1
|
Tiberio F, Coda ARD, Tosi DD, Luzi D, Polito L, Liso A, Lattanzi W. Mechanobiology and Primary Cilium in the Pathophysiology of Bone Marrow Myeloproliferative Diseases. Int J Mol Sci 2024; 25:8860. [PMID: 39201546 PMCID: PMC11354938 DOI: 10.3390/ijms25168860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Philadelphia-Negative Myeloproliferative neoplasms (MPNs) are a diverse group of blood cancers leading to excessive production of mature blood cells. These chronic diseases, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), can significantly impact patient quality of life and are still incurable in the vast majority of the cases. This review examines the mechanobiology within a bone marrow niche, emphasizing the role of mechanical cues and the primary cilium in the pathophysiology of MPNs. It discusses the influence of extracellular matrix components, cell-cell and cell-matrix interactions, and mechanosensitive structures on hematopoietic stem cell (HSC) behavior and disease progression. Additionally, the potential implications of the primary cilium as a chemo- and mechanosensory organelle in bone marrow cells are explored, highlighting its involvement in signaling pathways crucial for hematopoietic regulation. This review proposes future research directions to better understand the dysregulated bone marrow niche in MPNs and to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Federica Tiberio
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Domiziano Dario Tosi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Debora Luzi
- S.C. Oncoematologia, Azienda Ospedaliera di Terni, 05100 Terni, Italy;
| | - Luca Polito
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Arcangelo Liso
- Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
2
|
Luo J, Zhang Y, Jayaprakash S, Zhuang L, He J. Cross-Species Insights into Autosomal Dominant Polycystic Kidney Disease: Provide an Alternative View on Research Advancement. Int J Mol Sci 2024; 25:5646. [PMID: 38891834 PMCID: PMC11171680 DOI: 10.3390/ijms25115646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a prevalent hereditary disorder that affects the kidneys, characterized by the development of an excessive number of fluid-filled cysts of varying sizes in both kidneys. Along with the progression of ADPKD, these enlarged cysts displace normal kidney tissue, often accompanied by interstitial fibrosis and inflammation, and significantly impair renal function, leading to end-stage renal disease. Currently, the precise mechanisms underlying ADPKD remain elusive, and a definitive cure has yet to be discovered. This review delineates the epidemiology, pathological features, and clinical diagnostics of ADPKD or ADPKD-like disease across human populations, as well as companion animals and other domesticated species. A light has been shed on pivotal genes and biological pathways essential for preventing and managing ADPKD, which underscores the importance of cross-species research in addressing this complex condition. Treatment options are currently limited to Tolvaptan, dialysis, or surgical excision of large cysts. However, comparative studies of ADPKD across different species hold promise for unveiling novel insights and therapeutic strategies to combat this disease.
Collapse
Affiliation(s)
- Jianing Luo
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Yuan Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Sakthidasan Jayaprakash
- Department of Biotechnology, Hindustan Institute of Technology and Science, Tamil Nadu 603103, India;
| | - Lenan Zhuang
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Jin He
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| |
Collapse
|
3
|
Pyrshev K, Stavniichuk A, Tomilin VN, Khayyat NH, Ren G, Kordysh M, Zaika O, Mamenko M, Pochynyuk O. TRPV4 functional status in cystic cells regulates cystogenesis in autosomal recessive polycystic kidney disease during variations in dietary potassium. Physiol Rep 2023; 11:e15641. [PMID: 36946001 PMCID: PMC10031299 DOI: 10.14814/phy2.15641] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/23/2023] Open
Abstract
Mechanosensitive TRPV4 channel plays a dominant role in maintaining [Ca2+ ]i homeostasis and flow-sensitive [Ca2+ ]i signaling in the renal tubule. Polycystic kidney disease (PKD) manifests as progressive cyst growth due to cAMP-dependent fluid secretion along with deficient mechanosensitivity and impaired TRPV4 activity. Here, we tested how regulation of renal TRPV4 function by dietary K+ intake modulates the rate of cystogenesis and mechanosensitive [Ca2+ ]i signaling in cystic cells of PCK453 rats, a homologous model of human autosomal recessive PKD (ARPKD). One month treatment with both high KCl (5% K+ ) and KB/C (5% K+ with bicarbonate/citrate) diets significantly increased TRPV4 levels when compared to control (0.9% K+ ). High KCl diet caused an increased TRPV4-dependent Ca2+ influx, and partial restoration of mechanosensitivity in freshly isolated monolayers of cystic cells. Unexpectedly, high KB/C diet induced an opposite effect by reducing TRPV4 activity and worsening [Ca2+ ]i homeostasis. Importantly, high KCl diet decreased cAMP, whereas high KB/C diet further increased cAMP levels in cystic cells (assessed as AQP2 distribution). At the systemic level, high KCl diet fed PCK453 rats had significantly lower kidney-to-bodyweight ratio and reduced cystic area. These beneficial effects were negated by a concomitant administration of an orally active TRPV4 antagonist, GSK2193874, resulting in greater kidney weight, accelerated cystogenesis, and augmented renal injury. High KB/C diet also exacerbated renal manifestations of ARPKD, consistent with deficient TRPV4 activity in cystic cells. Overall, we demonstrate that TRPV4 channel activity negatively regulates cAMP levels in cystic cells thus attenuating (high activity) or accelerating (low activity) ARPKD progression.
Collapse
Affiliation(s)
- Kyrylo Pyrshev
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Anna Stavniichuk
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Viktor N. Tomilin
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Naghmeh Hassanzadeh Khayyat
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Guohui Ren
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Mariya Kordysh
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Oleg Zaika
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Mykola Mamenko
- Department of PhysiologyAugusta UniversityAugustaGeorgiaUSA
| | - Oleh Pochynyuk
- Department of Integrative Biology and PharmacologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
4
|
Mohamed R, Liu Y, Kistler AD, Harris PC, Thangaraju M. Netrin-1 Overexpression Induces Polycystic Kidney Disease: A Novel Mechanism Contributing to Cystogenesis in Autosomal Dominant Polycystic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:862-875. [PMID: 35358475 DOI: 10.1016/j.ajpath.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
Despite recent advances in understanding the pathogenesis of polycystic kidney disease (PKD), the underlying molecular mechanisms involved in cystogenesis are not fully understood. This study describes a novel pathway involved in cyst formation. Transgenic mice overexpressing netrin-1 in proximal tubular cells showed increased production and urinary excretion of netrin-1. Although no cysts were detectable immediately after birth, numerous small cysts were evident by the age of 4 weeks, and disease was accelerated along with age. Surprisingly, cyst formation in the kidney was restricted to male mice, with 80% penetrance. However, ovariectomy induced kidney cyst growth in netrin-1-overexpressing female mice. Cyst development in males was associated with albuminuria and polyuria and increased cAMP excretion in netrin-1 transgenic mice. Netrin-1 overexpression significantly increased extracellular signal-regulated kinase and focal adhesion kinase phosphorylation and vimentin expression. Interestingly, p53 expression was increased but in an inactive form. Furthermore, netrin-1 expression was increased in cystic epithelia and urine of various rodent models of PKD. siRNA-mediated suppression of netrin-1 significantly reduced cyst growth and improved kidney function in netrin-1 transgenic mice and in two genetic animal models of PKD. Together, these data demonstrate that netrin-1 up-regulation induced cyst formation in autosomal dominant PKD.
Collapse
Affiliation(s)
- Riyaz Mohamed
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia.
| | - Yang Liu
- Department of Internal Medicine, Cantonal Hospital Frauenfeld, Frauenfeld, Switzerland
| | - Andreas D Kistler
- Department of Internal Medicine, Cantonal Hospital Frauenfeld, Frauenfeld, Switzerland
| | - Peter C Harris
- Division of Nephrology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia; Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
5
|
Cabrita I, Talbi K, Kunzelmann K, Schreiber R. Loss of PKD1 and PKD2 share common effects on intracellular Ca 2+ signaling. Cell Calcium 2021; 97:102413. [PMID: 33915319 DOI: 10.1016/j.ceca.2021.102413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022]
Abstract
In polycystic kidney disease (PKD) multiple bilateral renal cysts gradually enlarge causing a decline in renal function. Transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and TMEM16A (anoctamin 1) drive cyst enlargement. We demonstrated recently that a loss of PKD1 increases expression and function of TMEM16A in murine kidneys and in mouse M1 collecting duct cells. The data demonstrated that TMEM16A contributes essentially to cyst growth by upregulating intracellular Ca2+ signaling. Enhanced expression of TMEM16A and Ca2+ signaling increased both cell proliferation and fluid secretion, which suggested inhibition of TMEM16A as a novel therapy in ADPKD. About 15 % of all ADPKD cases are caused by mutations in PKD2. To analyze the effects of loss of function of PKD2 on Ca2+ signaling, we knocked-down Pkd2 in mouse primary renal epithelial cells in the present study, using viral transfection of shRNA. Unlike in Pkd1-/- cells, knockdown of PKD2 lowered basal Ca2+ and augmented store-operated Ca2+ entry, which was both independent of TMEM16A. However, disease causing purinergic Ca2+ store release was enhanced, similar to that observed in Pkd1-/- renal epithelial cells. The present data suggest pharmacological inhibition of TMEM16A as a treatment in ADPKD caused by mutations in both PKD1 and PKD2.
Collapse
Affiliation(s)
- Ines Cabrita
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Khaoula Talbi
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| |
Collapse
|
6
|
Cardiac Involvement in Autosomal Dominant Polycystic Kidney Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disorders are the main complication in autosomal dominant polycystic kidney disease (ADPKD). contributing to both morbidity and mortality. This review considers clinical studies unveiling cardiovascular features in patients with ADPKD. Additionally, it focuses on basic science studies addressing the dysfunction of the polycystin proteins located in the cardiovascular system as a contributing factor to cardiovascular abnormalities. In particular, the effects of polycystin proteins’ deficiency on the cardiomyocyte function have been considered.
Collapse
|
7
|
Park SJ, Li C, Chen YM. Endoplasmic Reticulum Calcium Homeostasis in Kidney Disease: Pathogenesis and Therapeutic Targets. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:256-265. [PMID: 33245915 DOI: 10.1016/j.ajpath.2020.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/14/2020] [Accepted: 11/06/2020] [Indexed: 01/15/2023]
Abstract
Calcium (Ca2+) homeostasis is a crucial determinant of cellular function and survival. Endoplasmic reticulum (ER) acts as the largest intracellular Ca2+ store that maintains Ca2+ homeostasis through the ER Ca2+ uptake pump, sarco/ER Ca2+ ATPase, ER Ca2+ release channels, inositol 1,4,5-trisphosphate receptor channel, ryanodine receptor, and Ca2+-binding proteins inside of the ER lumen. Alterations in ER homeostasis trigger ER Ca2+ depletion and ER stress, which have been associated with the development of a variety of diseases. In addition, recent studies have highlighted the role of ER Ca2+ imbalance caused by dysfunction of sarco/ER Ca2+ ATPase, ryanodine receptor, and inositol 1,4,5-trisphosphate receptor channel in various kidney diseases. Despite progress in the understanding of the importance of these ER Ca2+ channels, pumps, and binding proteins in the pathogenesis of kidney disease, treatment is still lacking. This mini-review is focused on: i) Ca2+ homeostasis in the ER, ii) ER Ca2+ dyshomeostasis and apoptosis, and iii) altered ER Ca2+ homeostasis in kidney disease, including podocytopathy, diabetic nephropathy, albuminuria, autosomal dominant polycystic kidney disease, and ischemia/reperfusion-induced acute kidney injury.
Collapse
Affiliation(s)
- Sun-Ji Park
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Chuang Li
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ying Maggie Chen
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
8
|
Novel Potential Application of Chitosan Oligosaccharide for Attenuation of Renal Cyst Growth in the Treatment of Polycystic Kidney Disease. Molecules 2020; 25:molecules25235589. [PMID: 33261193 PMCID: PMC7730275 DOI: 10.3390/molecules25235589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Chitosan oligosaccharide (COS), a natural polymer derived from chitosan, exerts several biological activities including anti-inflammation, anti-tumor, anti-metabolic syndrome, and drug delivery enhancer. Since COS is vastly distributed to kidney and eliminated in urine, it may have a potential advantage as the therapeutics of kidney diseases. Polycystic kidney disease (PKD) is a common genetic disorder characterized by multiple fluid-filled cysts, replacing normal renal parenchyma and leading to impaired renal function and end-stage renal disease (ESRD). The effective treatment for PKD still needs to be further elucidated. Interestingly, AMP-activated protein kinase (AMPK) has been proposed as a drug target for PKD. This study aimed to investigate the effect of COS on renal cyst enlargement and its underlying mechanisms. We found that COS at the concentrations of 50 and 100 µg/mL decreased renal cyst growth without cytotoxicity, as measured by MTT assay. Immunoblotting analysis showed that COS at 100 µg/mL activated AMPK, and this effect was abolished by STO-609, a calcium/calmodulin-dependent protein kinase kinase beta (CaMKKβ) inhibitor. Moreover, COS elevated the level of intracellular calcium. These results suggest that COS inhibits cyst progression by activation of AMPK via CaMKKβ. Therefore, COS may hold the potential for pharmaceutical application in PKD.
Collapse
|
9
|
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
|
10
|
TMEM16A drives renal cyst growth by augmenting Ca 2+ signaling in M1 cells. J Mol Med (Berl) 2020; 98:659-671. [PMID: 32185407 PMCID: PMC7220898 DOI: 10.1007/s00109-020-01894-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Abstract Polycystic kidney disease (PKD) leads to continuous decline of renal function by growth of renal cysts. Enhanced proliferation and transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and Ca2+-activated TMEM16A Cl− channels is thought to cause an increase in cyst volume. Recent work shows the pro-proliferative role of the Ca2+ activated Cl− channel TMEM16A (anoctamin 1), and demonstrates the essential contribution of TMEM16A to CFTR-dependent Cl− secretion. The present data demonstrate an increase in intracellular Ca2+ ([Ca2+]i) signals and Cl− secretion by TMEM16A, in renal collecting duct principle cells from dog (MDCK) and mouse (M1) as well as primary tubular epithelial cells from PKD1−/− knockout mice. M1 organoids proliferated, increased expression of TMEM16A, and secreted Cl− upon knockdown of endogenous polycystin 1 or 2 (PKD1,2), by retroviral transfection with shPKD1 and shPKD2, respectively. Knockdown of PKD1 or PKD2 increased basal intracellular Ca2+ levels and enhanced purinergic Ca2+ release from endoplasmic reticulum. In contrast, ryanodine receptors were found not to be expressed in mouse renal epithelial cells and caffeine had no effects on [Ca2+]i. Ca2+ signals, proliferation, and Cl− secretion were largely reduced by knockdown or blockade of TMEM16A. TMEM16A may be therefore important for enhanced Ca2+ release from IP3-sensitive Ca2+ stores in polycystic kidney disease. Key messages • ADPKD leads to continuous decline of renal function by growth of renal cysts. • Knockdown of PKD1 or PKD2 increases TMEM16A expression. • TMEM16A enhanced intracellular Ca2+ signals, Cl− secretion, and proliferation. • TMEM16A contributes to cyst growth in ADPKD. Electronic supplementary material The online version of this article (10.1007/s00109-020-01894-y) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Weydert C, Decuypere JP, De Smedt H, Janssens P, Vennekens R, Mekahli D. Fundamental insights into autosomal dominant polycystic kidney disease from human-based cell models. Pediatr Nephrol 2019; 34:1697-1715. [PMID: 30215095 DOI: 10.1007/s00467-018-4057-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
Several animal- and human-derived models are used in autosomal dominant polycystic kidney disease (ADPKD) research to gain insight in the disease mechanism. However, a consistent correlation between animal and human ADPKD models is lacking. Therefore, established human-derived models are relevant to affirm research results and translate findings into a clinical set-up. In this review, we give an extensive overview of the existing human-based cell models. We discuss their source (urine, nephrectomy and stem cell), immortalisation procedures, genetic engineering, kidney segmental origin and characterisation with nephron segment markers. We summarise the most studied pathways and lessons learned from these different ADPKD models. Finally, we issue recommendations for the derivation of human-derived cell lines and for experimental set-ups with these cell lines.
Collapse
Affiliation(s)
- Caroline Weydert
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Jean-Paul Decuypere
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Peter Janssens
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium
- Department of Nephrology, University Hospitals Brussels, Brussels, Belgium
| | - Rudi Vennekens
- VIB Center for Brain and Disease Research, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, GPURE, KU Leuven, Leuven, Belgium.
- Department of Pediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
12
|
Fu W, Zhou W, Chu PK, Yu X. Inherent Chemotherapeutic Anti‐Cancer Effects of Low‐Dimensional Nanomaterials. Chemistry 2019; 25:10995-11006. [DOI: 10.1002/chem.201901841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Wen Fu
- Materials Interference CenterShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences Shenzhen 518055 P.R. China
- Shenzhen College of Advanced TechnologyUniversity of Chinese Academy of Sciences Shenzhen 518055 P.R. China
| | - Wenhua Zhou
- Materials Interference CenterShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences Shenzhen 518055 P.R. China
| | - Paul K. Chu
- Department of Physics and Department of Materials Science and EngineeringCity University of Hong Kong Tat Chee Avenue Kowloon, Hong Kong P.R. China
| | - Xue‐Feng Yu
- Materials Interference CenterShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences Shenzhen 518055 P.R. China
| |
Collapse
|
13
|
Nishimura Y, Kasahara K, Shiromizu T, Watanabe M, Inagaki M. Primary Cilia as Signaling Hubs in Health and Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801138. [PMID: 30643718 PMCID: PMC6325590 DOI: 10.1002/advs.201801138] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/20/2018] [Indexed: 05/13/2023]
Abstract
Primary cilia detect extracellular cues and transduce these signals into cells to regulate proliferation, migration, and differentiation. Here, the function of primary cilia as signaling hubs of growth factors and morphogens is in focus. First, the molecular mechanisms regulating the assembly and disassembly of primary cilia are described. Then, the role of primary cilia in mediating growth factor and morphogen signaling to maintain human health and the potential mechanisms by which defects in these pathways contribute to human diseases, such as ciliopathy, obesity, and cancer are described. Furthermore, a novel signaling pathway by which certain growth factors stimulate cell proliferation through suppression of ciliogenesis is also described, suggesting novel therapeutic targets in cancer.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Kousuke Kasahara
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Takashi Shiromizu
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masatoshi Watanabe
- Department of Oncologic PathologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masaki Inagaki
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| |
Collapse
|
14
|
Leal-Esteban LC, Rothé B, Fortier S, Isenschmid M, Constam DB. Role of Bicaudal C1 in renal gluconeogenesis and its novel interaction with the CTLH complex. PLoS Genet 2018; 14:e1007487. [PMID: 29995892 PMCID: PMC6056059 DOI: 10.1371/journal.pgen.1007487] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/23/2018] [Accepted: 06/13/2018] [Indexed: 01/06/2023] Open
Abstract
Altered glucose and lipid metabolism fuel cystic growth in polycystic kidneys, but the cause of these perturbations is unclear. Renal cysts also associate with mutations in Bicaudal C1 (Bicc1) or in its self-polymerizing sterile alpha motif (SAM). Here, we found that Bicc1 maintains normoglycemia and the expression of the gluconeogenic enzymes FBP1 and PEPCK in kidneys. A proteomic screen revealed that Bicc1 interacts with the C-Terminal to Lis-Homology domain (CTLH) complex. Since the orthologous Gid complex in S. cerevisae targets FBP1 and PEPCK for degradation, we mapped the topology among CTLH subunits and found that SAM-mediated binding controls Bicc1 protein levels, whereas Bicc1 inhibited the accumulation of several CTLH subunits. Under the conditions analyzed, Bicc1 increased FBP1 protein levels independently of the CTLH complex. Besides linking Bicc1 to cell metabolism, our findings reveal new layers of complexity in the regulation of renal gluconeogenesis compared to lower eukaryotes. Polycystic kidney diseases (PKD) are incurable inherited chronic disorders marked by fluid-filled cysts that frequently cause renal failure. A glycolytic metabolism reminiscent of cancerous cells accelerates cystic growth, but the mechanism underlying such metabolic re-wiring is poorly understood. PKD-like cystic kidneys also develop in mice that lack the RNA-binding protein Bicaudal-C (Bicc1), and mutations in a single copy of human BICC1 associate with renal cystic dysplasia. Here, we report that Bicc1 regulates renal gluconeogenesis. A screen for interacting factors revealed that Bicc1 binds the C-Terminal to Lis-Homology domain (CTLH) complex, which in lower eukaryotes mediates degradation of gluconeogenic enzymes. By contrast, Bicc1 and the mammalian CTLH complex regulated each other, and Bicc1 stimulated the accumulation of the rate-limiting gluconeogenic enzyme even in cells depleted of CTLH subunits. Our finding that Bicc1 is required for normoglycemia implies that renal gluconeogenesis may be important to inhibit cyst formation.
Collapse
Affiliation(s)
- Lucia Carolina Leal-Esteban
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Benjamin Rothé
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Simon Fortier
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Manuela Isenschmid
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Daniel B. Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
15
|
Abstract
The liver and kidneys are often similarly affected by a single disease. This is the case in metabolic, immunological, toxic, and infectious diseases, and in the different congenital malformation syndromes. Also, an enzymatic defect in an otherwise healthy liver or the consequences of advanced liver disease by itself can cause kidney disease as a secondary phenomenon. In this review, we describe numerous pathogenic mechanisms leading to dysfunction or malformations of the liver and kidneys in children. We encourage multidisciplinary management for optimal care. A combined liver-kidney transplantation is sometimes needed.
Collapse
|
16
|
Di Mise A, Tamma G, Ranieri M, Centrone M, van den Heuvel L, Mekahli D, Levtchenko EN, Valenti G. Activation of Calcium-Sensing Receptor increases intracellular calcium and decreases cAMP and mTOR in PKD1 deficient cells. Sci Rep 2018; 8:5704. [PMID: 29632324 PMCID: PMC5890283 DOI: 10.1038/s41598-018-23732-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/13/2018] [Indexed: 12/25/2022] Open
Abstract
Clinical and fundamental research suggest that altered calcium and cAMP signaling might be the most proximal events in ADPKD pathogenesis. Cells from ADPKD cysts have a reduced resting cytosolic calcium [Ca2+]i and increased cAMP levels. CaSR plays an essential role in regulating calcium homeostasis. Its activation is associated with [Ca2+]i increase and cAMP decrease, making CaSR a possible therapeutic target. Human conditionally immortalized Proximal Tubular Epithelial cells (ciPTEC) with stable knockdown of PKD1 (ciPTEC-PC1KD) and ciPTEC generated from an ADPKD1 patient (ciPTEC-PC1Pt) were used as experimental tools. CaSR functional expression was confirmed by studies showing that the calcimimetic NPS-R568 induced a significant increase in [Ca2+]i in ciPTEC-PC1KD and ciPTEC-PC1Pt. Resting [Ca2+]i were significantly lower in ciPTEC-PC1KD with respect to ciPTECwt, confirming calcium dysregulation. As in native cyst cells, significantly higher cAMP levels and mTOR activity were found in ciPTEC-PC1KD compared to ciPTECwt. Of note, NPS-R568 treatment significantly reduced intracellular cAMP and mTOR activity in ciPTEC-PC1KD and ciPTEC-PC1Pt. To conclude, we demonstrated that selective CaSR activation in human ciPTEC carrying PKD1 mutation increases [Ca2+]i, reduces intracellular cAMP and mTOR activity, reversing the principal dysregulations considered the most proximal events in ADPKD pathogenesis, making CaSR a possible candidate as therapeutic target.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70125, Italy.
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70125, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, Roma, 00136, Italy
| | - Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70125, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70125, Italy
| | - Lambertus van den Heuvel
- Department of Pediatric Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, 6525 HP, The Netherlands
| | - Djalila Mekahli
- Department of Pediatric Nephrology, University Hospital Gasthuisberg, Leuven, 3000, Belgium.,Department of Development & Regeneration, University of Leuven (KU Leuven), Leuven, 3000, Belgium
| | - Elena N Levtchenko
- Department of Pediatric Nephrology, University Hospital Gasthuisberg, Leuven, 3000, Belgium.,Department of Development & Regeneration, University of Leuven (KU Leuven), Leuven, 3000, Belgium
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70125, Italy. .,Istituto Nazionale di Biostrutture e Biosistemi, Roma, 00136, Italy. .,Center of Excellence in Comparative Genomics (CEGBA), University of Bari, Bari, 70125, Italy.
| |
Collapse
|
17
|
Ranjzad F, Aghdami N, Tara A, Mohseni M, Moghadasali R, Basiri A. Identification of Three Novel Frameshift Mutations in the PKD1 Gene in Iranian Families with Autosomal Dominant Polycystic Kidney Disease Using Efficient Targeted Next-Generation Sequencing. Kidney Blood Press Res 2018; 43:471-478. [PMID: 29590654 DOI: 10.1159/000488471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/14/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common inherited cystic kidney diseases caused by mutations in two large multi-exon genes, PKD1 and PKD2. High allelic heterogeneity and duplication of PKD1 exons 1-32 as six pseudo genes on chromosome 16 complicate molecular analysis of this disease. METHODS We applied targeted next-generation sequencing (NGS) in 9 non-consanguineous unrelated Iranian families with ADPKD to identify the genes hosting disease-causing mutations. This approach was confirmed by Sanger sequencing. RESULTS Here, we determined three different novel frameshift mutations and four previously reported nonsense mutations in the PKD1 gene encoding polycystin1 in heterozygotes. CONCLUSION This study demonstrates the effectiveness of NGS in significantly reducing the cost and time for simultaneous sequence analysis of PKD1 and PKD2, simplifying the genetic diagnostics of ADPKD. Although a probable correlation between the mutation types and phenotypic outcome is possible, however for more extensive studies in future, the consideration of renal hypouricemia (RHUC) and PKD1 coexistence may be helpful. The novel frameshift mutations reported by this study are p. Q1997X, P. D73X and p. V336X.
Collapse
Affiliation(s)
- Fariba Ranjzad
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ahmad Tara
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Mohseni
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Tomilin V, Reif GA, Zaika O, Wallace DP, Pochynyuk O. Deficient transient receptor potential vanilloid type 4 function contributes to compromised [Ca 2+] i homeostasis in human autosomal-dominant polycystic kidney disease cells. FASEB J 2018; 32:4612-4623. [PMID: 29553832 DOI: 10.1096/fj.201701535rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a devastating disorder that is characterized by a progressive decline in renal function as a result of the development of fluid-filled cysts. Defective flow-mediated [Ca2+]i responses and disrupted [Ca2+]i homeostasis have been repeatedly associated with cyst progression in ADPKD. We have previously demonstrated that the transient receptor potential vanilloid type 4 (TRPV4) channel is imperative for flow-mediated [Ca2+]i responses in murine distal renal tubule cells. To determine whether compromised TRPV4 function contributes to aberrant Ca2+ regulation in ADPKD, we assessed TRPV4 function in primary cells that were cultured from ADPKD and normal human kidneys (NHKs). Single-channel TRPV4 activity and TRPV4-dependent Ca2+ influxes were drastically reduced in ADPKD cells, which correlated with distorted [Ca2+]i signaling. Whereas total TRPV4 protein levels were comparable in NHK and ADPKD cells, we detected a marked decrease in TRPV4 glycosylation in ADPKD cells. Tunicamycin-induced deglycosylation inhibited TRPV4 activity and compromised [Ca2+]i signaling in NHK cells. Overall, we demonstrate that TRPV4 glycosylation and channel activity are diminished in human ADPKD cells compared with NHK cells, and that this contributes significantly to the distorted [Ca2+]i dynamics. We propose that TRPV4 stimulation may be beneficial for restoring [Ca2+]i homeostasis in cyst cells, thereby interfering with ADPKD progression.-Tomilin, V., Reif, G. A., Zaika, O., Wallace, D. P., Pochynyuk, O. Deficient transient receptor potential vanilloid type 4 function contributes to compromised [Ca2+]i homeostasis in human autosomal-dominant polycystic kidney disease cells.
Collapse
Affiliation(s)
- Viktor Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA; and
| | - Gail A Reif
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA; and
| | - Darren P Wallace
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA; and
| |
Collapse
|
19
|
Janssens P, Weydert C, De Rechter S, Wissing KM, Liebau MC, Mekahli D. Expanding the role of vasopressin antagonism in polycystic kidney diseases: From adults to children? Pediatr Nephrol 2018; 33:395-408. [PMID: 28455745 DOI: 10.1007/s00467-017-3672-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/21/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Polycystic kidney disease (PKD) encompasses a group of genetic disorders that are common causes of renal failure. The two classic forms of PKD are autosomal recessive polycystic kidney disease (ARPKD) and autosomal dominant polycystic kidney disease (ADPKD). Despite their clinical differences, ARPKD and ADPKD share many similarities. Altered intracellular Ca2+ and increased cyclic adenosine monophosphate (cAMP) concentrations have repetitively been described as central anomalies that may alter signaling pathways leading to cyst formation. The vasopressin V2 receptor (V2R) antagonist tolvaptan lowers cAMP in cystic tissues and slows renal cystic progression and kidney function decline when given over 3 years in adult ADPKD patients. Tolvaptan is currently approved for the treatment of rapidly progressive disease in adult ADPKD patients. On the occasion of the recent initiation of a clinical trial with tolvaptan in pediatric ADPKD patients, we aim to describe the most important aspects in the literature regarding the AVP-cAMP axis and the clinical use of tolvaptan in PKD.
Collapse
Affiliation(s)
- Peter Janssens
- Laboratory of Pediatrics, University Hospitals Leuven, Leuven, Belgium. .,Department of Nephrology, University Hospitals Brussel, Brussel, Belgium.
| | - Caroline Weydert
- Laboratory of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Stephanie De Rechter
- Laboratory of Pediatrics, University Hospitals Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | | | - Max Christoph Liebau
- Pediatric Nephrology, Department of Pediatrics and Center for Molecular Medicine, University Hospital of Cologne, Cologne, Germany.,Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Djalila Mekahli
- Laboratory of Pediatrics, University Hospitals Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Fliniaux I, Germain E, Farfariello V, Prevarskaya N. TRPs and Ca2+ in cell death and survival. Cell Calcium 2018; 69:4-18. [DOI: 10.1016/j.ceca.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
|
21
|
Li W, Lu M, Zhang Y, Xia D, Chen Z, Wang L, Yin N, Wang Z. Puerarin attenuates the daunorubicin-induced apoptosis of H9c2 cells by activating the PI3K/Akt signaling pathway via the inhibition of Ca2+ influx. Int J Mol Med 2017; 40:1889-1894. [PMID: 29039532 DOI: 10.3892/ijmm.2017.3186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/20/2017] [Indexed: 11/06/2022] Open
Abstract
Puerarin extracted from Radix Puerariae is well known for its pharmacological effects, including antioxidant, anti‑inflammatory, neuroprotective and cardioprotective properties. In this study, we aimed to investigate the effects of puerarin on the daunorubicin (DNR)-induced apoptosis of H9c2 cells and to elucidate the potential mechanisms involved. MTT assay and flow cytometry were performed to evaluate cell cytotoxicity and apoptosis, respectively. Western blot analysis was used to assess changes in the expression levels of proteins, including caspase-3, Akt and phosphorylated Akt (p-Akt). Ratiometric imaging of intracellular calcium (Ca2+) using cells loaded with Fura-2 was also carried out. Our results revealed that puerarin pre-treatment protected the H9c2 cells against DNR-induced cytotoxicity by inhibiting cell apoptosis, which was also confirmed by the decrease in the expression of cleaved caspase-3. Additionally, p-Akt activation was associated with the suppressive effects of puerarin. Following pre-treatment with puerarin, the extracellular Ca2+ influx was restrained and this resulted in a reduction in the intracellular Ca2+ levels; these effects were abrogated by LY294002 [an inhibitor of phosphatidylinositol 3-kinase (PI3K)]. The inhibition of Ca2+ influx suggested that the PI3K/Akt signaling pathway participated in the suppressive effects of puerarin against H9c2 cell apoptosis. Taken togher, our findings demonstrate that puerarin attenuates the DNR-induced apoptosis of H9c2 cells by activating the PI3K/Akt signaling pathway via the inhibition of Ca2+ influx, suggesting that puerarin may be a potential cardioprotective agent for use in the clinical treatment of cardiomyopathy triggered by DNR.
Collapse
Affiliation(s)
- Weihua Li
- Department of Cardiology, Affiliated Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Min Lu
- Department of Human Anatomy and Embryology, Medical College of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Yanhong Zhang
- Department of Anatomy, College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, P.R. China
| | - Danqin Xia
- Department of Cardiology, Affiliated Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Zebin Chen
- Acupuncture and Moxibustion College, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, Hubei 430065, P.R. China
| | - Linhua Wang
- Department of Traditional Chinese Medicine, Hubei Rongjun Hospital, Wuhan, Hubei 430079, P.R. China
| | - Nina Yin
- Department of Anatomy, College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, P.R. China
| | - Zhigang Wang
- Department of Pathogen Biology, College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, P.R. China
| |
Collapse
|
22
|
de Almeida RMC, Clendenon SG, Richards WG, Boedigheimer M, Damore M, Rossetti S, Harris PC, Herbert BS, Xu WM, Wandinger-Ness A, Ward HH, Glazier JA, Bacallao RL. Transcriptome analysis reveals manifold mechanisms of cyst development in ADPKD. Hum Genomics 2016; 10:37. [PMID: 27871310 PMCID: PMC5117508 DOI: 10.1186/s40246-016-0095-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) causes progressive loss of renal function in adults as a consequence of the accumulation of cysts. ADPKD is the most common genetic cause of end-stage renal disease. Mutations in polycystin-1 occur in 87% of cases of ADPKD and mutations in polycystin-2 are found in 12% of ADPKD patients. The complexity of ADPKD has hampered efforts to identify the mechanisms underlying its pathogenesis. No current FDA (Federal Drug Administration)-approved therapies ameliorate ADPKD progression. Results We used the de Almeida laboratory’s sensitive new transcriptogram method for whole-genome gene expression data analysis to analyze microarray data from cell lines developed from cell isolates of normal kidney and of both non-cystic nephrons and cysts from the kidney of a patient with ADPKD. We compared results obtained using standard Ingenuity Volcano plot analysis, Gene Set Enrichment Analysis (GSEA) and transcriptogram analysis. Transcriptogram analysis confirmed the findings of Ingenuity, GSEA, and published analysis of ADPKD kidney data and also identified multiple new expression changes in KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways related to cell growth, cell death, genetic information processing, nucleotide metabolism, signal transduction, immune response, response to stimulus, cellular processes, ion homeostasis and transport and cofactors, vitamins, amino acids, energy, carbohydrates, drugs, lipids, and glycans. Transcriptogram analysis also provides significance metrics which allow us to prioritize further study of these pathways. Conclusions Transcriptogram analysis identifies novel pathways altered in ADPKD, providing new avenues to identify both ADPKD’s mechanisms of pathogenesis and pharmaceutical targets to ameliorate the progression of the disease. Electronic supplementary material The online version of this article (doi:10.1186/s40246-016-0095-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rita M C de Almeida
- Biocomplexity Institute and Department of Physics, Indiana University, Bloomington, IN, 47405, USA.,Instituto de Física and Instituto Nacional de Ciência e Tecnologia, Universidade Federal do Rio Grande do Sul, 91501-970, Porto Alegre, RS, Brazil
| | - Sherry G Clendenon
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | | | | | - Michael Damore
- AMGEN Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320-1799, USA
| | - Sandro Rossetti
- Division of Nephrology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Peter C Harris
- Division of Nephrology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Britney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Wei Min Xu
- Division of Nephrology, Department of Medicine, Richard Roudebush VAMC and Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Angela Wandinger-Ness
- Department of Pathology MSC08-4640 and Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Heather H Ward
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - James A Glazier
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Robert L Bacallao
- Division of Nephrology, Department of Medicine, Richard Roudebush VAMC and Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
23
|
Belmonte JM, Clendenon SG, Oliveira GM, Swat MH, Greene EV, Jeyaraman S, Glazier JA, Bacallao RL. Virtual-tissue computer simulations define the roles of cell adhesion and proliferation in the onset of kidney cystic disease. Mol Biol Cell 2016; 27:3673-3685. [PMID: 27193300 PMCID: PMC5221598 DOI: 10.1091/mbc.e16-01-0059] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022] Open
Abstract
Virtual-tissue modeling is used to model emergent cyst growth in polycystic kidney disease. Model predictions, confirmed experimentally, show that decreased cell adhesion is necessary to produce stalk cysts, and loss of contact inhibition causes saccular cysts. Virtual-tissue modeling can be used to fully explore cell- and tissue-based behaviors. In autosomal dominant polycystic kidney disease (ADPKD), cysts accumulate and progressively impair renal function. Mutations in PKD1 and PKD2 genes are causally linked to ADPKD, but how these mutations drive cell behaviors that underlie ADPKD pathogenesis is unknown. Human ADPKD cysts frequently express cadherin-8 (cad8), and expression of cad8 ectopically in vitro suffices to initiate cystogenesis. To explore cell behavioral mechanisms of cad8-driven cyst initiation, we developed a virtual-tissue computer model. Our simulations predicted that either reduced cell–cell adhesion or reduced contact inhibition of proliferation triggers cyst induction. To reproduce the full range of cyst morphologies observed in vivo, changes in both cell adhesion and proliferation are required. However, only loss-of-adhesion simulations produced morphologies matching in vitro cad8-induced cysts. Conversely, the saccular cysts described by others arise predominantly by decreased contact inhibition, that is, increased proliferation. In vitro experiments confirmed that cell–cell adhesion was reduced and proliferation was increased by ectopic cad8 expression. We conclude that adhesion loss due to cadherin type switching in ADPKD suffices to drive cystogenesis. Thus, control of cadherin type switching provides a new target for therapeutic intervention.
Collapse
Affiliation(s)
- Julio M Belmonte
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Sherry G Clendenon
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Guilherme M Oliveira
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Maciej H Swat
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Evan V Greene
- Division of Nephrology, Richard L. Roudebush VA Medical Center, and Indiana University School of Medicine, Indianapolis, IN 46202
| | - Srividhya Jeyaraman
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - James A Glazier
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Robert L Bacallao
- Division of Nephrology, Richard L. Roudebush VA Medical Center, and Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
24
|
De Rechter S, Decuypere JP, Ivanova E, van den Heuvel LP, De Smedt H, Levtchenko E, Mekahli D. Autophagy in renal diseases. Pediatr Nephrol 2016; 31:737-52. [PMID: 26141928 DOI: 10.1007/s00467-015-3134-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/14/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
Abstract
Autophagy is the cell biology process in which cytoplasmic components are degraded in lysosomes to maintain cellular homeostasis and energy production. In the healthy kidney, autophagy plays an important role in the homeostasis and viability of renal cells such as podocytes and tubular epithelial cells and of immune cells. Recently, evidence is mounting that (dys)regulation of autophagy is implicated in the pathogenesis of various renal diseases, and might be an attractive target for new renoprotective therapies. In this review, we provide an overview of the role of autophagy in kidney physiology and kidney diseases.
Collapse
Affiliation(s)
- Stéphanie De Rechter
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium. .,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium.
| | - Jean-Paul Decuypere
- Laboratory of Abdominal Transplantation, Department of Microbiology and Immunology Biomedical Sciences Group, KU Leuven, Leuven, Belgium.,Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | | | - Lambertus P van den Heuvel
- Laboratory of Paediatrics, KU Leuven, Leuven, Belgium.,Translational Metabolic Laboratory and Department of Paediatric Nephrology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Mangolini A, de Stephanis L, Aguiari G. Role of calcium in polycystic kidney disease: From signaling to pathology. World J Nephrol 2016; 5:76-83. [PMID: 26788466 PMCID: PMC4707171 DOI: 10.5527/wjn.v5.i1.76] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/21/2015] [Accepted: 12/11/2015] [Indexed: 02/06/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited monogenic kidney disease. Characterized by the development and growth of cysts that cause progressive kidney enlargement, it ultimately leads to end-stage renal disease. Approximately 85% of ADPKD cases are caused by mutations in the PKD1 gene, while mutations in the PKD2 gene account for the remaining 15% of cases. The PKD1 gene encodes for polycystin-1 (PC1), a large multi-functional membrane receptor protein able to regulate ion channel complexes, whereas polycystin-2 (PC2), encoded by the PKD2 gene, is an integral membrane protein that functions as a calcium-permeable cation channel, located mainly in the endoplasmic reticulum (ER). In the primary cilia of the epithelial cells, PC1 interacts with PC2 to form a polycystin complex that acts as a mechanosensor, regulating signaling pathways involved in the differentiation of kidney tubular epithelial cells. Despite progress in understanding the function of these proteins, the molecular mechanisms associated with the pathogenesis of ADPKD remain unclear. In this review we discuss how an imbalance between functional PC1 and PC2 proteins may disrupt calcium channel activities in the cilium, plasma membrane and ER, thereby altering intracellular calcium signaling and leading to the aberrant cell proliferation and apoptosis associated with the development and growth of renal cysts. Research in this field could lead to the discovery of new molecules able to rebalance intracellular calcium, thereby normalizing cell proliferation and reducing kidney cyst progression.
Collapse
|
26
|
Mamenko M, Zaika O, Boukelmoune N, O'Neil RG, Pochynyuk O. Deciphering physiological role of the mechanosensitive TRPV4 channel in the distal nephron. Am J Physiol Renal Physiol 2015; 308:F275-86. [PMID: 25503733 PMCID: PMC4329491 DOI: 10.1152/ajprenal.00485.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/08/2014] [Indexed: 12/14/2022] Open
Abstract
Long-standing experimental evidence suggests that epithelial cells in the renal tubule are able to sense osmotic and pressure gradients caused by alterations in ultrafiltrate flow by elevating intracellular Ca(2+) concentration. These responses are viewed as critical regulators of a variety of processes ranging from transport of water and solutes to cellular growth and differentiation. A loss in the ability to sense mechanical stimuli has been implicated in numerous pathologies associated with systemic imbalance of electrolytes and to the development of polycystic kidney disease. The molecular mechanisms conferring mechanosensitive properties to epithelial tubular cells involve activation of transient receptor potential (TRP) channels, such as TRPV4, allowing direct Ca(2+) influx to increase intracellular Ca(2+) concentration. In this review, we critically analyze the current evidence about signaling determinants of TRPV4 activation by luminal flow in the distal nephron and discuss how dysfunction of this mechanism contributes to the progression of polycystic kidney disease. We also review the physiological relevance of TRPV4-based mechanosensitivity in controlling flow-dependent K(+) secretion in the distal renal tubule.
Collapse
Affiliation(s)
- M Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - O Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - N Boukelmoune
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - R G O'Neil
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - O Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
27
|
Anoctamin 1 induces calcium-activated chloride secretion and proliferation of renal cyst-forming epithelial cells. Kidney Int 2013; 85:1058-67. [PMID: 24152967 DOI: 10.1038/ki.2013.418] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 08/20/2013] [Accepted: 08/22/2013] [Indexed: 01/08/2023]
Abstract
Polycystic kidney diseases are characterized by multiple bilateral renal cysts that gradually enlarge and lead to a decline in renal function. Cyst enlargement is driven by transepithelial chloride secretion, stimulated by enhanced levels of cyclic adenosine monophosphate, which activates apical cystic fibrosis transmembrane conductance regulator chloride channels. However, chloride secretion by calcium-dependent chloride channels, activated through stimulation of purinergic receptors, also has a major impact. To identify the molecular basis of calcium-dependent chloride secretion in cyst expansion, we determined the role of anoctamin 1 and 6, two recently discovered calcium-activated chloride channels both of which are expressed in epithelial cells. We found that anoctamin 1, which plays a role in epithelial fluid secretion and proliferation, is strongly expressed in principal-like MDCK cells (PLCs) forming cysts within a collagen matrix, in an embryonic kidney cyst model, and in human autosomal dominant polycystic kidney disease tissue. Knockdown of anoctamin 1 but not anoctamin 6 strongly diminished the calcium-dependent chloride secretion of PLCs. Moreover, two inhibitors of anoctamin ion channels, tannic acid and a more selective inhibitor of anoctamin 1, significantly inhibited PLC cyst growth and cyst enlargement in an embryonic kidney cyst model. Knockdown of ANO1 by morpholino analogs also attenuated embryonic cyst growth. Thus, calcium-activated chloride secretion by anoctamin 1 appears to be a crucial component of renal cyst growth.
Collapse
|