1
|
Richfield O, Cortez R, Kulthinee S, Franco M, Navar LG. Purinergic Receptor Activation Protects Glomerular Microvasculature from Increased Mechanical Stress in Angiotensin II-Induced Hypertension: A Modeling Study. Int J Mol Sci 2025; 26:1928. [PMID: 40076554 PMCID: PMC11900630 DOI: 10.3390/ijms26051928] [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: 12/31/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Angiotensin II (Ang II)-induced hypertension increases afferent (AA) and efferent (EA) arteriole resistances via the actions of Ang II on the AT1 receptor. In addition to the increased interstitial levels of Ang II, the increased arterial pressure increases interstitial ATP concentrations. In turn, ATP acts on the purinergic receptors P2X1 and P2X7 to constrict the AA, preventing increases in plasma flow and single-nephron GFR (SNGFR). While the hemodynamic effects of P2 activation have been characterized, the resulting increases in mechanical stresses (shear stress and circumferential hoop stress) on the glomerular microvasculature have not been quantified. A mathematical microvascular hemodynamic glomerular model was developed to simulate blood flow and plasma filtration in an anatomically accurate rat glomerular capillary network. AA and EA resistances were adjusted to match glomerular hemodynamic data for control, Ang II-induced hypertension, and P2X1-blocked conditions. A blockade of the purinergic receptors reduced both afferent and efferent resistances, maintaining glomerular pressure at hypertensive levels but increasing blood flow and sheer stress significantly. Because glomerular pressure was maintained, hoop stress barely changed. Our results indicate that the activation of the purinergic system protects the glomerular microvasculature from elevated shear stress caused by increased blood flow that would occur in the absence of purinergic stimulation.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation IGERT PhD Program, Tulane University, New Orleans, LA 70118, USA
- Department of Physiology and the Tulane Hypertension and Renal Center of Excellence, Tulane University, New Orleans, LA 70112, USA
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA 70118, USA;
| | - Supaporn Kulthinee
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Martha Franco
- Renal Pathophysiology Laboratory, Department of Nephrology, Instituto Nacional de CardiologÍa “Ignacio Chávez”, México City 14080, Mexico;
| | - L. Gabriel Navar
- Department of Physiology and the Tulane Hypertension and Renal Center of Excellence, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
2
|
Briggs JP, Knepper MA. Shear Forces and the Vulnerability of the Podocyte. J Am Soc Nephrol 2025; 36:169-170. [PMID: 39729364 PMCID: PMC11801737 DOI: 10.1681/asn.0000000590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024] Open
Affiliation(s)
| | - Mark A. Knepper
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
3
|
Fuhrmann A, Pritz B, Endlich K, Kriz W. Numerical Flow Simulations of the Shear Stress Forces Arising in Filtration Slits during Glomerular Filtration in Rat Kidney. J Am Soc Nephrol 2025; 36:219-230. [PMID: 39348197 PMCID: PMC11801757 DOI: 10.1681/asn.0000000513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/25/2024] [Indexed: 10/02/2024] Open
Abstract
Key Points Computational fluid dynamics were applied to estimate the shear stress challenge to the filtration barrier during glomerular filtration in rats. Shear forces were especially relevant in pathologic situations where they contribute to the loss of viable podocytes. Background The flow dynamic forces during glomerular filtration challenging the fixation of podocytes to the glomerular basement membrane (GBM) are insufficiently understood. Methods Numerical flow simulations were used to estimate these forces in the rat kidney. Simulations were run with a three-dimensional (3D) model of the slit diaphragm as a zipper structure according to Rodewald and Karnovsky. The GBM was modeled as a porous medium. Results Filtrate flow exerted a mean wall shear stress of 39 Pa with a maximum of 152 Pa on the plasma membrane of foot processes and up to 250 Pa on internal surfaces of the slit diaphragm. The slit diaphragm accounted for 25% of the hydrodynamic resistance of the glomerular filtration barrier. Based on the results of the 3D model, we developed a two-dimensional (2D) model that allowed us to perform extensive parameter variations. Reducing the filtration slit width from 40 to 30 nm almost doubled wall shear stress. Furthermore, increasing filtrate flow velocity by 50% increased wall shear stress by 47%. When increasing the viscous resistance of the slit diaphragm, the pressure drop across the slit diaphragm increased to intolerably high values. A lower viscous resistance of the slit diaphragm than that of the GBM accounted for a gradual pressure decline along the filtration barrier. The subpodocyte space tempered these challenges in circumscribed areas of filtration surface but had only a marginal impact on overall forces. Conclusions The filtration barrier experiences high levels of shear and pressure stress accounting for the detachment of injured but viable podocytes from the GBM—a hallmark in many glomerular diseases.
Collapse
Affiliation(s)
- Alexander Fuhrmann
- Institute of Thermal Turbomachinery, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Balazs Pritz
- Institute of Thermal Turbomachinery, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Wilhelm Kriz
- Department of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Zhang Y, Arzaghi H, Ma Z, Roye Y, Musah S. Epigenetics of Hypertensive Nephropathy. Biomedicines 2024; 12:2622. [PMID: 39595187 PMCID: PMC11591919 DOI: 10.3390/biomedicines12112622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertensive nephropathy (HN) is a leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD), contributing to significant morbidity, mortality, and rising healthcare costs. In this review article, we explore the role of epigenetic mechanisms in HN progression and their potential therapeutic implications. We begin by examining key epigenetic modifications-DNA methylation, histone modifications, and non-coding RNAs-observed in kidney disease. Next, we discuss the underlying pathophysiology of HN and highlight current in vitro and in vivo models used to study the condition. Finally, we compare various types of HN-induced renal injury and their associated epigenetic mechanisms with those observed in other kidney injury models, drawing inferences on potential epigenetic therapies for HN. The information gathered in this work indicate that epigenetic mechanisms can drive the progression of HN by regulating key molecular signaling pathways involved in renal damage and fibrosis. The limitations of Renin-Angiotensin-Aldosterone System (RAAS) inhibitors underscore the need for alternative treatments targeting epigenetic pathways. This review emphasizes the importance of further research into the epigenetic regulation of HN to develop more effective therapies and preventive strategies. Identifying novel epigenetic markers could provide new therapeutic opportunities for managing CKD and reducing the burden of ESRD.
Collapse
Affiliation(s)
- Yize Zhang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hamidreza Arzaghi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Zhehan Ma
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27708, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, and Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
5
|
Ma R, Tao Y, Wade ML, Mallet RT. Non-voltage-gated Ca 2+ channel signaling in glomerular cells in kidney health and disease. Am J Physiol Renal Physiol 2024; 327:F249-F264. [PMID: 38867675 PMCID: PMC11460346 DOI: 10.1152/ajprenal.00130.2024] [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: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Positioned at the head of the nephron, the renal corpuscle generates a plasma ultrafiltrate to initiate urine formation. Three major cell types within the renal corpuscle, the glomerular mesangial cells, podocytes, and glomerular capillary endothelial cells, communicate via endocrine- and paracrine-signaling mechanisms to maintain the structure and function of the glomerular capillary network and filtration barrier. Ca2+ signaling mediated by several distinct plasma membrane Ca2+ channels impacts the functions of all three cell types. The past two decades have witnessed pivotal advances in understanding of non-voltage-gated Ca2+ channel function and regulation in the renal corpuscle in health and renal disease. This review summarizes the current knowledge of the physiological and pathological impact of non-voltage-gated Ca2+ channel signaling in mesangial cells, podocytes and glomerular capillary endothelium. The main focus is on transient receptor potential and store-operated Ca2+ channels, but ionotropic N-methyl-d-aspartate receptors and purinergic receptors also are discussed. This update of Ca2+ channel functions and their cellular signaling cascades in the renal corpuscle is intended to inform the development of therapeutic strategies targeting these channels to treat kidney diseases, particularly diabetic nephropathy.
Collapse
Affiliation(s)
- Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Michael L Wade
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
6
|
Pajenda S, Gerges D, Wagner L, O’Connell D, Aiad M, Imre R, Mechtler K, Zimprich A, Schmidt A, Sengoelge G, Winnicki W. Identification of Pathogenic Pathways for Recurrence of Focal Segmental Glomerulosclerosis after Kidney Transplantation. Diagnostics (Basel) 2024; 14:1591. [PMID: 39125467 PMCID: PMC11312181 DOI: 10.3390/diagnostics14151591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Primary focal segmental glomerulosclerosis (FSGS) is a disease of the podocytes and glomerulus, leading to nephrotic syndrome and progressive loss of renal function. One of the most serious aspects is its recurrence of disease in over 30% of patients following allogeneic kidney transplantation, leading to early graft loss. This research investigates the individual genetic predispositions and differences in the immune responses leading to recurrence of FSGS after transplantation. We performed exome sequencing on six patients with recurrent FSGS to identify variants in fifty-one genes and found significant variations in the alpha-2-macroglobulin (A2M). Immunoblotting was used to investigate effects of specific gene variants at the protein level. Further expression analysis identified A2M, exophilin 5 (EXPH5) and plectin (PLEC) as specific proteins linked to podocytes, endothelial cells, and the glomerulus. Subsequent protein array screening revealed the presence of non-HLA-specific antibodies, including TRIM21, after transplantation. Using Metascape for pathway and process enrichment analysis, we focused on the IL-17 signaling and chemotaxis pathways. ELISA measurements showed significantly elevated IL-17 levels in patients with recurrent FSGS (32.30 ± 9.12 pg/mL) compared to individuals with other glomerular diseases (23.16 ± 2.49 pg/mL; p < 0.01) and healthy subjects (22.28 ± 0.94 pg/mL; p < 0.01), with no significant difference in plasma CCL2/MCP-1 levels between groups. This study explores the molecular dynamics underlying recurrence of FSGS after transplantation, offering insights into potential biomarkers and therapeutic targets for the future development of individualized treatments for transplant patients.
Collapse
Affiliation(s)
- Sahra Pajenda
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - Daniela Gerges
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - Ludwig Wagner
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - David O’Connell
- BiOrbic—Bioeconomy Research Centre, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Monika Aiad
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - Richard Imre
- ProtChem Facility, IMP-IMBA, Research Institute of Molecular Pathology—Institute of Molecular Biotechnology, 1030 Vienna, Austria; (R.I.); (K.M.)
| | - Karl Mechtler
- ProtChem Facility, IMP-IMBA, Research Institute of Molecular Pathology—Institute of Molecular Biotechnology, 1030 Vienna, Austria; (R.I.); (K.M.)
| | - Alexander Zimprich
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Alice Schmidt
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - Guerkan Sengoelge
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| | - Wolfgang Winnicki
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (D.G.); (M.A.); (A.S.); (G.S.); (W.W.)
| |
Collapse
|
7
|
Kumar D, Nadda R, Repaka R. Advances and challenges in organ-on-chip technology: toward mimicking human physiology and disease in vitro. Med Biol Eng Comput 2024; 62:1925-1957. [PMID: 38436835 DOI: 10.1007/s11517-024-03062-7] [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: 10/19/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Organs-on-chips have been tissues or three-dimensional (3D) mini-organs that comprise numerous cell types and have been produced on microfluidic chips to imitate the complicated structures and interactions of diverse cell types and organs under controlled circumstances. Several morphological and physiological distinctions exist between traditional 2D cultures, animal models, and the growing popular 3D cultures. On the other hand, animal models might not accurately simulate human toxicity because of physiological variations and interspecies metabolic capability. The on-chip technique allows for observing and understanding the process and alterations occurring in metastases. The present study aimed to briefly overview single and multi-organ-on-chip techniques. The current study addresses each platform's essential benefits and characteristics and highlights recent developments in developing and utilizing technologies for single and multi-organs-on-chips. The study also discusses the drawbacks and constraints associated with these models, which include the requirement for standardized procedures and the difficulties of adding immune cells and other intricate biological elements. Finally, a comprehensive review demonstrated that the organs-on-chips approach has a potential way of investigating organ function and disease. The advancements in single and multi-organ-on-chip structures can potentially increase drug discovery and minimize dependency on animal models, resulting in improved therapies for human diseases.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| | - Rahul Nadda
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India.
| | - Ramjee Repaka
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| |
Collapse
|
8
|
Richfield O, Cortez R, Navar LG. Modeling the interaction between tubuloglomerular feedback and myogenic mechanisms in the control of glomerular mechanics. Front Physiol 2024; 15:1410764. [PMID: 38966231 PMCID: PMC11223525 DOI: 10.3389/fphys.2024.1410764] [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: 04/01/2024] [Accepted: 05/24/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction: Mechanical stresses and strains exerted on the glomerular cells have emerged as potentially influential factors in the progression of glomerular disease. Renal autoregulation, the feedback process by which the afferent arteriole changes in diameter in response to changes in blood pressure, is assumed to control glomerular mechanical stresses exerted on the glomerular capillaries. However, it is unclear how the two major mechanisms of renal autoregulation, the afferent arteriole myogenic mechanism and tubuloglomerular feedback (TGF), each contribute to the maintenance of glomerular mechanical homeostasis. Methods: In this study, we made a mathematical model of renal autoregulation and combined this model with an anatomically accurate model of glomerular blood flow and filtration, developed previously by us. We parameterized the renal autoregulation model based on data from previous literature, and we found evidence for an increased myogenic mechanism sensitivity when TGF is operant, as has been reported previously. We examined the mechanical effects of each autoregulatory mechanism (the myogenic, TGF and modified myogenic) by simulating blood flow through the glomerular capillary network with and without each mechanism operant. Results: Our model results indicate that the myogenic mechanism plays a central role in maintaining glomerular mechanical homeostasis, by providing the most protection to the glomerular capillaries. However, at higher perfusion pressures, the modulation of the myogenic mechanism sensitivity by TGF is crucial for the maintenance of glomerular mechanical homeostasis. Overall, a loss of renal autoregulation increases mechanical strain by up to twofold in the capillaries branching off the afferent arteriole. This further corroborates our previous simulation studies, that have identified glomerular capillaries nearest to the afferent arteriole as the most prone to mechanical injury in cases of disturbed glomerular hemodynamics. Discussion: Renal autoregulation is a complex process by which multiple feedback mechanisms interact to control blood flow and filtration in the glomerulus. Importantly, our study indicates that another function of renal autoregulation is control of the mechanical stresses on the glomerular cells, which indicates that loss or inhibition of renal autoregulation may have a mechanical effect that may contribute to glomerular injury in diseases such as hypertension or diabetes. This study highlights the utility of mathematical models in integrating data from previous experimental studies, estimating variables that are difficult to measure experimentally (i.e. mechanical stresses in microvascular networks) and testing hypotheses that are historically difficult or impossible to measure.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation PhD Program, Tulane University, New Orleans, LA, United States
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA, United States
| | - L. Gabriel Navar
- Department of Physiology, Tulane School of Medicine, New Orleans, LA, United States
| |
Collapse
|
9
|
Nishimura Y. Revolutionizing renal research: The future of kidney-on-a-chip in biotechnology. Regen Ther 2024; 26:275-280. [PMID: 38993536 PMCID: PMC11237358 DOI: 10.1016/j.reth.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 07/13/2024] Open
Abstract
In vitro models of kidneys have limited effectiveness owing to the complex structure and functions of the kidney when compared with other organs. Therefore many renal function evaluations are currently being carried out through animal experiments. In contrast, efforts are being made to apply biomimetic systems, such as organ-on-a-chip, which is based on microfluidic device technology, to serve as an in vitro model for the kidney. These systems aimed to recreate a physiological cultivation environment. This review has provided an overview of organ-on-a-chip research focused on glomeruli and tubules as in vitro models for the kidney and discusses future prospects.
Collapse
Affiliation(s)
- Yusuke Nishimura
- Department of Clinical Engineering, Faculty of Medical Science and Technology, Gunma Paz University, 3-3-4 Tonyamachi, Takasaki-shi, Gunma, 370-0006, Japan
| |
Collapse
|
10
|
Kliewe F, Siegerist F, Hammer E, Al-Hasani J, Amling TRJ, Hollemann JZE, Schindler M, Drenic V, Simm S, Amann K, Daniel C, Lindenmeyer M, Hecker M, Völker U, Endlich N. Zyxin is important for the stability and function of podocytes, especially during mechanical stretch. Commun Biol 2024; 7:446. [PMID: 38605154 PMCID: PMC11009394 DOI: 10.1038/s42003-024-06125-5] [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: 07/18/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Podocyte detachment due to mechanical stress is a common issue in hypertension-induced kidney disease. This study highlights the role of zyxin for podocyte stability and function. We have found that zyxin is significantly up-regulated in podocytes after mechanical stretch and relocalizes from focal adhesions to actin filaments. In zyxin knockout podocytes, we found that the loss of zyxin reduced the expression of vinculin and VASP as well as the expression of matrix proteins, such as fibronectin. This suggests that zyxin is a central player in the translation of mechanical forces in podocytes. In vivo, zyxin is highly up-regulated in patients suffering from diabetic nephropathy and in hypertensive DOCA-salt treated mice. Furthermore, zyxin loss in mice resulted in proteinuria and effacement of podocyte foot processes that was measured by super resolution microscopy. This highlights the essential role of zyxin for podocyte maintenance in vitro and in vivo, especially under mechanical stretch.
Collapse
Affiliation(s)
- Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jaafar Al-Hasani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | | | | | - Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Vedran Drenic
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| |
Collapse
|
11
|
Mascharak S, Guo JL, Griffin M, Berry CE, Wan DC, Longaker MT. Modelling and targeting mechanical forces in organ fibrosis. NATURE REVIEWS BIOENGINEERING 2024; 2:305-323. [PMID: 39552705 PMCID: PMC11567675 DOI: 10.1038/s44222-023-00144-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 11/19/2024]
Abstract
Few efficacious therapies exist for the treatment of fibrotic diseases, such as skin scarring, liver cirrhosis and pulmonary fibrosis, which is related to our limited understanding of the fundamental causes and mechanisms of fibrosis. Mechanical forces from cell-matrix interactions, cell-cell contact, fluid flow and other physical stimuli may play a central role in the initiation and propagation of fibrosis. In this Review, we highlight the mechanotransduction mechanisms by which various sources of physical force drive fibrotic disease processes, with an emphasis on central pathways that may be therapeutically targeted to prevent and reverse fibrosis. We then discuss engineered models of mechanotransduction in fibrosis, as well as molecular and biomaterials-based therapeutic approaches for limiting fibrosis and promoting regenerative healing phenotypes in various organs. Finally, we discuss challenges within fibrosis research that remain to be addressed and that may greatly benefit from next-generation bioengineered model systems.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Jason L. Guo
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Charlotte E. Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T. Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
12
|
Schindler M, Endlich N. Zebrafish as a model for podocyte research. Am J Physiol Renal Physiol 2024; 326:F369-F381. [PMID: 38205541 DOI: 10.1152/ajprenal.00335.2023] [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: 10/18/2023] [Revised: 12/25/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes, specialized postmitotic cells, are central players in various kidney-related diseases. Zebrafish have become a valuable model system for studying podocyte biology because they are genetically easy to manipulate, transparent, and their glomerular structure is similar to that of mammals. This review provides an overview of the knowledge of podocyte biology in zebrafish larvae, with particular focus on their essential contribution to understanding the mechanisms that underlie kidney diseases as well as supporting drug development. In addition, special attention is given to advances in live-imaging techniques allowing the observation of dynamic processes, including podocyte motility, podocyte process behavior, and glomerulus maturation. The review further addresses the functional aspects of podocytes in zebrafish larvae. This includes topics such as glomerular filtration, ultrastructural analyses, and evaluation of podocyte response to nephrotoxic insults. Studies presented in this context have provided important insights into the maintenance and resistance of the glomerular filtration barrier in zebrafish larvae and explored the potential transferability of these findings to mammals such as mice, rats, and most importantly, humans. The recent ability to identify potential therapeutic targets represents a promising new way to identify drugs that could effectively treat podocyte-associated glomerulopathies in humans. In summary, this review gives an overview about the importance of zebrafish as a model for podocyte-related disease and targeted drug development. It also highlights the key role of advanced imaging techniques in transparent zebrafish larvae, improving our understanding of glomerular diseases and the significant potential for translation of these findings to humans.
Collapse
Affiliation(s)
- Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
13
|
Wang Y, Huang H, Weng H, Jia C, Liao B, Long Y, Yu F, Nie Y. Talin mechanotransduction in disease. Int J Biochem Cell Biol 2024; 166:106490. [PMID: 37914021 DOI: 10.1016/j.biocel.2023.106490] [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: 07/29/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
Talin protein (Talin 1/2) is a mechanosensitive cytoskeleton protein. The unique structure of the Talin plays a vital role in transmitting mechanical forces. Talin proteins connect the extracellular matrix to the cytoskeleton by linking to integrins and actin, thereby mediating the conversion of mechanical signals into biochemical signals and influencing disease progression as potential diagnostic indicators, therapeutic targets, and prognostic indicators of various diseases. Most studies in recent years have confirmed that mechanical forces also have a crucial role in the development of disease, and Talin has been found to play a role in several diseases. Still, more studies need to be done on how Talin is involved in mechanical signaling in disease. This review focuses on the mechanical signaling of Talin in disease, aiming to summarize the mechanisms by which Talin plays a role in disease and to provide references for further studies.
Collapse
Affiliation(s)
- Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Haozhong Huang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Huimin Weng
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Chunsen Jia
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yongmei Nie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China.
| |
Collapse
|
14
|
Korolj A, Kohler RH, Scott E, Halabi EA, Lucas K, Carlson JC, Weissleder R. Perfusion Window Chambers Enable Interventional Analyses of Tumor Microenvironments. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304886. [PMID: 37870204 PMCID: PMC10700240 DOI: 10.1002/advs.202304886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/29/2023] [Indexed: 10/24/2023]
Abstract
Intravital microscopy (IVM) allows spatial and temporal imaging of different cell types in intact live tissue microenvironments. IVM has played a critical role in understanding cancer biology, invasion, metastases, and drug development. One considerable impediment to the field is the inability to interrogate the tumor microenvironment and its communication cascades during disease progression and therapeutic interventions. Here, a new implantable perfusion window chamber (PWC) is described that allows high-fidelity in vivo microscopy, local administration of stains and drugs, and longitudinal sampling of tumor interstitial fluid. This study shows that the new PWC design allows cyclic multiplexed imaging in vivo, imaging of drug action, and sampling of tumor-shed materials. The PWC will be broadly useful as a novel perturbable in vivo system for deciphering biology in complex microenvironments.
Collapse
Affiliation(s)
- Anastasia Korolj
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
| | - Rainer H. Kohler
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Ella Scott
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Elias A. Halabi
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Kilean Lucas
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Jonathan C.T. Carlson
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Cancer CenterMassachusetts General Hospital55 Fruit StreetBostonMA02114USA
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
- Cancer CenterMassachusetts General Hospital55 Fruit StreetBostonMA02114USA
| |
Collapse
|
15
|
Kourpa A, Schulz A, Mangelsen E, Kaiser-Graf D, Koppers N, Stoll M, Rothe M, Bader M, Purfürst B, Kunz S, Gladytz T, Niendorf T, Bachmann S, Mutig K, Bolbrinker J, Panáková D, Kreutz R. Studies in Zebrafish and Rat Models Support Dual Blockade of EP2 and EP4 (Prostaglandin E 2 Receptors Type 2 and 4) for Renoprotection in Glomerular Hyperfiltration and Albuminuria. Hypertension 2023; 80:771-782. [PMID: 36715011 DOI: 10.1161/hypertensionaha.122.20392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Glomerular hyperfiltration (GH) is an important mechanism in the development of albuminuria in hypertension. Upregulation of COX2 (cyclooxygenase 2) and prostaglandin E2 (PGE2) was linked to podocyte damage in GH. We explored the potential renoprotective effects of either separate or combined pharmacological blockade of EP2 (PGE2 receptor type 2) and EP4 (PGE2 receptor type 4) in GH. METHODS We conducted in vivo studies in a transgenic zebrafish model (Tg[fabp10a:gc-EGFP]) suitable for analysis of glomerular filtration barrier function and a genetic rat model with GH, albuminuria, and upregulation of PGE2. Similar pharmacological interventions and primary outcome analysis on albuminuria phenotype development were conducted in both model systems. RESULTS Stimulation of zebrafish embryos with PGE2 induced an albuminuria-like phenotype, thus mimicking the suggested PGE2 effects on glomerular filtration barrier dysfunction. Both separate and combined blockade of EP2 and EP4 reduced albuminuria phenotypes in zebrafish and rat models. A significant correlation between albuminuria and podocyte damage in electron microscopy imaging was identified in the rat model. Dual blockade of both receptors showed a pronounced synergistic suppression of albuminuria. Importantly, this occurred without changes in arterial blood pressure, glomerular filtration rate, or tissue oxygenation in magnetic resonance imaging, while RNA sequencing analysis implicated a potential role of circadian clock genes. CONCLUSIONS Our findings confirm a role of PGE2 in the development of albuminuria in GH and support the renoprotective potential of combined pharmacological blockade of EP2 and EP4 receptors. These data support further translational research to explore this therapeutic option and a possible role of circadian clock genes.
Collapse
Affiliation(s)
- Aikaterini Kourpa
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Angela Schulz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Eva Mangelsen
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Debora Kaiser-Graf
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Nils Koppers
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | - Monika Stoll
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | | | - Michael Bader
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (M.B.).,Charité-Universitätsmedizin Berlin, Germany (M.B.).,Institute for Biology, University of Lübeck, Germany (M.B.)
| | - Bettina Purfürst
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Severine Kunz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thomas Gladytz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thoralf Niendorf
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Sebastian Bachmann
- Institute of Vegetative Anatomy (S.B.), Charité-Universitätsmedizin Berlin, Germany
| | - Kerim Mutig
- Institute of Translational Physiology (K.M.), Charité-Universitätsmedizin Berlin, Germany
| | - Juliane Bolbrinker
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Daniela Panáková
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
16
|
Lee JB, Kim H, Kim S, Sung GY. Fabrication and Evaluation of Tubule-on-a-Chip with RPTEC/HUVEC Co-Culture Using Injection-Molded Polycarbonate Chips. MICROMACHINES 2022; 13:mi13111932. [PMID: 36363953 PMCID: PMC9698344 DOI: 10.3390/mi13111932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 05/27/2023]
Abstract
To simulate the ADME process such as absorption, distribution, metabolism, and excretion in the human body after drug administration and to confirm the applicability of the mass production process, a microfluidic chip injection molded with polycarbonate (injection-molded chip (I-M chip)) was fabricated. Polycarbonate materials were selected to minimize drug absorption. As a first step to evaluate the I-M chip, RPTEC (Human Renal Proximal Tubule Epithelial Cells) and HUVEC (Human Umbilical Vein Endothelial Cells) were co-cultured, and live and dead staining, TEER (trans-epithelial electrical resistance), glucose reabsorption, and permeability were compared using different membrane pore sizes of 0.4 μm and 3 μm. Drug excretion was confirmed through a pharmacokinetic test with metformin and cimetidine, and the gene expression of drug transporters was confirmed. As a result, it was confirmed that the cell viability was higher in the 3 μm pore size than in the 0.4 μm, the cell culture performed better, and the drug secretion was enhanced when the pore size was large. The injection-molded polycarbonate microfluidic chip is anticipated to be commercially viable for drug screening devices, particularly ADME tests.
Collapse
Affiliation(s)
- Ju-Bi Lee
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Hyoungseob Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Sol Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
- Major in Materials Science and Engineering, School of Future Convergence, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
17
|
Srivastava T, Garola RE, Zhou J, Boinpelly VC, Priya L, Ali MF, Rezaiekhaligh MH, Heruth DP, Novak J, Alon US, Joshi T, Jiang Y, McCarthy ET, Savin VJ, Johnson ML, Sharma R, Sharma M. Prostanoid receptors in hyperfiltration-mediated glomerular injury: Novel agonists and antagonists reveal opposing roles for EP2 and EP4 receptors. FASEB J 2022; 36:e22559. [PMID: 36125047 DOI: 10.1096/fj.202200875r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
Increased fluid-flow shear stress (FFSS) contributes to hyperfiltration-induced podocyte and glomerular injury resulting in progression of chronic kidney disease (CKD). We reported that increased FFSS in vitro and in vivo upregulates PGE2 receptor EP2 (but not EP4 expression), COX2-PGE2 -EP2 axis, and EP2-linked Akt-GSK3β-β-catenin signaling pathway in podocytes. To understand and use the disparities between PGE2 receptors, specific agonists, and antagonists of EP2 and EP4 were used to assess phosphorylation of Akt, GSK3β and β-catenin in podocytes using Western blotting, glomerular filtration barrier function using in vitro albumin permeability (Palb ) assay, and mitigation of hyperfiltration-induced injury in unilaterally nephrectomized (UNX) mice at 1 and 6 months. Results show an increase in Palb by PGE2 , EP2 agonist (EP2AGO ) and EP4 antagonist (EP4ANT ), but not by EP2 antagonist (EP2ANT ) or EP4 agonist (EP4AGO ). Pretreatment with EP2ANT blocked the effect of PGE2 or EP2AGO on Palb . Modulation of EP2 and EP4 also induced opposite effects on phosphorylation of Akt and β-Catenin. Individual agonists or antagonists of EP2 or EP4 did not induce significant improvement in albuminuria in UNX mice. However, treatment with a combination EP2ANT + EP4AGO for 1 or 6 months caused a robust decrease in albuminuria. EP2ANT + EP4AGO combination did not impact adaptive hypertrophy or increased serum creatinine. Observed differences between expression of EP2 and EP4 on the glomerular barrier highlight these receptors as potential targets for intervention. Safe and effective mitigating effect of EP2ANT + EP4AGO presents a novel opportunity to delay the progression of hyperfiltration-associated CKD as seen in transplant donors.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA.,Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Lakshmi Priya
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammed Farhan Ali
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammad H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Ellen T McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Virginia J Savin
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Ram Sharma
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
18
|
Wang D, Gust M, Ferrell N. Kidney-on-a-Chip: Mechanical Stimulation and Sensor Integration. SENSORS (BASEL, SWITZERLAND) 2022; 22:6889. [PMID: 36146238 PMCID: PMC9503911 DOI: 10.3390/s22186889] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 06/16/2023]
Abstract
Bioengineered in vitro models of the kidney offer unprecedented opportunities to better mimic the in vivo microenvironment. Kidney-on-a-chip technology reproduces 2D or 3D features which can replicate features of the tissue architecture, composition, and dynamic mechanical forces experienced by cells in vivo. Kidney cells are exposed to mechanical stimuli such as substrate stiffness, shear stress, compression, and stretch, which regulate multiple cellular functions. Incorporating mechanical stimuli in kidney-on-a-chip is critically important for recapitulating the physiological or pathological microenvironment. This review will explore approaches to applying mechanical stimuli to different cell types using kidney-on-a-chip models and how these systems are used to study kidney physiology, model disease, and screen for drug toxicity. We further discuss sensor integration into kidney-on-a-chip for monitoring cellular responses to mechanical or other pathological stimuli. We discuss the advantages, limitations, and challenges associated with incorporating mechanical stimuli in kidney-on-a-chip models for a variety of applications. Overall, this review aims to highlight the importance of mechanical stimuli and sensor integration in the design and implementation of kidney-on-a-chip devices.
Collapse
Affiliation(s)
- Dan Wang
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Matthew Gust
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Statistics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Nicholas Ferrell
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
19
|
Effects of Genistein on Common Kidney Diseases. Nutrients 2022; 14:nu14183768. [PMID: 36145144 PMCID: PMC9506319 DOI: 10.3390/nu14183768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 12/16/2022] Open
Abstract
Genistein is a naturally occurring phytoestrogen (soy or soybean products) that is classified as an isoflavone, and its structure is similar to that of endogenous estrogens; therefore, genistein can exert an estrogen-like effect via estrogen receptors. Additionally, genistein is a tyrosine kinase inhibitor, which enables it to block abnormal cell growth and proliferation signals through the inhibition of tyrosine kinase. Genistein is also an angiogenesis inhibitor and an antioxidant. Genistein has effects on kidney cells, some of the kidney’s physiological functions, and a variety of kidney diseases. First, genistein exerts a protective effect on normal cells by reducing the inflammatory response, inhibiting apoptosis, inhibiting oxidative stress, inhibiting remodeling, etc., but after cell injury, the protective effect of genistein decreases or even has the opposite effect. Second, genistein can regulate renin intake to maintain blood pressure balance, regulate calcium uptake to regulate Ca2+ and Pi balances, and reduce vasodilation to promote diuresis. Third, genistein has beneficial effects on a variety of kidney diseases (including acute kidney disease, kidney cancer, and different chronic kidney diseases), such as reducing symptoms, delaying disease progression, and improving prognosis. Therefore, this paper reviews animal and human studies on the protective effects of genistein on the kidney in vivo and in vitro to provide a reference for clinical research in the future.
Collapse
|
20
|
Madhavan SM, Konieczkowski M, Bruggeman LA, DeWalt M, Nguyen JK, O'Toole JF, Sedor JR. Essential role of Wtip in mouse development and maintenance of the glomerular filtration barrier. Am J Physiol Renal Physiol 2022; 323:F272-F287. [PMID: 35862649 PMCID: PMC9394782 DOI: 10.1152/ajprenal.00051.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Wilms' tumor interacting protein (Wtip) has been implicated in cell junction assembly and cell differentiation and interacts with proteins in the podocyte slit diaphragm, where it regulates podocyte phenotype. To define Wtip expression and function in the kidney, we created a Wtip-deleted mouse model using β-galactosidase-neomycin (β-geo) gene trap technology. Wtip gene trap mice were embryonic lethal, suggesting additional developmental roles outside kidney function. Using β-geo heterozygous and normal mice, Wtip expression was identified in the developing kidneys, heart, and eyes. In the kidney, expression was restricted to podocytes, which appeared initially at the capillary loop stage coinciding with terminal podocyte differentiation. Heterozygous mice had an expected lifespan and showed no evidence of proteinuria or glomerular pathology. However, heterozygous mice were more susceptible to glomerular injury than wild-type littermates and developed more significant and prolonged proteinuria in response to lipopolysaccharide or adriamycin. In normal human kidneys, WTIP expression patterns were consistent with observations in mice and were lost in glomeruli concurrent with loss of synaptopodin expression in disease. Mechanistically, we identified the Rho guanine nucleotide exchange factor 12 (ARHGEF12) as a binding partner for WTIP. ARHGEF12 was expressed in human podocytes and formed high-affinity interactions through their LIM- and PDZ-binding domains. Our findings suggest that Wtip is essential for early murine embryonic development and maintaining normal glomerular filtration barrier function, potentially regulating slit diaphragm and foot process function through Rho effector proteins.NEW & NOTEWORTHY This study characterized dynamic expression patterns of Wilms' tumor interacting protein (Wtip) and demonstrates the novel role of Wtip in murine development and maintenance of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Sethu M Madhavan
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Leslie A Bruggeman
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
| | - Megan DeWalt
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Jane K Nguyen
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio
| | - John F O'Toole
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
| | - John R Sedor
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
21
|
Kim H, Lee JB, Kim K, Sung GY. Effect of shear stress on the proximal tubule-on-a-chip for multi-organ microphysiological system. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
22
|
Journey of organ on a chip technology and its role in future healthcare scenario. APPLIED SURFACE SCIENCE ADVANCES 2022; 9. [PMCID: PMC9000345 DOI: 10.1016/j.apsadv.2022.100246] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Organ on a chip refers to microengineered biomimetic system which reflects structural and functional characteristics of human tissue. It involves biomaterial technology, cell biology and engineering combined together in a miniaturized platform. Several models using different organs such as lungs on a chip, liver on a chip, kidney on a chip, heart on a chip, intestine on a chip and skin on a chip have been successfully developed. Food and Drug administration (FDA) has also shown confidence in this technology and has partnered with industries/institutes which are working with this technology. In this review, the concepts and applications of Organ on a chip model in different scientific domains including disease model development, drug screening, toxicology, pathogenesis study, efficacy testing and virology is discussed. It is envisaged that amalgamation of various organs on chip modules into a unified body on chip device is of utmost importance for diagnosis and treatment, especially considering the complications due to the ongoing COVID-19 pandemic. It is expected that the market demand for developing organ on chip devices to skyrocket in the near future.
Collapse
|
23
|
Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci (Lond) 2022; 136:493-520. [PMID: 35415751 PMCID: PMC9008595 DOI: 10.1042/cs20210625] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Albuminuria is the hallmark of both primary and secondary proteinuric glomerulopathies, including focal segmental glomerulosclerosis (FSGS), obesity-related nephropathy, and diabetic nephropathy (DN). Moreover, albuminuria is an important feature of all chronic kidney diseases (CKDs). Podocytes play a key role in maintaining the permselectivity of the glomerular filtration barrier (GFB) and injury of the podocyte, leading to foot process (FP) effacement and podocyte loss, the unifying underlying mechanism of proteinuric glomerulopathies. The metabolic insult of hyperglycemia is of paramount importance in the pathogenesis of DN, while insults leading to podocyte damage are poorly defined in other proteinuric glomerulopathies. However, shared mechanisms of podocyte damage have been identified. Herein, we will review the role of haemodynamic and oxidative stress, inflammation, lipotoxicity, endocannabinoid (EC) hypertone, and both mitochondrial and autophagic dysfunction in the pathogenesis of the podocyte damage, focussing particularly on their role in the pathogenesis of DN. Gaining a better insight into the mechanisms of podocyte injury may provide novel targets for treatment. Moreover, novel strategies for boosting podocyte repair may open the way to podocyte regenerative medicine.
Collapse
|
24
|
Chen J, Wang X, He Q, Harris RC. TAZ is important for maintenance of the integrity of podocytes. Am J Physiol Renal Physiol 2022; 322:F419-F428. [PMID: 35157550 PMCID: PMC8934679 DOI: 10.1152/ajprenal.00426.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022] Open
Abstract
The podocyte is an important component of the glomerular filtration barrier, and maintenance of the integrity of its highly specified structure and function is critical for normal kidney function. Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) are two crucial effectors of the Hippo signaling pathway, and recent studies have shown that podocyte-specific YAP deletion causes podocyte apoptosis and the development of focal segmental glomerulosclerosis followed by progressive renal failure. In the present study, we investigated a potential role of the YAP paralog TAZ in podocytes. TAZ was found to be constitutively active in podocytes, and mice with podocyte-specific deletion of TAZ (TazpodKO) developed proteinuria starting at 4 wk of age and had increased podocyte apoptosis. Using primary cultured podocytes or immortalized mouse podocytes from Tazflox/flox mice, we found that TAZ is a transcriptional activator for TEAD-dependent expression of synaptopodin, zonula occludens-1, and zonula occludens-2. This is the first study to determine that TAZ plays an important role in the maintenance of the structure and function of podocytes.NEW & NOTEWORTHY Podocytes play an important role in maintaining the integrity of the structure and function of the kidney. We observed that mice with selective deletion of transcriptional coactivator with PDZ-binding motif (TAZ) in podocytes developed proteinuria. TAZ is constitutively active and critical for expression of synaptopodin, zonula occludens-1, and zonula occludens-2 in podocytes. The findings of this study implicate TAZ as an important mediator of podocyte structural integrity and provide further insights into the role of Hippo-Yes-associated protein/TAZ in podocyte biology.
Collapse
Affiliation(s)
- Jianchun Chen
- United States Department of Veterans Affairs, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xiaoyong Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qian He
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Raymond C Harris
- United States Department of Veterans Affairs, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
25
|
Valverde MG, Mille LS, Figler KP, Cervantes E, Li VY, Bonventre JV, Masereeuw R, Zhang YS. Biomimetic models of the glomerulus. Nat Rev Nephrol 2022; 18:241-257. [PMID: 35064233 PMCID: PMC9949601 DOI: 10.1038/s41581-021-00528-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/17/2022]
Abstract
The use of biomimetic models of the glomerulus has the potential to improve our understanding of the pathogenesis of kidney diseases and to enable progress in therapeutics. Current in vitro models comprise organ-on-a-chip, scaffold-based and organoid approaches. Glomerulus-on-a-chip designs mimic components of glomerular microfluidic flow but lack the inherent complexity of the glomerular filtration barrier. Scaffold-based 3D culture systems and organoids provide greater microenvironmental complexity but do not replicate fluid flows and dynamic responses to fluidic stimuli. As the available models do not accurately model the structure or filtration function of the glomerulus, their applications are limited. An optimal approach to glomerular modelling is yet to be developed, but the field will probably benefit from advances in biofabrication techniques. In particular, 3D bioprinting technologies could enable the fabrication of constructs that recapitulate the complex structure of the glomerulus and the glomerular filtration barrier. The next generation of in vitro glomerular models must be suitable for high(er)-content or/and high(er)-throughput screening to enable continuous and systematic monitoring. Moreover, coupling of glomerular or kidney models with those of other organs is a promising approach to enable modelling of partial or full-body responses to drugs and prediction of therapeutic outcomes.
Collapse
Affiliation(s)
- Marta G Valverde
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Luis S Mille
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Kianti P Figler
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Ernesto Cervantes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Vanessa Y Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Joseph V Bonventre
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA.
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
26
|
Sharma M, Singh V, Sharma R, Koul A, McCarthy ET, Savin VJ, Joshi T, Srivastava T. Glomerular Biomechanical Stress and Lipid Mediators during Cellular Changes Leading to Chronic Kidney Disease. Biomedicines 2022; 10:407. [PMID: 35203616 PMCID: PMC8962328 DOI: 10.3390/biomedicines10020407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperfiltration is an important underlying cause of glomerular dysfunction associated with several systemic and intrinsic glomerular conditions leading to chronic kidney disease (CKD). These include obesity, diabetes, hypertension, focal segmental glomerulosclerosis (FSGS), congenital abnormalities and reduced renal mass (low nephron number). Hyperfiltration-associated biomechanical forces directly impact the cell membrane, generating tensile and fluid flow shear stresses in multiple segments of the nephron. Ongoing research suggests these biomechanical forces as the initial mediators of hyperfiltration-induced deterioration of podocyte structure and function leading to their detachment and irreplaceable loss from the glomerular filtration barrier. Membrane lipid-derived polyunsaturated fatty acids (PUFA) and their metabolites are potent transducers of biomechanical stress from the cell surface to intracellular compartments. Omega-6 and ω-3 long-chain PUFA from membrane phospholipids generate many versatile and autacoid oxylipins that modulate pro-inflammatory as well as anti-inflammatory autocrine and paracrine signaling. We advance the idea that lipid signaling molecules, related enzymes, metabolites and receptors are not just mediators of cellular stress but also potential targets for developing novel interventions. With the growing emphasis on lifestyle changes for wellness, dietary fatty acids are potential adjunct-therapeutics to minimize/treat hyperfiltration-induced progressive glomerular damage and CKD.
Collapse
Affiliation(s)
- Mukut Sharma
- Research and Development Service, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, MO 66160, USA;
| | - Vikas Singh
- Neurology, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Ram Sharma
- Research and Development Service, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Arnav Koul
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
| | - Ellen T. McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, MO 66160, USA;
| | - Virginia J. Savin
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO 65201, USA;
| | - Tarak Srivastava
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri, Kansas City, MO 64108, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, MO 64108, USA
| |
Collapse
|
27
|
Fanizza F, Campanile M, Forloni G, Giordano C, Albani D. Induced pluripotent stem cell-based organ-on-a-chip as personalized drug screening tools: A focus on neurodegenerative disorders. J Tissue Eng 2022; 13:20417314221095339. [PMID: 35570845 PMCID: PMC9092580 DOI: 10.1177/20417314221095339] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/04/2022] [Indexed: 01/15/2023] Open
Abstract
The Organ-on-a-Chip (OoC) technology shows great potential to revolutionize the drugs development pipeline by mimicking the physiological environment and functions of human organs. The translational value of OoC is further enhanced when combined with patient-specific induced pluripotent stem cells (iPSCs) to develop more realistic disease models, paving the way for the development of a new generation of patient-on-a-chip devices. iPSCs differentiation capacity leads to invaluable improvements in personalized medicine. Moreover, the connection of single-OoC into multi-OoC or body-on-a-chip allows to investigate drug pharmacodynamic and pharmacokinetics through the study of multi-organs cross-talks. The need of a breakthrough thanks to this technology is particularly relevant within the field of neurodegenerative diseases, where the number of patients is increasing and the successful rate in drug discovery is worryingly low. In this review we discuss current iPSC-based OoC as drug screening models and their implication in development of new therapies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fanizza
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
28
|
Yu P, Duan Z, Liu S, Pachon I, Ma J, Hemstreet GP, Zhang Y. Drug-Induced Nephrotoxicity Assessment in 3D Cellular Models. MICROMACHINES 2021; 13:mi13010003. [PMID: 35056167 PMCID: PMC8780064 DOI: 10.3390/mi13010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
The kidneys are often involved in adverse effects and toxicity caused by exposure to foreign compounds, chemicals, and drugs. Early predictions of these influences are essential to facilitate new, safe drugs to enter the market. However, in current drug treatments, drug-induced nephrotoxicity accounts for 1/4 of reported serious adverse reactions, and 1/3 of them are attributable to antibiotics. Drug-induced nephrotoxicity is driven by multiple mechanisms, including altered glomerular hemodynamics, renal tubular cytotoxicity, inflammation, crystal nephropathy, and thrombotic microangiopathy. Although the functional proteins expressed by renal tubules that mediate drug sensitivity are well known, current in vitro 2D cell models do not faithfully replicate the morphology and intact renal tubule function, and therefore, they do not replicate in vivo nephrotoxicity. The kidney is delicate and complex, consisting of a filter unit and a tubular part, which together contain more than 20 different cell types. The tubular epithelium is highly polarized, and maintaining cellular polarity is essential for the optimal function and response to environmental signals. Cell polarity depends on the communication between cells, including paracrine and autocrine signals, as well as biomechanical and chemotaxis processes. These processes affect kidney cell proliferation, migration, and differentiation. For drug disposal research, the microenvironment is essential for predicting toxic reactions. This article reviews the mechanism of drug-induced kidney injury, the types of nephrotoxicity models (in vivo and in vitro models), and the research progress related to drug-induced nephrotoxicity in three-dimensional (3D) cellular culture models.
Collapse
Affiliation(s)
- Pengfei Yu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Shuang Liu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Ivan Pachon
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | - Jianxing Ma
- Department of Biochemistry, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | | | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
- Correspondence: ; Tel.: +1-336-713-1189
| |
Collapse
|
29
|
Butt L, Unnersjö-Jess D, Höhne M, Schermer B, Edwards A, Benzing T. A mathematical estimation of the physical forces driving podocyte detachment. Kidney Int 2021; 100:1054-1062. [PMID: 34332959 DOI: 10.1016/j.kint.2021.06.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/27/2021] [Accepted: 06/18/2021] [Indexed: 01/21/2023]
Abstract
Loss of podocytes, possibly through the detachment of viable cells, is a hallmark of progressive glomerular disease. Podocytes are exposed to considerable physical forces due to pressure and flow resulting in circumferential wall stress and tangential shear stress exerted on the podocyte cell body, which have been proposed to contribute to podocyte depletion. However, estimations of in vivo alterations of physical forces in glomerular disease have been hampered by a lack of quantitative functional and morphological data. Here, we used ultra-resolution data and computational analyses in a mouse model of human disease, hereditary late-onset focal segmental glomerular sclerosis, to calculate increased mechanical stress upon podocyte injury. Transversal shear stress on the lateral walls of the foot processes was prominently increased during the initial stages of podocyte detachment. Thus, our study highlights the importance of targeting glomerular hemodynamics to treat glomerular disease.
Collapse
Affiliation(s)
- Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - David Unnersjö-Jess
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
30
|
Greiten JK, Kliewe F, Schnarre A, Artelt N, Schröder S, Rogge H, Amann K, Daniel C, Lindenmeyer MT, Cohen CD, Endlich K, Endlich N. The role of filamins in mechanically stressed podocytes. FASEB J 2021; 35:e21560. [PMID: 33860543 DOI: 10.1096/fj.202001179rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 11/11/2022]
Abstract
Glomerular hypertension induces mechanical load to podocytes, often resulting in podocyte detachment and the development of glomerulosclerosis. Although it is well known that podocytes are mechanosensitive, the mechanosensors and mechanotransducers are still unknown. Since filamin A, an actin-binding protein, is already described to be a mechanosensor and mechanotransducer, we hypothesized that filamins could be important for the outside-in signaling as well as the actin cytoskeleton of podocytes under mechanical stress. In this study, we demonstrate that filamin A is the main isoform of the filamin family that is expressed in cultured podocytes. Together with filamin B, filamin A was significantly up-regulated during mechanical stretch (3 days, 0.5 Hz, and 5% extension). To study the role of filamin A in cultured podocytes under mechanical stress, filamin A was knocked down (Flna KD) by specific siRNA. Additionally, we established a filamin A knockout podocyte cell line (Flna KO) by CRISPR/Cas9. Knockdown and knockout of filamin A influenced the expression of synaptopodin, a podocyte-specific protein, focal adhesions as well as the morphology of the actin cytoskeleton. Moreover, the cell motility of Flna KO podocytes was significantly increased. Since the knockout of filamin A has had no effect on cell adhesion of podocytes during mechanical stress, we simultaneously knocked down the expression of filamin A and B. Thereby, we observed a significant loss of podocytes during mechanical stress indicating a compensatory mechanism. Analyzing hypertensive mice kidneys as well as biopsies of patients suffering from diabetic nephropathy, we found an up-regulation of filamin A in podocytes in contrast to the control. In summary, filamin A and B mediate matrix-actin cytoskeleton interactions which are essential for the adaptation of cultured podocyte to mechanical stress.
Collapse
Affiliation(s)
- Jonas K Greiten
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Annabel Schnarre
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nadine Artelt
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Sindy Schröder
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Rogge
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja T Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
31
|
Srivastava T, Heruth DP, Duncan RS, Rezaiekhaligh MH, Garola RE, Priya L, Zhou J, Boinpelly VC, Novak J, Ali MF, Joshi T, Alon US, Jiang Y, McCarthy ET, Savin VJ, Sharma R, Johnson ML, Sharma M. Transcription Factor β-Catenin Plays a Key Role in Fluid Flow Shear Stress-Mediated Glomerular Injury in Solitary Kidney. Cells 2021; 10:cells10051253. [PMID: 34069476 PMCID: PMC8159099 DOI: 10.3390/cells10051253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 01/21/2023] Open
Abstract
Increased fluid flow shear stress (FFSS) in solitary kidney alters podocyte function in vivo. FFSS-treated cultured podocytes show upregulated AKT-GSK3β-β-catenin signaling. The present study was undertaken to confirm (i) the activation of β-catenin signaling in podocytes in vivo using unilaterally nephrectomized (UNX) TOPGAL mice with the β-galactosidase reporter gene for β-catenin activation, (ii) β-catenin translocation in FFSS-treated mouse podocytes, and (iii) β-catenin signaling using publicly available data from UNX mice. The UNX of TOPGAL mice resulted in glomerular hypertrophy and increased the mesangial matrix consistent with hemodynamic adaptation. Uninephrectomized TOPGAL mice showed an increased β-galactosidase expression at 4 weeks but not at 12 weeks, as assessed using immunofluorescence microscopy (p < 0.001 at 4 weeks; p = 0.16 at 12 weeks) and X-gal staining (p = 0.008 at 4 weeks; p = 0.65 at 12 weeks). Immunofluorescence microscopy showed a significant increase in phospho-β-catenin (Ser552, p = 0.005) at 4 weeks but not at 12 weeks (p = 0.935) following UNX, and the levels of phospho-β-catenin (Ser675) did not change. In vitro FFSS caused a sustained increase in the nuclear translocation of phospho-β-catenin (Ser552) but not phospho-β-catenin (Ser675) in podocytes. The bioinformatic analysis of the GEO dataset, #GSE53996, also identified β-catenin as a key upstream regulator. We conclude that transcription factor β-catenin mediates FFSS-induced podocyte (glomerular) injury in solitary kidney.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
- Correspondence: ; Tel.: +1-816-234-3010; Fax: +1-816-302-9919
| | - Daniel P. Heruth
- Children’s Mercy Research Institute, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - R. Scott Duncan
- School of Biological Sciences, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Mohammad H. Rezaiekhaligh
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Robert E. Garola
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Lakshmi Priya
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Jianping Zhou
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Varun C. Boinpelly
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35487, USA;
| | - Mohammed Farhan Ali
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO 65211, USA;
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA;
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- MU Data Science and Informatics Institute, University of Missouri, Columbia, MO 65211, USA
| | - Uri S. Alon
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA;
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Ellen T. McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Virginia J. Savin
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Ram Sharma
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Mark L. Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Mukut Sharma
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| |
Collapse
|
32
|
Affiliation(s)
- Thomas Benzing
- From Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine, and University Hospital Cologne, and the Excellence Cluster CECAD, University of Cologne, Cologne, Germany (T.B.); and the Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston (D.S.)
| | - David Salant
- From Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine, and University Hospital Cologne, and the Excellence Cluster CECAD, University of Cologne, Cologne, Germany (T.B.); and the Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston (D.S.)
| |
Collapse
|
33
|
Dembele M, Delafosse M, Yousfi N, Debiec H, Ngo K, Plaisier E, Ronco P, Perry G. [Models of glomerular filtration barrier : New developments]. Med Sci (Paris) 2021; 37:242-248. [PMID: 33739271 DOI: 10.1051/medsci/2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In this article, we present the latest innovations to generate in vitro models of the glomerular filtration barrier. There is currently a growing interest for such model systems that allow to reduce the use of animal models. Methodologies to improve their physiological relevance have taken advantage of the development of induced pluripotent stem cells and of bioengineering, particularly tissue engineering. Here, we first introduce the methods to overcome the limitations of the currently used glomerular cells based on the use of stem cells. The different approaches to obtain podocytes, the most important cells in the glomerulus, are presented. Finally, we emphasize the importance of the glomerular microenvironment in maintaining the glomerular cell phenotype, which can be achieved by co-culturing different glomerular cells, integration of biomaterials mimicking the extracellular matrix and introduction of flows with microfluidics.
Collapse
Affiliation(s)
- Mahamadou Dembele
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Marion Delafosse
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Nadhir Yousfi
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Hanna Debiec
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Kieu Ngo
- Sorbonne Université, CNRS, Laboratoire interfaces et systèmes électrochimiques, LISE, Campus Pierre et Marie Curie, 4 place Jussieu 75252 Paris, France
| | - Emmanuelle Plaisier
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Pierre Ronco
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Guillaume Perry
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France - CNRS, Sorbonne université, Laboratoire de génie électrique et électronique de Paris, GeePs, Campus Pierre et Marie Curie, 4 Place Jussieu 75252 Paris, France
| |
Collapse
|
34
|
Ayuso JM, Park KY, Virumbrales-Muñoz M, Beebe DJ. Toward improved in vitro models of human cancer. APL Bioeng 2021; 5:010902. [PMID: 33532672 PMCID: PMC7822630 DOI: 10.1063/5.0026857] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of death across the world and continues to increase in incidence. Despite years of research, multiple tumors (e.g., glioblastoma, pancreatic cancer) still have limited treatment options in the clinic. Additionally, the attrition rate and cost of drug development have continued to increase. This trend is partly explained by the poor predictive power of traditional in vitro tools and animal models. Moreover, multiple studies have highlighted that cell culture in traditional Petri dishes commonly fail to predict drug sensitivity. Conversely, animal models present differences in tumor biology compared with human pathologies, explaining why promising therapies tested in animal models often fail when tested in humans. The surging complexity of patient management with the advent of cancer vaccines, immunotherapy, and precision medicine demands more robust and patient-specific tools to better inform our understanding and treatment of human cancer. Advances in stem cell biology, microfluidics, and cell culture have led to the development of sophisticated bioengineered microscale organotypic models (BMOMs) that could fill this gap. In this Perspective, we discuss the advantages and limitations of patient-specific BMOMs to improve our understanding of cancer and how these tools can help to confer insight into predicting patient response to therapy.
Collapse
Affiliation(s)
| | - Keon-Young Park
- Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
35
|
Agarwal S, Sudhini YR, Reiser J, Altintas MM. From Infancy to Fancy: A Glimpse into the Evolutionary Journey of Podocytes in Culture. KIDNEY360 2020; 2:385-397. [PMID: 35373019 PMCID: PMC8740988 DOI: 10.34067/kid.0006492020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/22/2020] [Indexed: 02/04/2023]
Abstract
Podocytes are critical components of the filtration barrier and responsible for maintaining healthy kidney function. An assault on podocytes is generally associated with progression of chronic glomerular diseases. Therefore, podocyte pathophysiology is a favorite research subject for nephrologists. Despite this, podocyte research has lagged because of the unavailability of techniques for culturing such specialized cells ex vivo in quantities that are adequate for mechanistic studies. In recent years, this problem was circumvented by the efforts of researchers, who successfully developed several in vitro podocyte cell culture model systems that paved the way for incredible discoveries in the field of nephrology. This review sets us on a journey that provides a comprehensive insight into the groundbreaking breakthroughs and novel technologic advances made in the field of podocyte cell culture so far, beginning from its inception, evolution, and progression. In this study, we also describe in detail the pros and cons of different models that are being used to culture podocytes. Our extensive and exhaustive deliberation on the status of podocyte cell culture will facilitate researchers to choose wisely an appropriate model for their own research to avoid potential pitfalls in the future.
Collapse
|
36
|
The mechanobiology of kidney podocytes in health and disease. Clin Sci (Lond) 2020; 134:1245-1253. [PMID: 32501496 DOI: 10.1042/cs20190764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023]
Abstract
Chronic kidney disease (CKD) substantially reduces quality of life and leads to premature death for thousands of people each year. Dialysis and kidney organ transplants remain prevalent therapeutic avenues but carry significant medical, economic and social burden. Podocytes are responsible for blood filtration selectivity in the kidney, where they extend a network of foot processes (FPs) from their cell bodies which surround endothelial cells and interdigitate with those on neighbouring podocytes to form narrow slit diaphragms (SDs). During aging, some podocytes are lost naturally but accelerated podocyte loss is a hallmark of CKD. Insights into the origin of degenerative podocyte loss will help answer important questions about kidney function and lead to substantial health benefits. Here, approaches that uncover insights into podocyte mechanobiology are reviewed, both those that interrogate the biophysical properties of podocytes and how the external physical environment affects podocyte behaviour, and also those that interrogate the biophysical effects that podocytes exert on their surroundings.
Collapse
|
37
|
Flegeau K, Rubin S, Mucha S, Bur P, Préterre J, Siadous R, L'Azou B, Fricain JC, Combe C, Devillard R, Kalisky J, Rigothier C. Towards an in vitro model of the glomerular barrier unit with an innovative bioassembly method. Nephrol Dial Transplant 2020; 35:240-250. [PMID: 31121032 DOI: 10.1093/ndt/gfz094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The development of an artificial glomerular unit may be pivotal for renal pathophysiology studies at a multicellular scale. Using a tissue engineering approach, we aimed to reproduce in part the specific glomerular barrier architecture by manufacturing a glomerular microfibre (Mf). METHODS Immortalized human glomerular cell lines of endothelial cells (GEnCs) and podocytes were used. Cells and a three-dimensional (3D) matrix were characterized by immunofluorescence with confocal analysis, Western blot and polymerase chain reaction. Optical and electron microscopy were used to study Mf and cell shapes. We also analysed cell viability and cell metabolism within the 3D construct at 14 days. RESULTS Using the Mf manufacturing method, we repeatedly obtained a cellularized Mf sorting human glomerular cells in 3D. Around a central structure made of collagen I, we obtained an internal layer composed of GEnC, a newly formed glomerular basement membrane rich in α5 collagen IV and an external layer of podocytes. The cell concentration, optimal seeding time and role of physical stresses were modulated to obtain the Mf. Cell viability and expression of specific proteins (nephrin, synaptopodin, vascular endothelial growth factor receptor 2 (VEGFR2) and von Willebrandt factor (vWF)) were maintained for 19 days in the Mf system. Mf ultrastructure, observed with EM, had similarities with the human glomerular barrier. CONCLUSION In summary, with our 3D bio-engineered glomerular fibre, GEnC and podocytes produced a glomerular basement membrane. In the future, this glomerular Mf will allow us to study cell interactions in a 3D system and increase our knowledge of glomerular pathophysiology.
Collapse
Affiliation(s)
- Killian Flegeau
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Sébastien Rubin
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Simon Mucha
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Pauline Bur
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Julie Préterre
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Robin Siadous
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Béatrice L'Azou
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Jean-Christophe Fricain
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service d'odontologie et de Santé Buccale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Christian Combe
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Raphaël Devillard
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service d'odontologie et de Santé Buccale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Jérôme Kalisky
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Claire Rigothier
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| |
Collapse
|
38
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
39
|
Blaine J, Dylewski J. Regulation of the Actin Cytoskeleton in Podocytes. Cells 2020; 9:cells9071700. [PMID: 32708597 PMCID: PMC7408282 DOI: 10.3390/cells9071700] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Podocytes are an integral part of the glomerular filtration barrier, a structure that prevents filtration of large proteins and macromolecules into the urine. Podocyte function is dependent on actin cytoskeleton regulation within the foot processes, structures that link podocytes to the glomerular basement membrane. Actin cytoskeleton dynamics in podocyte foot processes are complex and regulated by multiple proteins and other factors. There are two key signal integration and structural hubs within foot processes that regulate the actin cytoskeleton: the slit diaphragm and focal adhesions. Both modulate actin filament extension as well as foot process mobility. No matter what the initial cause, the final common pathway of podocyte damage is dysregulation of the actin cytoskeleton leading to foot process retraction and proteinuria. Disruption of the actin cytoskeleton can be due to acquired causes or to genetic mutations in key actin regulatory and signaling proteins. Here, we describe the major structural and signaling components that regulate the actin cytoskeleton in podocytes as well as acquired and genetic causes of actin dysregulation.
Collapse
Affiliation(s)
- Judith Blaine
- Renal Division, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - James Dylewski
- Renal Division, University of Colorado Anschutz Medical Campus and Denver Health Medical Center, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +303-724-4841
| |
Collapse
|
40
|
Srivastava T, Joshi T, Jiang Y, Heruth DP, Rezaiekhaligh MH, Novak J, Staggs VS, Alon US, Garola RE, El-Meanawy A, McCarthy ET, Zhou J, Boinpelly VC, Sharma R, Savin VJ, Sharma M. Upregulated proteoglycan-related signaling pathways in fluid flow shear stress-treated podocytes. Am J Physiol Renal Physiol 2020; 319:F312-F322. [PMID: 32628542 DOI: 10.1152/ajprenal.00183.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The ultrafiltrate flow over the major processes and cell body generates fluid flow shear stress (FFSS) on podocytes. Hyperfiltration-associated increase in FFSS can lead to podocyte injury and detachment. Previously, we showed that FFSS-induced upregulation of the cyclooxygenase 2 (COX2)-PGE2-prostaglandin E receptor 2 (EP2) axis in podocytes activates Akt-glycogen synthase kinase-3β-β-catenin and MAPK/ERK signaling in response to FFSS. Integrative MultiOmics Pathway Resolution (IMPRes) is a new bioinformatic tool that enables simultaneous time-series analysis of more than two groups to identify pathways and molecular connections. In the present study, we used previously characterized COX2 [prostaglandin-endoperoxide synthase 2 (Ptgs2)], EP2 (Ptger2), and β1-catenin (Ctnnb1) as "seed genes" from an array data set of four groups analyzed over a time course. The 3 seed genes shared 7 pathways and 50 genes of 14 pathways and 89 genes identified by IMPRes. A composite of signaling pathways highlighted the temporal molecular connections during mechanotransduction signaling in FFSS-treated podocytes. We investigated the "proteoglycans in cancer" and "galactose metabolism" pathways predicted by IMPRes. A custom-designed PCR array validated 60.7% of the genes predicted by IMPRes analysis, including genes for the above-named pathways. Further validation using Western blot analysis showed increased expression of phosho-Erbb2, phospho-mammalian target of rapamycin (mTOR), CD44, and hexokinase II (Hk2); decreased total Erbb2, galactose mutarotase (Galm), and β-1,4-galactosyltransferase 1 (B4galt1); and unchanged total mTOR and AKT3. These findings corroborate our previously reported results. This study demonstrates the potential of the IMPRes method to identify novel pathways. Identifying the "proteoglycans in cancer" and "galactose metabolism" pathways has generated a lead to study the significance of FFSS-induced glycocalyx remodeling and possible detachment of podocytes from the glomerular matrix.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri.,Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Department of Oral and Craniofacial Sciences, University of Missouri School of Dentistry, Kansas City, Missouri
| | - Trupti Joshi
- Department of Health Management and Informatics and University of Missouri Informatics Institute, University of Missouri, Columbia, Missouri.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,MU Data Science and Informatics Institute, University of Missouri, Columbia, Missouri
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Mohamed H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Jan Novak
- Department of Microbiology, University of Alabama, Birmingham, Alabama
| | - Vincent S Staggs
- Biostatistics and Epidemiology Core, Children's Mercy Kansas City, Department of Pediatrics, University of Missouri, Kansas City, Missouri
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri, Kansas City
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ellen T McCarthy
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Ram Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Virginia J Savin
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
41
|
Xie R, Korolj A, Liu C, Song X, Lu RXZ, Zhang B, Ramachandran A, Liang Q, Radisic M. h-FIBER: Microfluidic Topographical Hollow Fiber for Studies of Glomerular Filtration Barrier. ACS CENTRAL SCIENCE 2020; 6:903-912. [PMID: 32607437 PMCID: PMC7318083 DOI: 10.1021/acscentsci.9b01097] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Indexed: 05/07/2023]
Abstract
Kidney-on-a-chip devices may revolutionize the discovery of new therapies. However, fabricating a 3D glomerulus remains a challenge, due to a requirement for a microscale soft material with complex topography to support cell culture in a native configuration. Here, we describe the use of microfluidic spinning to recapitulate complex concave and convex topographies over multiple length scales, required for biofabrication of a biomimetic 3D glomerulus. We produced a microfluidic extruded topographic hollow fiber (h-FIBER), consisting of a vessel-like perfusable tubular channel for endothelial cell cultivation, and a glomerulus-like knot with microconvex topography on its surface for podocyte cultivation. Meter long h-FIBERs were produced in microfluidics within minutes, followed by chemically induced inflation for generation of topographical cues on the 3D scaffold surface. The h-FIBERs were assembled into a hot-embossed plastic 96-well plate. Long-term perfusion, podocyte barrier formation, endothelialization, and permeability tests were easily performed by a standard pipetting technique on the platform. Following long-term culture (1 month), a functional filtration barrier, measured by the transfer of albumin from the blood vessel side to the ultrafiltrate side, suggested the establishment of an engineered glomerulus.
Collapse
Affiliation(s)
- Ruoxiao Xie
- MOE
Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology,
Beijing Key Lab of Microanalytical Methods & Instrumentation,
Department of Chemistry, Centre for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, P. R. China
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Anastasia Korolj
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Chuan Liu
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Xin Song
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Rick Xing Ze Lu
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Boyang Zhang
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Arun Ramachandran
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Qionglin Liang
- MOE
Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology,
Beijing Key Lab of Microanalytical Methods & Instrumentation,
Department of Chemistry, Centre for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, P. R. China
| | - Milica Radisic
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| |
Collapse
|
42
|
Shiva N, Sharma N, Kulkarni YA, Mulay SR, Gaikwad AB. Renal ischemia/reperfusion injury: An insight on in vitro and in vivo models. Life Sci 2020; 256:117860. [PMID: 32534037 DOI: 10.1016/j.lfs.2020.117860] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023]
Abstract
Optimal tissue oxygenation is essential for its normal function. Suboptimal oxygenation or ischemia contributes to increased mortalities during various pathological conditions such as stroke, acute kidney injury (AKI), cardiac failure. Despite the rapid progression of renal tissue injury, the mechanism underlying renal ischemia/reperfusion injury (IRI) remains highly unclear. Experimental in vitro and in vivo models epitomizing the fundamental process is critical to the research of the pathogenesis of IRI and the development of plausible therapeutics. In this review, we describe the in vitro and in vivo models of IRI, ranges from proximal tubular cell lines to surgery-based animal models like clamping of both renal pedicles (bilateral IRI), clamping of one renal pedicle (unilateral IRI), clamping of one/or both renal arteries/or vein, or unilateral IRI with contralateral nephrectomy (uIRIx). Also, advanced technologies like three-dimensional kidney organoids, kidney-on-a-chip are explained. This review provides thoughtful information for establishing reliable and pertinent models for studying IRI-associated acute renal pathologies.
Collapse
Affiliation(s)
- Niharika Shiva
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Shrikant R Mulay
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
43
|
Concerted EP2 and EP4 Receptor Signaling Stimulates Autocrine Prostaglandin E 2 Activation in Human Podocytes. Cells 2020; 9:cells9051256. [PMID: 32438662 PMCID: PMC7290667 DOI: 10.3390/cells9051256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/29/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Glomerular hyperfiltration is an important mechanism in the development of albuminuria. During hyperfiltration, podocytes are exposed to increased fluid flow shear stress (FFSS) in Bowman’s space. Elevated Prostaglandin E2 (PGE2) synthesis and upregulated cyclooxygenase 2 (Cox2) are associated with podocyte injury by FFSS. We aimed to elucidate a PGE2 autocrine/paracrine pathway in human podocytes (hPC). We developed a modified liquid chromatography tandem mass spectrometry (LC/ESI-MS/MS) protocol to quantify cellular PGE2, 15-keto-PGE2, and 13,14-dihydro-15-keto-PGE2 levels. hPC were treated with PGE2 with or without separate or combined blockade of prostaglandin E receptors (EP), EP2, and EP4. Furthermore, the effect of FFSS on COX2, PTGER2, and PTGER4 expression in hPC was quantified. In hPC, stimulation with PGE2 led to an EP2- and EP4-dependent increase in cyclic adenosine monophosphate (cAMP) and COX2, and induced cellular PGE2. PTGER4 was downregulated after PGE2 stimulation in hPC. In the corresponding LC/ESI-MS/MS in vivo analysis at the tissue level, increased PGE2 and 15-keto-PGE2 levels were observed in isolated glomeruli obtained from a well-established rat model with glomerular hyperfiltration, the Munich Wistar Frömter rat. COX2 and PTGER2 were upregulated by FFSS. Our data thus support an autocrine/paracrine COX2/PGE2 pathway in hPC linked to concerted EP2 and EP4 signaling.
Collapse
|
44
|
Mechanisms of Synergistic Interactions of Diabetes and Hypertension in Chronic Kidney Disease: Role of Mitochondrial Dysfunction and ER Stress. Curr Hypertens Rep 2020; 22:15. [PMID: 32016622 DOI: 10.1007/s11906-020-1016-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To discuss the importance of synergistic interactions of diabetes mellitus (DM) and hypertension (HT) in causing chronic kidney disease and the potential molecular mechanisms involved. RECENT FINDINGS DM and HT are the two most important risk factors for chronic kidney disease (CKD) and development of end-stage renal disease (ESRD). The combination of HT and DM may synergistically promote the progression of renal injury through mechanisms that have not been fully elucidated. Hyperglycemia and other metabolic changes in DM initiate endoplasmic reticulum (ER) stress and mitochondrial (MT) adaptation in different types of glomerular cells. These adaptations appear to make the cells more vulnerable to HT-induced mechanical stress. Excessive activation of mechanosensors, possibly via transient receptor potential cation channel subfamily C member 6 (TRPC6), may lead to impaired calcium (Ca2+) homeostasis and further exacerbate ER stress and MT dysfunction promoting cellular apoptosis and glomerular injury. The synergistic effects of HT and DM to promote kidney injury may be mediated by increased intraglomerular pressure. Chronic activation of mechanotransduction signaling may amplify metabolic effects of DM causing cellular injury through a vicious cycle of impaired Ca2+ homeostasis, mitochondrial dysfunction, and ER stress.
Collapse
|
45
|
Kriz W. The Inability of Podocytes to Proliferate: Cause, Consequences, and Origin. Anat Rec (Hoboken) 2019; 303:2588-2596. [PMID: 31606944 DOI: 10.1002/ar.24291] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 12/24/2022]
Abstract
This study presents a theoretical analysis of the problems related to the inability of podocytes to proliferate. The basis of these problems is the very high rate of glomerular filtration. Podocytes do not in general die by apoptosis or necrosis but are lost by detachment from the glomerular basement membrane (GBM) as viable cells. Podocytes situated on the outside of the filtration barrier and attached to the GBM only by their foot processes are permanently exposed to the flow dynamic forces of the high filtration rate tending to detach them from the GBM. The major challenge seems to consist of the high shear stresses on the foot processes within the filtration slits due to filtrate flow. Healthy podocytes are able to resist this challenge, injured podocytes are not, and may undergo foot process detachment, leading to a gap in the podocyte cover of the GBM. This represents a mortal event. Like a dam break, such a leak cannot be repaired. The ongoing exposure to filtrate flow prevents any attempt to close the gap, thus preventing any regeneration including cell proliferation. An improvement of this precarious situation consists of healing by scarring that may involve only one lobule of the glomerulus, permitting the remaining lobules to maintain filtration. An answer to the question of which waste product requires such a high filtration rate for its excretion may be in the huge quantity of circulating peptides, a problem that dates far back in evolution.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Department of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
46
|
Abstract
Finding new therapeutic targets of glomerulosclerosis treatment is an ongoing quest. Due to a living environment of various stresses and pathological stimuli, podocytes are prone to injuries; moreover, as a cell without proliferative potential, loss of podocytes is vital in the pathogenesis of glomerulosclerosis. Thus, sufficient understanding of factors and underlying mechanisms of podocyte injury facilitates the advancement of treating and prevention of glomerulosclerosis. The clinical symptom of podocyte injury is proteinuria, sometimes with loss of kidney functions progressing to glomerulosclerosis. Injury-induced changes in podocyte physiology and function are actually not a simple passive process, but a complex interaction of proteins that comprise the anatomical structure of podocytes at molecular levels. This chapter lists several aspects of podocyte injuries along with potential mechanisms, including glucose and lipid metabolism disorder, hypertension, RAS activation, micro-inflammation, immune disorder, and other factors. These aspects are not technically separated items, but intertwined with each other in the pathogenesis of podocyte injuries.
Collapse
|
47
|
Abstract
Kidney donors face a small but definite risk of end-stage renal disease 15 to 30 years postdonation. The development of proteinuria, hypertension with gradual decrease in kidney function in the donor after surgical resection of 1 kidney, has been attributed to hyperfiltration. Genetic variations, physiological adaptations, and comorbidities exacerbate the hyperfiltration-induced loss of kidney function in the years after donation. A focus on glomerular hemodynamics and capillary pressure has led to the development of drugs that target the renin-angiotensin-aldosterone system (RAAS), but these agents yield mixed results in transplant recipients and donors. Recent work on glomerular biomechanical forces highlights the differential effects of tensile stress and fluid flow shear stress (FFSS) from hyperfiltration. Capillary wall stretch due to glomerular capillary pressure increases tensile stress on podocyte foot processes that cover the capillary. In parallel, increased flow of the ultrafiltrate due to single-nephron glomerular filtration rate elevates FFSS on the podocyte cell body. Although tensile stress invokes the RAAS, FFSS predominantly activates the cyclooxygenase 2-prostaglandin E2-EP2 receptor axis. Distinguishing these 2 mechanisms is critical, as current therapeutic approaches focus on the RAAS system. A better understanding of the biomechanical forces can lead to novel therapeutic agents to target FFSS through the cyclooxygenase 2-prostaglandin E2-EP2 receptor axis in hyperfiltration-mediated injury. We present an overview of several aspects of the risk to transplant donors and discuss the relevance of FFSS in podocyte injury, loss of glomerular barrier function leading to albuminuria and gradual loss of renal function, and potential therapeutic strategies to mitigate hyperfiltration-mediated injury to the remaining kidney.
Collapse
|
48
|
Tsuda A, Ishimura E, Uedono H, Ochi A, Nakatani S, Morioka T, Mori K, Uchida J, Emoto M, Nakatani T, Inaba M. Association of Albuminuria With Intraglomerular Hydrostatic Pressure and Insulin Resistance in Subjects With Impaired Fasting Glucose and/or Impaired Glucose Tolerance. Diabetes Care 2018; 41:2414-2420. [PMID: 30217931 DOI: 10.2337/dc18-0718] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/06/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Little is known about the relationships between insulin resistance, intrarenal hemodynamics, and urinary albumin excretion (UAE) in humans with impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT). The aim of the current study was to examine intrarenal hemodynamic abnormalities, insulin resistance, and UAE in subjects with IFG or IGT. We hypothesized that intrarenal hemodynamic abnormalities would be associated with insulin resistance. RESEARCH DESIGN AND METHODS Fifty-four kidney donors underwent 75-g oral glucose tolerance and inulin and para-aminohippuric acid clearance testing. Insulin sensitivity index (ISI) was evaluated by the Matsuda index. Intrarenal hemodynamic parameters were calculated by the Gomez formulae. RESULTS Of the 54 subjects, 33 exhibited IFG or IGT and 31 exhibited normal glucose tolerance (NGT). Glomerular hydrostatic pressure (Pglo) and UAE were significantly higher in the IFG or IGT subjects with obesity (P = 0.015 and 0.0001, respectively). Log ISI correlated significantly and negatively with Pglo (r = -0.351, P = 0.009) in all subjects. In multiple regression analyses among all subjects, log ISI was associated significantly and independently with Pglo (β = -0.316, P = 0.015), after adjustment for age, sex, and systolic blood pressure. Further, BMI (β = 0.517, P = 0.0004), Pglo (β = 0.420, P = 0.004), and log ISI (β = -0.366, P = 0.008) were each associated significantly and independently with UAE after adjustment. CONCLUSIONS We demonstrated that increased insulin resistance is associated with increased Pglo and UAE in IFG or IGT subjects. These hemodynamic burdens and insulin resistance may cause injury to the glomeruli even in subjects with IFG or IGT.
Collapse
Affiliation(s)
- Akihiro Tsuda
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Eiji Ishimura
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hideki Uedono
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akinobu Ochi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsuhito Mori
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Junji Uchida
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masanori Emoto
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsuya Nakatani
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaaki Inaba
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
49
|
Artelt N, Ludwig TA, Rogge H, Kavvadas P, Siegerist F, Blumenthal A, van den Brandt J, Otey CA, Bang ML, Amann K, Chadjichristos CE, Chatziantoniou C, Endlich K, Endlich N. The Role of Palladin in Podocytes. J Am Soc Nephrol 2018; 29:1662-1678. [PMID: 29720549 DOI: 10.1681/asn.2017091039] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 03/28/2018] [Indexed: 11/03/2022] Open
Abstract
Background Podocyte loss and effacement of interdigitating podocyte foot processes are the major cause of a leaky filtration barrier and ESRD. Because the complex three-dimensional morphology of podocytes depends on the actin cytoskeleton, we studied the role in podocytes of the actin bundling protein palladin, which is highly expressed therein.Methods We knocked down palladin in cultured podocytes by siRNA transfection or in zebrafish embryos by morpholino injection and studied the effects by immunofluorescence and live imaging. We also investigated kidneys of mice with podocyte-specific knockout of palladin (PodoPalld-/- mice) by immunofluorescence and ultrastructural analysis and kidney biopsy specimens from patients by immunostaining for palladin.Results Compared with control-treated podocytes, palladin-knockdown podocytes had reduced actin filament staining, smaller focal adhesions, and downregulation of the podocyte-specific proteins synaptopodin and α-actinin-4. Furthermore, palladin-knockdown podocytes were more susceptible to disruption of the actin cytoskeleton with cytochalasin D, latrunculin A, or jasplakinolide and showed altered migration dynamics. In zebrafish embryos, palladin knockdown compromised the morphology and dynamics of epithelial cells at an early developmental stage. Compared with PodoPalld+/+ controls, PodoPalld-/- mice developed glomeruli with a disturbed morphology, an enlarged subpodocyte space, mild effacement, and significantly reduced expression of nephrin and vinculin. Furthermore, nephrotoxic serum injection led to significantly higher levels of proteinuria in PodoPalld-/- mice than in controls. Kidney biopsy specimens from patients with diabetic nephropathy and FSGS showed downregulation of palladin in podocytes as well.Conclusions Palladin has an important role in podocyte function in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | - Panagiotis Kavvadas
- National Institute for Health and Medical Research (INSERM), Unité Mixte de Recherche (UMR)-S1155, Tenon Hospital, Sorbonne Universités, Paris, France
| | | | | | - Jens van den Brandt
- Central Core and Research Facility of Laboratory Animals (ZSFV), University Medicine Greifswald, Greifswald, Germany
| | - Carol A Otey
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina
| | - Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Milan, Italy.,Humanitas Clinical and Research Center, Rozzano, Milan, Italy; and
| | - Kerstin Amann
- Department of Nephropathology, University Medicine Erlangen, Erlangen, Germany
| | - Christos E Chadjichristos
- National Institute for Health and Medical Research (INSERM), Unité Mixte de Recherche (UMR)-S1155, Tenon Hospital, Sorbonne Universités, Paris, France
| | - Christos Chatziantoniou
- National Institute for Health and Medical Research (INSERM), Unité Mixte de Recherche (UMR)-S1155, Tenon Hospital, Sorbonne Universités, Paris, France
| | | | | |
Collapse
|
50
|
Kriz W, Lemley KV. Potential relevance of shear stress for slit diaphragm and podocyte function. Kidney Int 2018; 91:1283-1286. [PMID: 28501303 DOI: 10.1016/j.kint.2017.02.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/07/2017] [Accepted: 02/24/2017] [Indexed: 11/30/2022]
Abstract
Filtrate flow through the glomerular barrier produces shear stresses that tend to disconnect podocytes from the glomerular basement membrane. Forces are highest within the filtration slits. The slit diaphragm mechanically balances the lateral components of the shear stresses on opposing foot processes, preventing widening of the slit.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Department of Neuroanatomy, Medical Faculty Mannheim, Germany.
| | - Kevin V Lemley
- Division of Nephrology, Children's Hospital Los Angeles, California, USA
| |
Collapse
|