1
|
Lapin B, Vandensteen J, Gropplero G, Mazloum M, Bienaimé F, Descroix S, Coscoy S. Decoupling shear stress and pressure effects in the biomechanics of autosomal dominant polycystic kidney disease using a perfused kidney-on-chip. Acta Biomater 2025; 197:326-338. [PMID: 40089130 DOI: 10.1016/j.actbio.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Kidney tubular cells are submitted to two distinct mechanical forces generated by the urine flow: shear stress and hydrostatic pressure. In addition, the mechanical properties of the surrounding extracellular matrix modulate tubule deformation under constraints. These mechanical factors likely play a role in the pathophysiology of kidney as exemplified by autosomal dominant polycystic kidney disease, in which pressure, flow and matrix stiffness have been proposed to modulate the cystic dilation of tubules with PKD1 mutations. The lack of in vitro systems recapitulating the mechanical environment of kidney tubules impedes our ability to dissect the role of these mechanical factors. Here we describe a perfused kidney-on-chip with tunable extracellular matrix mechanical properties and hydrodynamic constraints, that allows a decoupling of shear stress and flow. We used this system to dissect how these mechanical cues affect Pkd1-/- tubule dilation. We investigated cell behavior for a flow shear stress of 1 dyn/cm², combined or not with a 10-mbar intraluminal pressure. Our results showed two distinct mechanisms leading to tubular dilation. For Pkd1-/- PCT cells (proximal tubule), overproliferation mechanically leads to tubular dilation of 1.5-2-fold in 5 days, regardless of the mechanical context. For mIMCD-3 cells (collecting duct), tube dilation was associated with a squamous cell morphology but not with overproliferation and was highly sensitive to extracellular matrix properties, with suppression of the dilation when switching extracellular matrix composition from 6 to 9 mg/ml collagen. Contrary to PCT, mIMCD-3 tube dilation was highly sensitive to the nature of hydrodynamic constraint. Surprisingly, flow alone suppressed Pkd1-/- mIMCD-3 tubule dilation observed in static conditions, while the addition of luminal pressure restored it. Our in vitro model emulating nephron geometrical and mechanical organization sheds light on the roles of mechanical constraints in ADPKD and demonstrates the importance of controlling intraluminal pressure in kidney tubule models. STATEMENT OF SIGNIFICANCE: In autosomal dominant polycystic kidney disease, the development of numerous renal cysts leads to renal failure, with no curative therapy available. The initial stage of cyst formation, local tubule dilation, remains poorly understood. Although mechanical cues may be decisive, there is a lack of biomimetic systems recapitulating them. Here, an innovative kidney-on-a-chip was designed to decouple different hydrodynamic cues. We observed disease-specific tube dilation, driven by distinct mechanisms based or not on proliferation, in proximal tubule or collecting duct cell lines. Strikingly in the latter case, dilation, highly dependent on mechanical conditions, was suppressed by flow but restored by luminal pressure. Our model highlights the role of mechanical constraints in ADPKD and the importance of pressure control in renal models.
Collapse
Affiliation(s)
- Brice Lapin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris 75005, France
| | - Jessica Vandensteen
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris 75005, France
| | - Giacomo Gropplero
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris 75005, France
| | - Manal Mazloum
- Université de Paris Cité, Institut Necker Enfants Malades-INEM, Département 'Croissance et Signalisation', INSERM UMR1151, CNRS UMR 8253, Paris, France
| | - Frank Bienaimé
- Université de Paris Cité, Institut Necker Enfants Malades-INEM, Département 'Croissance et Signalisation', INSERM UMR1151, CNRS UMR 8253, Paris, France; Service de Physiologie Hôpital Necker Enfants-Malades, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Stéphanie Descroix
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris 75005, France.
| | - Sylvie Coscoy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris 75005, France.
| |
Collapse
|
2
|
Smith AO, Frantz WT, Preval KM, Edwards YJK, Ceol CJ, Jonassen JA, Pazour GJ. The Tumor-Associated Calcium Signal Transducer 2 (TACSTD2) oncogene is upregulated in cystic epithelial cells revealing a potential new target for polycystic kidney disease. PLoS Genet 2024; 20:e1011510. [PMID: 39666736 DOI: 10.1371/journal.pgen.1011510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/26/2024] [Accepted: 11/23/2024] [Indexed: 12/14/2024] Open
Abstract
Polycystic kidney disease (PKD) is an important cause of kidney failure, but treatment options are limited. While later stages of the disease have been extensively studied, mechanisms driving the initial conversion of kidney tubules into cysts are not understood. To identify genes with the potential to promote cyst initiation, we deleted polycystin-2 (Pkd2) in mice and surveyed transcriptional changes before and immediately after cysts developed. We identified 74 genes which we term cyst initiation candidates (CICs). To identify conserved changes with relevance to human disease we compared these murine CICs to single cell transcriptomic data derived from patients with PKD and from healthy controls. Tumor-associated calcium signal transducer 2 (Tacstd2) stood out as an epithelial-expressed gene with elevated levels early in cystic transformation that further increased with disease progression. Human tissue biopsies and organoids show that TACSTD2 protein is low in normal kidney cells but is elevated in cyst lining cells, making it an excellent candidate for mechanistic exploration of its role in cyst initiation. While TACSTD2 has not been studied in PKD, it has been studied in cancer where it is highly expressed in solid tumors while showing minimal expression in normal tissue. This property is being exploited by antibody drug conjugates that target TACSTD2 for the delivery of cytotoxic drugs. Our finding that Tacstd2/TACSTD2 is prevalent in cysts, but not normal tissue, suggests that it should be explored as a candidate for drug development in PKD. More immediately, our work suggests that PKD patients undergoing TACSTD2-directed treatment for breast and urothelial cancer should be monitored for kidney effects.
Collapse
Affiliation(s)
- Abigail O Smith
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
- Morningside Graduate School of Biological Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - William Tyler Frantz
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
- Morningside Graduate School of Biological Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Kenley M Preval
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
- Morningside Graduate School of Biological Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Yvonne J K Edwards
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Craig J Ceol
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Julie A Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
3
|
Lapin B, Gropplero G, Vandensteen J, Mazloum M, Bienaimé F, Descroix S, Coscoy S. Decoupling shear stress and pressure effects in the biomechanics of autosomal dominant polycystic kidney disease using a perfused kidney-on-chip. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599137. [PMID: 38948811 PMCID: PMC11212944 DOI: 10.1101/2024.06.18.599137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Kidney tubular cells are submitted to two distinct mechanical forces generated by the urine flow: shear stress and hydrostatic pressure. In addition, the mechanical properties of the surrounding extracellular matrix modulate tubule deformation under constraints. These mechanical factors likely play a role in the pathophysiology of kidney diseases as exemplified by autosomal dominant polycystic kidney disease, in which pressure, flow and matrix stiffness have been proposed to modulate the cystic dilation of tubules with PKD1 mutations. The lack of in vitro systems recapitulating the mechanical environment of kidney tubules impedes our ability to dissect the role of these mechanical factors. Here we describe a perfused kidney-on-chip with tunable extracellular matrix mechanical properties and hydrodynamic constraints, that allows a decoupling of shear stress and flow. We used this system to dissect how these mechanical cues affect Pkd1 -/- tubule dilation. Our results show two distinct mechanisms leading to tubular dilation. For PCT cells (proximal tubule), overproliferation mechanically leads to tubular dilation, regardless of the mechanical context. For mIMCD-3 cells (collecting duct), tube dilation is associated with a squamous cell morphology but not with overproliferation and is highly sensitive to extracellular matrix properties and hydrodynamic constraints. Surprisingly, flow alone suppressed Pkd1 -/- mIMCD-3 tubule dilation observed in static conditions, while the addition of luminal pressure restored it. Our in vitro model emulating nephron geometrical and mechanical organization sheds light on the roles of mechanical constraints in ADPKD and demonstrates the importance of controlling intraluminal pressure in kidney tubule models.
Collapse
Affiliation(s)
- Brice Lapin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, 75005 Paris, France
| | - Giacomo Gropplero
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, 75005 Paris, France
| | - Jessica Vandensteen
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, 75005 Paris, France
| | - Manal Mazloum
- Université de Paris Cité, Institut Necker Enfants Malades-INEM, Département ‘Croissance et Signalisation’, INSERM UMR1151, CNRS UMR 8253 Paris, France
| | - Frank Bienaimé
- Université de Paris Cité, Institut Necker Enfants Malades-INEM, Département ‘Croissance et Signalisation’, INSERM UMR1151, CNRS UMR 8253 Paris, France
- Service de Physiologie Hôpital Necker Enfants-Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Stéphanie Descroix
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, 75005 Paris, France
| | - Sylvie Coscoy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, 75005 Paris, France
| |
Collapse
|
4
|
Tröndle K, Rizzo L, Pichler R, Zimmermann S, Lienkamp SS. Flow induces common and specific transcriptional changes in renal tubular epithelial cells involving the PI3K pathway. FASEB J 2024; 38:e23329. [PMID: 38050412 DOI: 10.1096/fj.202300834r] [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: 05/12/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023]
Abstract
Flow-induced shear stress affects renal epithelial cells in the nephron tubule with potential implications for differential functionalities of the individual segments. Disruptions of cellular mechanosensation or flow conditions are associated with the development and progression of various renal diseases. This study investigates the effects of flow on the transcriptome of various renal tubular epithelial cell types. We analyzed the transcriptome of induced renal epithelial cells (iREC) cultured under physiological flow (0.57 ± 0.05 dyn/cm2 ) or in static conditions for 72 h. RNA sequencing showed 861 differentially expressed genes (DEGs), with 503 up- and 358 downregulated under flow. DEGs were linked to extracellular matrix (ECM) components (e.g. Col1a1, Col4a3, Col4a4, Fn1, Smoc2), junctions (Gja1, Tubb5), channel activities (Abcc4, Aqp1), and transcription factors (Foxq1, Lgr6). Next, we performed a meta-analysis comparing our data with three published datasets that subjected epithelial cell lines from distinct segments to flow, including proximal tubule and collecting duct cells. We found that TGF-ß, p53, MAPK, and PI3K are common flow-regulated pathways. Tfrc expression and thus the capability of iron uptake is commonly upregulated under flow. Many DEGs were related to kidney diseases, such as fibrosis (e.g. Tgfb1-3 and Serpine1). To obtain further mechanistic insights we investigated the role of the PI3K pathway in flow sensing. Applying flow and inhibition of PI3K showed significantly altered expression of transcripts related to ECM remodeling, angiogenesis, and ion transport. This suggests that the PI3K pathway is a critical mediator in flow-dependent cellular processes and gene expression, potentially influencing renal development and tissue remodeling. Finally, we derived a cross-cell-line summary of common as well as segment-specific transcriptomic effects, thus providing insights into the molecular mechanisms underlying flow sensing in the nephron tubule.
Collapse
Affiliation(s)
- Kevin Tröndle
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Ludovica Rizzo
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Roman Pichler
- Department of Medicine IV, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, Department of Microsystems Engineering, IMTEK, University of Freiburg, Freiburg, Germany
| | - Soeren S Lienkamp
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Smith AO, Frantz WT, Preval KM, Edwards YJK, Ceol CJ, Jonassen JA, Pazour GJ. The Tumor-Associated Calcium Signal Transducer 2 (TACSTD2) oncogene is upregulated in pre-cystic epithelial cells revealing a new target for polycystic kidney disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23299387. [PMID: 38106222 PMCID: PMC10723484 DOI: 10.1101/2023.12.04.23299387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Polycystic kidney disease (PKD) is an important cause of end stage renal disease, but treatment options are limited. While later stages of the disease have been extensively studied, mechanisms driving the initial conversion of renal tubules into cysts are not understood. To identify factors that promote the initiation of cysts we deleted polycystin-2 ( Pkd2 ) in mice and surveyed transcriptional changes before and immediately after cysts developed. We identified 74 genes which we term cyst initiation candidates (CICs). To identify conserved changes with relevance to human disease we compared these murine CICs to single cell transcriptomic data derived from patients with PKD and from healthy controls. Tumor-associated calcium signal transducer 2 ( Tacstd2 ) stood out as an epithelial-expressed gene whose levels were elevated prior to cystic transformation and further increased with disease progression. Human tissue biopsies and organoids show that TACSTD2 protein is low in normal kidney cells but is elevated in cyst lining cells. While TACSTD2 has not been studied in PKD, it has been studied in cancer where it is highly expressed in solid tumors while showing minimal expression in normal tissue. This property is being exploited by antibody drug conjugates that target TACSTD2 for the delivery of cytotoxic drugs. Our finding that Tacstd2 is highly expressed in cysts, but not normal tissue, suggests that it should be explored as a candidate for drug development in PKD. More immediately, our work suggests that PKD patients undergoing TACSTD2 treatment for cancer should be monitored for kidney effects. One Sentence Summary The oncogene, tumor-associated calcium signal transducer 2 (Tacstd2) mRNA increased in abundance shortly after Pkd2 loss and may be a driver of cyst initiation in polycystic kidney disease.
Collapse
|
6
|
Garfa Traoré M, Roccio F, Miceli C, Ferri G, Parisot M, Cagnard N, Lhomme M, Dupont N, Benmerah A, Saunier S, Delous M. Fluid shear stress triggers cholesterol biosynthesis and uptake in inner medullary collecting duct cells, independently of nephrocystin-1 and nephrocystin-4. Front Mol Biosci 2023; 10:1254691. [PMID: 37916190 PMCID: PMC10616263 DOI: 10.3389/fmolb.2023.1254691] [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: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 11/03/2023] Open
Abstract
Renal epithelial cells are subjected to fluid shear stress of urine flow. Several cellular structures act as mechanosensors-the primary cilium, microvilli and cell adhesion complexes-that directly relay signals to the cytoskeleton to regulate various processes including cell differentiation and renal cell functions. Nephronophthisis (NPH) is an autosomal recessive tubulointerstitial nephropathy leading to end-stage kidney failure before adulthood. NPHP1 and NPHP4 are the major genes which code for proteins that form a complex at the transition zone of the primary cilium, a crucial region required for the maintenance of the ciliary composition integrity. These two proteins also interact with signaling components and proteins associated with the actin cytoskeleton at cell junctions. Due to their specific subcellular localization, we wondered whether NPHP1 and NPHP4 could ensure mechanosensory functions. Using a microfluidic set up, we showed that murine inner medullary collecting ductal cells invalidated for Nphp1 or Nphp4 are more responsive to immediate shear exposure with a fast calcium influx, and upon a prolonged shear condition, an inability to properly regulate cilium length and actin cytoskeleton remodeling. Following a transcriptomic study highlighting shear stress-induced gene expression changes, we showed that prolonged shear triggers both cholesterol biosynthesis pathway and uptake, processes that do not seem to involve neither NPHP1 nor NPHP4. To conclude, our study allowed us to determine a moderate role of NPHP1 and NPHP4 in flow sensation, and to highlight a new signaling pathway induced by shear stress, the cholesterol biosynthesis and uptake pathways, which would allow cells to cope with mechanical stress by strengthening their plasma membrane through the supply of cholesterol.
Collapse
Affiliation(s)
- Meriem Garfa Traoré
- Laboratory of Hereditary Kidney Disease, INSERM UMR 1163, Imagine Institute, Université Paris Cité, Paris, France
- Cell Imaging Platform, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université Paris Cité, Paris, France
| | - Federica Roccio
- Institut Necker Enfants-Malades (INEM), INSERM U1151/CNRS UMR 8253, Université Paris Cité, Paris, France
| | - Caterina Miceli
- Institut Necker Enfants-Malades (INEM), INSERM U1151/CNRS UMR 8253, Université Paris Cité, Paris, France
| | - Giulia Ferri
- Laboratory of Hereditary Kidney Disease, INSERM UMR 1163, Imagine Institute, Université Paris Cité, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Université Paris Cité, Paris, France
| | - Nicolas Cagnard
- Bioinformatic Platform, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Université Paris Cité, Paris, France
| | - Marie Lhomme
- ICAN Omics, IHU ICAN Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Nicolas Dupont
- Institut Necker Enfants-Malades (INEM), INSERM U1151/CNRS UMR 8253, Université Paris Cité, Paris, France
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Disease, INSERM UMR 1163, Imagine Institute, Université Paris Cité, Paris, France
| | - Sophie Saunier
- Laboratory of Hereditary Kidney Disease, INSERM UMR 1163, Imagine Institute, Université Paris Cité, Paris, France
| | - Marion Delous
- Laboratory of Hereditary Kidney Disease, INSERM UMR 1163, Imagine Institute, Université Paris Cité, Paris, France
| |
Collapse
|
7
|
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: 0.3] [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
|
8
|
Sun Y, Jin D, Zhang Z, Jin D, Xue J, Duan L, Zhang Y, Kang X, Lian F. The critical role of the Hippo signaling pathway in kidney diseases. Front Pharmacol 2022; 13:988175. [PMID: 36483738 PMCID: PMC9723352 DOI: 10.3389/fphar.2022.988175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/03/2022] [Indexed: 09/14/2023] Open
Abstract
The Hippo signaling pathway is involved in cell growth, proliferation, and apoptosis, and it plays a key role in regulating organ size, tissue regeneration, and tumor development. The Hippo signaling pathway also participates in the occurrence and development of various human diseases. Recently, many studies have shown that the Hippo pathway is closely related to renal diseases, including renal cancer, cystic kidney disease, diabetic nephropathy, and renal fibrosis, and it promotes the transformation of acute kidney disease to chronic kidney disease (CKD). The present paper summarizes and analyzes the research status of the Hippo signaling pathway in different kidney diseases, and it also summarizes the expression of Hippo signaling pathway components in pathological tissues of kidney diseases. In addition, the present paper discusses the positive therapeutic significance of traditional Chinese medicine (TCM) in regulating the Hippo signaling pathway for treating kidney diseases. This article introduces new targets and ideas for drug development, clinical diagnosis, and treatment of kidney diseases.
Collapse
Affiliation(s)
- Yuting Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Di Jin
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - JiaoJiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - LiYun Duan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - YuQing Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - XiaoMin Kang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - FengMei Lian
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
9
|
Tholen LE, Hoenderop JGJ, de Baaij JHF. Mechanisms of ion transport regulation by HNF1β in the kidney: beyond transcriptional regulation of channels and transporters. Pflugers Arch 2022; 474:901-916. [PMID: 35554666 PMCID: PMC9338905 DOI: 10.1007/s00424-022-02697-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 01/01/2023]
Abstract
Hepatocyte nuclear factor 1β (HNF1β) is a transcription factor essential for the development and function of the kidney. Mutations in and deletions of HNF1β cause autosomal dominant tubule interstitial kidney disease (ADTKD) subtype HNF1β, which is characterized by renal cysts, diabetes, genital tract malformations, and neurodevelopmental disorders. Electrolyte disturbances including hypomagnesemia, hyperuricemia, and hypocalciuria are common in patients with ADTKD-HNF1β. Traditionally, these electrolyte disturbances have been attributed to HNF1β-mediated transcriptional regulation of gene networks involved in ion transport in the distal part of the nephron including FXYD2, CASR, KCNJ16, and FXR. In this review, we propose additional mechanisms that may contribute to the electrolyte disturbances observed in ADTKD-HNF1β patients. Firstly, kidney development is severely affected in Hnf1b-deficient mice. HNF1β is required for nephron segmentation, and the absence of the transcription factor results in rudimentary nephrons lacking mature proximal tubule, loop of Henle, and distal convoluted tubule cluster. In addition, HNF1β is proposed to be important for apical-basolateral polarity and tight junction integrity in the kidney. Interestingly, cilia formation is unaffected by Hnf1b defects in several models, despite the HNF1β-mediated transcriptional regulation of many ciliary genes. To what extent impaired nephron segmentation, apical-basolateral polarity, and cilia function contribute to electrolyte disturbances in HNF1β patients remains elusive. Systematic phenotyping of Hnf1b mouse models and the development of patient-specific kidney organoid models will be essential to advance future HNF1β research.
Collapse
Affiliation(s)
- Lotte E Tholen
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands.
| |
Collapse
|
10
|
Hu C, Beebe K, Hernandez EJ, Lazaro-Guevara JM, Revelo MP, Huang Y, Maschek JA, Cox JE, Kohan DE. Multiomic identification of factors associated with progression to cystic kidney disease in mice with nephron Ift88 disruption. Am J Physiol Renal Physiol 2022; 322:F175-F192. [PMID: 34927449 PMCID: PMC8782669 DOI: 10.1152/ajprenal.00409.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 02/03/2023] Open
Abstract
Ift88 gene mutations cause primary cilia loss and polycystic kidney disease (PKD) in mice. Nephron intraflagellar transport protein 88 (Ift88) knockout (KO) at 2 mo postnatal does not affect renal histology at 4 mo postnatal and causes PKD only in males by 11 mo postnatal. To identify factors associated with PKD development, kidneys from 4-mo-old male and female control and Ift88 KO mice underwent transcriptomic, proteomic, Western blot, metabolomic, and lipidomic analyses. mRNAs involved in extracellular matrix (ECM) synthesis and degradation were selectively upregulated in male KO mice. Proteomic analysis was insufficiently sensitive to detect most ECM components, while Western blot analysis paradoxically revealed reduced fibronectin and collagen type I in male KO mice. Only male KO mice had upregulated mRNAs encoding fibrinogen subunits and receptors for vascular endothelial growth factor and platelet-derived growth factor; period 2, period 3, and nuclear receptor subfamily 1 group D member 1 clock mRNAs were selectively decreased in male KO mice. Proteomic, metabolomic, and lipidomic analyses detected a relative (vs. the same-sex control) decrease in factors involved in fatty acid β-oxidation in female KO mice, while increased or unchanged levels in male KO mice, including medium-chain acyl-CoA dehydrogenase, 3-hydroxybutyrate, and acylcarnitine. Three putative mRNA biomarkers of cystogenesis in male Ift88 KO mice (similar control levels between sexes and uniquely altered by KO in males) were identified, including high levels (fibrinogen α-chain and stromal cell-derived factor 2-like 1) and low levels (BTG3-associated nuclear protein) in male KO mice. These findings suggest that relative alterations in renal ECM metabolism, fatty acid β-oxidation, and other pathways precede cystogenesis in Ift88 KO mice. In addition, potential novel biomarkers of cystogenesis in Ift88 KO mice have been identified.NEW & NOTEWORTHY Male, but not female, mice with nephron intraflagellar transport protein 88 (Ift88) gene knockout (KO) develop polycystic kidneys by ∼1 yr postnatal. We performed multiomic analysis of precystic male and female Ift88 KO and control kidneys. Precystic male Ift88 KO mice exhibited differential alterations (vs. females) in mRNA, proteins, metabolites, and/or lipids associated with renal extracellular matrix metabolism, fatty acid β-oxidation, circadian rhythm, and other pathways. These findings suggest targets for evaluation in the pathogenesis of Ift88 KO polycystic kidneys.
Collapse
Affiliation(s)
- Chunyan Hu
- Division of Nephrology, University of Utah Health, Salt Lake City, Utah
| | - Katherine Beebe
- Molecular Medicine Program, University of Utah Health, Salt Lake City, Utah
| | - Edgar J Hernandez
- Department of Human Genetics, University of Utah Health, Salt Lake City, Utah
- Utah Center for Genetic Discovery, Salt Lake City, Utah
| | - Jose M Lazaro-Guevara
- Division of Nephrology, University of Utah Health, Salt Lake City, Utah
- Department of Human Genetics, University of Utah Health, Salt Lake City, Utah
| | - Monica P Revelo
- Deparment of Pathology, University of Utah Health, Salt Lake City, Utah
| | - Yufeng Huang
- Division of Nephrology, University of Utah Health, Salt Lake City, Utah
| | - J Alan Maschek
- Deparment of Pathology, University of Utah Health, Salt Lake City, Utah
| | - James E Cox
- Department of Biochemistry, University of Utah Health, Salt Lake City, Utah
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health, Salt Lake City, Utah
| |
Collapse
|
11
|
Herault S, Naser J, Carassiti D, Chooi KY, Nikolopoulou R, Font ML, Patel M, Pedrigi R, Krams R. Mechanosensitive pathways are regulated by mechanosensitive miRNA clusters in endothelial cells. Biophys Rev 2021; 13:787-796. [PMID: 34777618 PMCID: PMC8555030 DOI: 10.1007/s12551-021-00839-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Shear stress is known to affect many processes in (patho-) physiology through a complex, multi-molecular mechanism, termed mechanotransduction. The sheer complexity of the process has raised questions how mechanotransduction is regulated. Here, we comprehensively evaluate the literature about the role of small non-coding miRNA in the regulation of mechanotransduction. Regulation of mRNA by miRNA is rather complex, depending not only on the concentration of mRNA to miRNA, but also on the amount of mRNA competing for a single mRNA. The only mechanism to counteract the latter factor is through overarching structures of miRNA. Indeed, two overarching structures are present miRNA families and miRNA clusters, and both will be discussed in details, regarding the latest literature and a previous conducted study focussed on mechanotransduction. Both the literature and our own data support a new hypothesis that miRNA-clusters predominantly regulate mechanotransduction, affecting 65% of signalling pathways. In conclusion, a new and important mode of regulation of mechanotransduction is proposed, based on miRNA clusters. This finding implicates new avenues for treatment of mechanotransduction and atherosclerosis.
Collapse
Affiliation(s)
- Sean Herault
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Daniele Carassiti
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | - K. Yean Chooi
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Marti Llopart Font
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Ryan Pedrigi
- College of Engineering, Mechanical & Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Rob Krams
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| |
Collapse
|
12
|
Identification of pathological transcription in autosomal dominant polycystic kidney disease epithelia. Sci Rep 2021; 11:15139. [PMID: 34301992 PMCID: PMC8302622 DOI: 10.1038/s41598-021-94442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/08/2021] [Indexed: 11/09/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) affects more than 12 million people worldwide. Mutations in PKD1 and PKD2 cause cyst formation through unknown mechanisms. To unravel the pathogenic mechanisms in ADPKD, multiple studies have investigated transcriptional mis-regulation in cystic kidneys from patients and mouse models, and numerous dysregulated genes and pathways have been described. Yet, the concordance between studies has been rather limited. Furthermore, the cellular and genetic diversity in cystic kidneys has hampered the identification of mis-expressed genes in kidney epithelial cells with homozygous PKD mutations, which are critical to identify polycystin-dependent pathways. Here we performed transcriptomic analyses of Pkd1- and Pkd2-deficient mIMCD3 kidney epithelial cells followed by a meta-analysis to integrate all published ADPKD transcriptomic data sets. Based on the hypothesis that Pkd1 and Pkd2 operate in a common pathway, we first determined transcripts that are differentially regulated by both genes. RNA sequencing of genome-edited ADPKD kidney epithelial cells identified 178 genes that are concordantly regulated by Pkd1 and Pkd2. Subsequent integration of existing transcriptomic studies confirmed 31 previously described genes and identified 61 novel genes regulated by Pkd1 and Pkd2. Cluster analyses then linked Pkd1 and Pkd2 to mRNA splicing, specific factors of epithelial mesenchymal transition, post-translational protein modification and epithelial cell differentiation, including CD34, CDH2, CSF2RA, DLX5, HOXC9, PIK3R1, PLCB1 and TLR6. Taken together, this model-based integrative analysis of transcriptomic alterations in ADPKD annotated a conserved core transcriptomic profile and identified novel candidate genes for further experimental studies.
Collapse
|
13
|
Role of Shear Stress on Renal Proximal Tubular Cells for Nephrotoxicity Assays. J Toxicol 2021; 2021:6643324. [PMID: 33976696 PMCID: PMC8084667 DOI: 10.1155/2021/6643324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/24/2021] [Indexed: 12/21/2022] Open
Abstract
Drug-induced nephrotoxicity causes huge morbidity and mortality at massive financial cost. The greatest burden of drug-induced acute kidney injury falls on the proximal tubular cells. To maintain their structure and function, renal proximal tubular cells need the shear stress from tubular fluid flow. Diverse techniques to reintroduce shear stress have been studied in a variety of proximal tubular like cell culture models. These studies often have limited replicates because of the huge cost of equipment and do not report all relevant parameters to allow reproduction and comparison of studies between labs. This review codifies the techniques used to reintroduce shear stress, the cell lines utilized, and the biological outcomes reported. Further, we propose a set of interventions to enhance future cell biology understanding of nephrotoxicity using cell culture models.
Collapse
|
14
|
Duong Phu M, Bross S, Burkhalter MD, Philipp M. Limitations and opportunities in the pharmacotherapy of ciliopathies. Pharmacol Ther 2021; 225:107841. [PMID: 33771583 DOI: 10.1016/j.pharmthera.2021.107841] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023]
Abstract
Ciliopathies are a family of rather diverse conditions, which have been grouped based on the finding of altered or dysfunctional cilia, potentially motile, small cellular antennae extending from the surface of postmitotic cells. Cilia-related disorders include embryonically arising conditions such as Joubert, Usher or Kartagener syndrome, but also afflictions with a postnatal or even adult onset phenotype, i.e. autosomal dominant polycystic kidney disease. The majority of ciliopathies are syndromic rather than affecting only a single organ due to cilia being found on almost any cell in the human body. Overall ciliopathies are considered rare diseases. Despite that, pharmacological research and the strive to help these patients has led to enormous therapeutic advances in the last decade. In this review we discuss new treatment options for certain ciliopathies, give an outlook on promising future therapeutic strategies, but also highlight the limitations in the development of therapeutic approaches of ciliopathies.
Collapse
Affiliation(s)
- Max Duong Phu
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Stefan Bross
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany.
| |
Collapse
|
15
|
Pathway identification through transcriptome analysis. Cell Signal 2020; 74:109701. [PMID: 32649993 DOI: 10.1016/j.cellsig.2020.109701] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Systems-based, agnostic approaches focusing on transcriptomics data have been employed to understand the pathogenesis of polycystic kidney diseases (PKD). While multiple signaling pathways, including Wnt, mTOR and G-protein-coupled receptors, have been implicated in late stages of disease, there were few insights into the transcriptional cascade immediately downstream of Pkd1 inactivation. One of the consistent findings has been transcriptional evidence of dysregulated metabolic and cytoskeleton remodeling pathways. Recent technical developments, including bulk and single-cell RNA sequencing technologies and spatial transcriptomics, offer new angles to investigate PKD. In this article, we review what has been learned based on transcriptional approaches and consider future opportunities.
Collapse
|
16
|
Verschuren EHJ, Rigalli JP, Castenmiller C, Rohrbach MU, Bindels RJM, Peters DJM, Arjona FJ, Hoenderop JGJ. Pannexin-1 mediates fluid shear stress-sensitive purinergic signaling and cyst growth in polycystic kidney disease. FASEB J 2020; 34:6382-6398. [PMID: 32159259 DOI: 10.1096/fj.201902901r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/07/2020] [Accepted: 03/01/2020] [Indexed: 12/16/2022]
Abstract
Tubular ATP release is regulated by mechanosensation of fluid shear stress (FSS). Polycystin-1/polycystin-2 (PC1/PC2) functions as a mechanosensory complex in the kidney. Extracellular ATP is implicated in polycystic kidney disease (PKD), where PC1/PC2 is dysfunctional. This study aims to provide new insights into the ATP signaling under physiological conditions and PKD. Microfluidics, pharmacologic inhibition, and loss-of-function approaches were combined to assess the ATP release in mouse distal convoluted tubule 15 (mDCT15) cells. Kidney-specific Pkd1 knockout mice (iKsp-Pkd1-/- ) and zebrafish pkd2 morphants (pkd2-MO) were as models for PKD. FSS-exposed mDCT15 cells displayed increased ATP release. Pannexin-1 inhibition and knockout decreased FSS-modulated ATP release. In iKsp-Pkd1-/- mice, elevated renal pannexin-1 mRNA expression and urinary ATP were observed. In Pkd1-/- mDCT15 cells, elevated ATP release was observed upon the FSS mechanosensation. In these cells, increased pannexin-1 mRNA expression was observed. Importantly, pannexin-1 inhibition in pkd2-MO decreased the renal cyst growth. Our results demonstrate that pannexin-1 channels mediate ATP release into the tubular lumen due to pro-urinary flow. We present pannexin-1 as novel therapeutic target to prevent the renal cyst growth in PKD.
Collapse
Affiliation(s)
- Eric H J Verschuren
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Juan P Rigalli
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Charlotte Castenmiller
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Meike U Rohrbach
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Francisco J Arjona
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
17
|
Verschuren EHJ, Castenmiller C, Peters DJM, Arjona FJ, Bindels RJM, Hoenderop JGJ. Sensing of tubular flow and renal electrolyte transport. Nat Rev Nephrol 2020; 16:337-351. [DOI: 10.1038/s41581-020-0259-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
|
18
|
Identification of ADPKD-Related Genes and Pathways in Cells Overexpressing PKD2. Genes (Basel) 2020; 11:genes11020122. [PMID: 31979107 PMCID: PMC7074416 DOI: 10.3390/genes11020122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 11/17/2022] Open
Abstract
Consistent with the gene dosage effect hypothesis, renal cysts can arise in transgenic murine models overexpressing either PKD1 or PKD2, which are causal genes for autosomal dominant polycystic kidney disease (ADPKD). To determine whether PKD gene overexpression is a universal mechanism driving cystogenesis or is merely restricted to rodents, other animal models are required. Previously, we failed to observe any renal cysts in a transgenic porcine model of PKD2 overexpression partially due to epigenetic silencing of the transgene. Thus, to explore the feasibility of porcine models and identify potential genes/pathways affected in ADPKD, LLC-PK1 cells with high PKD2 expression were generated. mRNA sequencing (RNA-seq) was performed, and MYC, IER3, and ADM were found to be upregulated genes common to the different PKD2 overexpression cell models. MYC is a well-characterized factor contributing to cystogenesis, and ADM is a biomarker for chronic kidney disease. Thus, these genes might be indicators of disease progression. Additionally, some ADPKD-associated pathways, e.g., the mitogen-activated protein kinase (MAPK) pathway, were enriched in the cells. Moreover, gene ontology (GO) analysis demonstrated that proliferation, apoptosis, and cell cycle regulation, which are hallmarks of ADPKD, were altered. Therefore, our experiment identified some biomarkers or indicators of ADPKD, indicating that high PKD2 expression would likely drive cystogenesis in future porcine models.
Collapse
|
19
|
Lee H, Kang J, Ahn S, Lee J. The Hippo Pathway Is Essential for Maintenance of Apicobasal Polarity in the Growing Intestine of Caenorhabditis elegans. Genetics 2019; 213:501-515. [PMID: 31358532 PMCID: PMC6781910 DOI: 10.1534/genetics.119.302477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022] Open
Abstract
Although multiple determinants for establishing polarity in membranes of epithelial cells have been identified, the mechanism for maintaining apicobasal polarity is not fully understood. Here, we show that the conserved Hippo kinase pathway plays a role in the maintenance of apicobasal polarity in the developing intestine of Caenorhabditis elegans We screened suppressors of the mutation in wts-1-the gene that encodes the LATS kinase homolog, deficiency of which leads to disturbance of the apicobasal polarity of the intestinal cells and to eventual death of the organism. We identified several alleles of yap-1 and egl-44 that suppress the effects of this mutation. yap-1 encodes a homolog of YAP/Yki, and egl-44 encodes a homolog of TEAD/Sd. WTS-1 bound directly to YAP-1 and inhibited its nuclear accumulation in intestinal cells. We also found that NFM-1, which is a homolog of NF2/Merlin, functioned in the same genetic pathway as WTS-1 to regulate YAP-1 to maintain cellular polarity. Transcriptome analysis identified several target candidates of the YAP-1-EGL-44 complex including TAT-2, which encodes a putative P-type ATPase. In summary, we have delineated the conserved Hippo pathway in C. elegans consisting of NFM-1-WTS-1-YAP-1-EGL-44 and proved that the proper regulation of YAP-1 by upstream NFM-1 and WTS-1 is essential for maintenance of apicobasal membrane identities of the growing intestine.
Collapse
Affiliation(s)
- Hanee Lee
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Junsu Kang
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Soungyub Ahn
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Junho Lee
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| |
Collapse
|
20
|
Vriend J, Peters JGP, Nieskens TTG, Škovroňová R, Blaimschein N, Schmidts M, Roepman R, Schirris TJJ, Russel FGM, Masereeuw R, Wilmer MJ. Flow stimulates drug transport in a human kidney proximal tubule-on-a-chip independent of primary cilia. Biochim Biophys Acta Gen Subj 2019; 1864:129433. [PMID: 31520681 DOI: 10.1016/j.bbagen.2019.129433] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Kidney disease modeling and assessment of drug-induced kidney injury can be advanced using three-dimensional (3D) microfluidic models that recapitulate in vivo characteristics. Fluid shear stress (FSS) has been depicted as main modulator improving in vitro physiology in proximal tubule epithelial cells (PTECs). We aimed to elucidate the role of FSS and primary cilia on transport activity and morphology in PTECs. METHODS Human conditionally immortalized PTEC (ciPTEC-parent) was cultured in a microfluidic 3D device, the OrganoPlate, under a physiological peak FSS of 2.0 dyne/cm2 or low peak FSS of 0.5 dyne/cm2. Upon a 9-day exposure to FSS, albumin-FITC uptake, activity of P-glycoprotein (P-gp) and multidrug resistance-associated proteins 2/4 (MRP2/4), cytotoxicity and cell morphology were determined. RESULTS A primary cilium knock-out cell model, ciPTEC-KIF3α-/-, was successfully established via CRISPR-Cas9 genome editing. Under physiological peak FSS, albumin-FITC uptake (p = .04) and P-gp efflux (p = .002) were increased as compared to low FSS. Remarkably, a higher albumin-FITC uptake (p = .03) and similar trends in activity of P-gp and MRP2/4 were observed in ciPTEC-KIF3α-/-. FSS induced cell elongation corresponding with the direction of flow in both cell models, but had no effect on cyclosporine A-induced cytotoxicity. CONCLUSIONS FSS increased albumin uptake, P-gp efflux and cell elongation, but this was not attributed to a mechanosensitive mechanism related to primary cilia in PTECs, but likely to microvilli present at the apical membrane. GENERAL SIGNIFICANCE FSS-induced improvements in biological characteristics and activity in PTECs was not mediated through a primary cilium-related mechanism.
Collapse
Affiliation(s)
- Jelle Vriend
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Janny G P Peters
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Renata Škovroňová
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nina Blaimschein
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Miriam Schmidts
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany
| | - Ronald Roepman
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|