor XIII. Our results demonstrate that our approach is useful for finding promising general and specific hNekscandidate inhibitors, which may also function as scaffolds to design more potent and selective inhibitors.
Collapse
Affiliation(s)
- Eduardo Cruz Moraes
- LaboratórioNacional de Biociências, Centro Nacional de PesquisaemEnergia e Materiais, Campinas, 13083-970 SP, Brazil.
| | - Gabriela Vaz Meirelles
- LaboratórioNacional de Biociências, Centro Nacional de PesquisaemEnergia e Materiais, Campinas, 13083-970 SP, Brazil.
| | - Rodrigo Vargas Honorato
- LaboratórioNacional de Biociências, Centro Nacional de PesquisaemEnergia e Materiais, Campinas, 13083-970 SP, Brazil.
| | | | - Edmarcia Elisa de Souza
- LaboratórioNacional de Biociências, Centro Nacional de PesquisaemEnergia e Materiais, Campinas, 13083-970 SP, Brazil.
| | - Mario Tyago Murakami
- LaboratórioNacional de Biociências, Centro Nacional de PesquisaemEnergia e Materiais, Campinas, 13083-970 SP, Brazil.
| | | | - Jörg Kobarg
- Programa de Pós-graduação em Biologia Funcional e Molecular, Departamento de Bioquímica e BiologiaTecidual, Instituto de Biologia, UniversidadeEstadual de Campinas, Campinas, 13083-862 SP, Brazil.
| |
Collapse
|
50
|
Huang L, Xiao A, Wecker A, McBride DA, Choi SY, Zhou W, Lipschutz JH. A possible zebrafish model of polycystic kidney disease: knockdown of wnt5a causes cysts in zebrafish kidneys. J Vis Exp 2014:52156. [PMID: 25489842 PMCID: PMC4354438 DOI: 10.3791/52156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Polycystic kidney disease (PKD) is one of the most common causes of end-stage kidney disease, a devastating disease for which there is no cure. The molecular mechanisms leading to cyst formation in PKD remain somewhat unclear, but many genes are thought to be involved. Wnt5a is a non-canonical glycoprotein that regulates a wide range of developmental processes. Wnt5a works through the planar cell polarity (PCP) pathway that regulates oriented cell division during renal tubular cell elongation. Defects of the PCP pathway have been found to cause kidney cyst formation. Our paper describes a method for developing a zebrafish cystic kidney disease model by knockdown of the wnt5a gene with wnt5a antisense morpholino (MO) oligonucleotides. Tg(wt1b:GFP) transgenic zebrafish were used to visualize kidney structure and kidney cysts following wnt5a knockdown. Two distinct antisense MOs (AUG - and splice-site) were used and both resulted in curly tail down phenotype and cyst formation after wnt5a knockdown. Injection of mouse Wnt5a mRNA, resistant to the MOs due to a difference in primary base pair structure, rescued the abnormal phenotype, demonstrating that the phenotype was not due to "off-target" effects of the morpholino. This work supports the validity of using a zebrafish model to study wnt5a function in the kidney.
Collapse
Affiliation(s)
- Liwei Huang
- Department of Medicine, Eastern Virginia Medical School;
| | - An Xiao
- Department of Medicine, Eastern Virginia Medical School
| | - Andrea Wecker
- Department of Medicine, Eastern Virginia Medical School
| | | | - Soo Young Choi
- Department of Medicine, Medical University of South Carolina
| | - Weibin Zhou
- Department of Pediatrics, University of Michigan
| | | |
Collapse
|