1
|
Antonowicz B, Błachnio-Zabielska A, Chlabicz U, Maciejczyk M, Borys J, Łukaszuk K, Zięba S, Lo Giudice R, Lo Giudice G, Szuta M, Zalewska A. Profile of the Main Representatives of Sphingolipid Metabolism in the Maxillary and Mandibular Periosteum of Patients with Dentofacial Deformities After Osteosynthesis Using Titanium Implants. J Clin Med 2025; 14:1929. [PMID: 40142736 PMCID: PMC11943205 DOI: 10.3390/jcm14061929] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: The objective of this study was to analyze the profiles of sphingosine (Sph), sphinganine (SPA), sphingosine-1-phosphate (S1P), and ceramides (C14 Cer, C16 Cer, C18:1 Cer, C18 Cer, C20 Cer, C22 Cer, C24:1 Cer, and C24 Cer), along with caspases (CAS-3, CAS-6, and CAS-9), in serum and in the periosteum of the maxilla and mandible in patients with dentofacial deformities undergoing treatment with titanium fixations (miniplates and miniscrews). Methods: The study group comprised 20 patients who underwent bilateral jaw osteotomy due to dentofacial deformities. The osteotomy segments were stabilized with titanium alloy miniplates and screws. The control group consisted of 20 patients who had not yet received surgical treatment for maxillofacial defects. Results: Sphinganine (SPA) and ceramide C22 (C22 Cer) were the only compounds found to be significantly elevated in the serum of the study group compared to the control group. The concentrations of Sph, SPA, C14 Cer, C16 Cer, C18 1 Cer, C18 Cer, C22 Cer, C24 1 Cer, C24 Cer, and S1P were significantly lower in the maxillary periosteum of patients in the study group compared to those in the control group. The concentration of C20 Cer was significantly higher in the maxillary periosteum of patients in the study group compared to the control group. In contrast, the concentrations of Sph, SPA, C14 Cer, C16 Cer, C18 1 Cer, C22 Cer, C24 1 Cer, and C24 Cer were significantly lower in the mandibular periosteum of the study group compared to the control group. The concentrations of C20 Cer and S1P were significantly elevated in the mandibular periosteum of patients in the study group compared with the control group. The activity of CAS-3 was significantly higher in the mandibular periosteum of patients in the study group compared to those in the control group. Conclusions: Titanium fixations induce local changes in the sphingolipid profile within the periosteum of the maxilla and mandible, while no systemic impact on this metabolism was observed.
Collapse
Affiliation(s)
- Bożena Antonowicz
- Department of Dental Surgery, Medical University in Bialystok, 15-089 Białystok, Poland
| | - Agnieszka Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 15-089 Białystok, Poland; (A.B.-Z.); (U.C.)
| | - Urszula Chlabicz
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 15-089 Białystok, Poland; (A.B.-Z.); (U.C.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Jan Borys
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Kamila Łukaszuk
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Sara Zięba
- Department of Restorative Dentistry, Medical University of Bialystok, 15-089 Białystok, Poland (A.Z.)
| | - Roberto Lo Giudice
- Department of Human Pathology of the Adult and Evolutive Age G. Barresi, Messina University, 98100 Messina, Italy;
| | - Giuseppe Lo Giudice
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy;
| | - Mariusz Szuta
- Department of Oral Surgery, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Anna Zalewska
- Department of Restorative Dentistry, Medical University of Bialystok, 15-089 Białystok, Poland (A.Z.)
- Independent Laboratory of Experimental Dentistry, Medical University of Bialystok, 15-089 Białystok, Poland
| |
Collapse
|
2
|
Seyedaghamiri F, Salimi L, Ghaznavi D, Sokullu E, Rahbarghazi R. Exosomes-based therapy of stroke, an emerging approach toward recovery. Cell Commun Signal 2022; 20:110. [PMID: 35869548 PMCID: PMC9308232 DOI: 10.1186/s12964-022-00919-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/11/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractBased on clinical observations, stroke is touted as one of the specific pathological conditions, affecting an individual’s life worldwide. So far, no effective treatment has been introduced to deal with stroke post-complications. Production and release of several neurotrophic factors by different cells exert positive effects on ischemic areas following stroke. As a correlate, basic and clinical studies have focused on the development and discovery of de novo modalities to introduce these factors timely and in appropriate doses into the affected areas. Exosomes (Exo) are non-sized vesicles released from many cells during pathological and physiological conditions and participate in intercellular communication. These particles transfer several arrays of signaling molecules, like several neurotrophic factors into the acceptor cells and induce specific signaling cascades in the favor of cell bioactivity. This review aimed to highlight the emerging role of exosomes as a therapeutic approach in the regeneration of ischemic areas.
Collapse
|
3
|
Du H, Xu Y, Zhu L. Role of Semaphorins in Ischemic Stroke. Front Mol Neurosci 2022; 15:848506. [PMID: 35350431 PMCID: PMC8957939 DOI: 10.3389/fnmol.2022.848506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is one of the major causes of neurological morbidity and mortality in the world. Although the management of ischemic stroke has been improved significantly, it still imposes a huge burden on the health and property. The integrity of the neurovascular unit (NVU) is closely related with the prognosis of ischemic stroke. Growing evidence has shown that semaphorins, a family of axon guidance cues, play a pivotal role in multiple pathophysiological processes in NVU after ischemia, such as regulating the immune system, angiogenesis, and neuroprotection. Modulating the NVU function via semaphorin signaling has a potential to develop a novel therapeutic strategy for ischemic stroke. We, therefore, review recent progresses on the role of semphorin family members in neurons, glial cells and vasculature after ischemic stroke.
Collapse
Affiliation(s)
- Huaping Du
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Yuan Xu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Li Zhu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Collaborative Innovation Center of Hematology of Jiangsu Province, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- *Correspondence: Li Zhu,
| |
Collapse
|
4
|
Lay AC, Hale LJ, Stowell-Connolly H, Pope RJP, Nair V, Ju W, Marquez E, Rollason R, Hurcombe JA, Hayes B, Roberts T, Gillam L, Allington J, Nelson RG, Kretzler M, Holly JMP, Perks CM, McArdle CA, Welsh GI, Coward RJM. IGFBP-1 expression is reduced in human type 2 diabetic glomeruli and modulates β1-integrin/FAK signalling in human podocytes. Diabetologia 2021; 64:1690-1702. [PMID: 33758952 PMCID: PMC8187213 DOI: 10.1007/s00125-021-05427-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/14/2021] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Podocyte loss or injury is one of the earliest features observed in the pathogenesis of diabetic kidney disease (DKD), which is the leading cause of end-stage renal failure worldwide. Dysfunction in the IGF axis, including in IGF binding proteins (IGFBPs), is associated with DKD, particularly in the early stages of disease progression. The aim of this study was to investigate the potential roles of IGFBPs in the development of type 2 DKD, focusing on podocytes. METHODS IGFBP expression was analysed in the Pima DKD cohort, alongside data from the Nephroseq database, and in ex vivo human glomeruli. Conditionally immortalised human podocytes and glomerular endothelial cells were studied in vitro, where IGFBP-1 expression was analysed using quantitative PCR and ELISAs. Cell responses to IGFBPs were investigated using migration, cell survival and adhesion assays; electrical cell-substrate impedance sensing; western blotting; and high-content automated imaging. RESULTS Data from the Pima DKD cohort and from the Nephroseq database demonstrated a significant reduction in glomerular IGFBP-1 in the early stages of human type 2 DKD. In the glomerulus, IGFBP-1 was predominantly expressed in podocytes and controlled by phosphoinositide 3-kinase (PI3K)-forkhead box O1 (FoxO1) activity. In vitro, IGFBP-1 signalled to podocytes via β1-integrins, resulting in increased phosphorylation of focal-adhesion kinase (FAK), increasing podocyte motility, adhesion, electrical resistance across the adhesive cell layer and cell viability. CONCLUSIONS/INTERPRETATION This work identifies a novel role for IGFBP-1 in the regulation of podocyte function and that the glomerular expression of IGFBP-1 is reduced in the early stages of type 2 DKD, via reduced FoxO1 activity. Thus, we hypothesise that strategies to maintain glomerular IGFBP-1 levels may be beneficial in maintaining podocyte function early in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lorna J Hale
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Robert J P Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Eva Marquez
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ruth Rollason
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jenny A Hurcombe
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Bryony Hayes
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Timothy Roberts
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lawrence Gillam
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jonathan Allington
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jeff M P Holly
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire M Perks
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Craig A McArdle
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
5
|
Kaminski A, Gupta KH, Goldufsky JW, Lee HW, Gupta V, Shafikhani SH. Pseudomonas aeruginosa ExoS Induces Intrinsic Apoptosis in Target Host Cells in a Manner That is Dependent on its GAP Domain Activity. Sci Rep 2018; 8:14047. [PMID: 30232373 PMCID: PMC6145893 DOI: 10.1038/s41598-018-32491-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/03/2018] [Indexed: 11/08/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that causes serious infections in immunocompromised individuals and cystic fibrosis patients. ExoS and ExoT are two homologous bifunctional Type III Secretion System (T3SS) virulence factors that induce apoptosis in target host cells. They possess a GTPase Activating Protein (GAP) domain at their N-termini, which share ~76% homology, and an ADP-ribosyltransferase (ADPRT) domain at their C-termini, which target non-overlapping substrates. Both the GAP and the ADPRT domains contribute to ExoT's cytotoxicity in target epithelial cells, whereas, ExoS-induced apoptosis is reported to be primarily due to its ADPRT domain. In this report, we demonstrate that ExoS/GAP domain is both necessary and sufficient to induce mitochondrial apoptosis. Our data demonstrate that intoxication with ExoS/GAP domain leads to enrichment of Bax and Bim into the mitochondrial outer-membrane, disruption of mitochondrial membrane and release of and cytochrome c into the cytosol, which activates initiator caspase-9 and effector caspase-3, that executes cellular death. We posit that the contribution of the GAP domain in ExoS-induced apoptosis was overlooked in prior studies due to its slower kinetics of cytotoxicity as compared to ADPRT. Our data clarify the field and reveal a novel virulence function for ExoS/GAP as an inducer of apoptosis.
Collapse
Affiliation(s)
- Amber Kaminski
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Kajal H Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Josef W Goldufsky
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Ha Won Lee
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vineet Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sasha H Shafikhani
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA.
- Cancer Center, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
6
|
Shen H, Yao X, Li H, Li X, Zhang T, Sun Q, Ji C, Chen G. Role of Exosomes Derived from miR-133b Modified MSCs in an Experimental Rat Model of Intracerebral Hemorrhage. J Mol Neurosci 2018; 64:421-430. [PMID: 29455449 DOI: 10.1007/s12031-018-1041-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhage (ICH) has poor outcomes due to high mortality and morbidity, but until now, the effective treatments remain limited. MicroRNAs (miRNAs) are vital regulators of gene expression and demonstrated to be linked to the pathogenesis of various central nervous system (CNS) diseases. Exosomes are considered as cell-to-cell communication vectors and secreted largely by mesenchymal stromal cells (MSCs). The present study investigated the role of miR-133b delivered by exosomes secreted from MSCs to brain tissues in rats after ICH. An autologous arterial blood ICH model in adult male Sprague-Dawley (SD) rats was used in this study. At 72 h after transfection with miR-133b mimics in MSCs, miR-133b-modified MSC-derived exosomes were collected from medium of MSCs and then injected to rats via tail vein. The levels of miR-133b in secreted exosomes and brain tissues of rats in various groups and the levels of RhoA, phosphorylations of extracellular signal regulating kinase (ERK1/2), and cAMP response element-binding protein (CREB) were detected by real-time PCR and western blot analysis, respectively. The effects of miR-133b on neuronal apoptosis and degeneration were respectively evaluated by TUNEL and fluoro-jade B staining. The miR-133b levels were reduced in brain tissues of rats at 24 h and peaked at 72 h after ICH. At 24 h after miR-133b-modified exosome administration, the level of miR-133b was significantly increased, while the apoptotic and neurodegenerative neurons were obviously reduced in brain tissues after ICH. The results of western blot analysis showed that miR-133b modified exosomes treatment remarkably suppressed RhoA expression and activated ERK1/2/CREB in brain tissues after ICH. Collectively, our investigation suggested that exosomes derived from miR-133b modified MSCs exhibited neuroprotective role for anti-apoptotic effect of miR-133b mediating RhoA and ERK1/2/CREB in rats after ICH.
Collapse
Affiliation(s)
- Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Tiejun Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Chengyuan Ji
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| |
Collapse
|
7
|
RhoA inhibits the hypoxia-induced apoptosis and mitochondrial dysfunction in chondrocytes via positively regulating the CREB phosphorylation. Biosci Rep 2017; 37:BSR20160622. [PMID: 28254846 PMCID: PMC5398256 DOI: 10.1042/bsr20160622] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 11/17/2022] Open
Abstract
Chondrocytes that are embedded within the growth plate or the intervertebral disc are sensitive to environmental stresses, such as inflammation and hypoxia. However, little is known about the molecular signalling pathways underlining the hypoxia-induced mitochondrial dysfunction and apoptosis in chondrocytes. In the present study, we firstly examined the hypoxia-induced apoptosis, mitochondrial dysfunction and the activation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) signalling in human chondrocyte cell line, C28/I2 and then investigated the regulatory role of RhoA, a well-recognized apoptosis suppressor, in such process, with gain-of-function strategy. Our results indicated that hypoxia induced apoptosis and inhibited CREB phosphprylation in chondrocytes, meanwhile, the dysfunctional mitochondria with up-regulated mitochondrial superoxide and reactive oxygen species (ROS) levels, whereas with a reduced mitochondrial membrane potential (MMP) and Complex IV activity were observed in the hypoxia-treated C28/I2 cells. However, the overexpressed RhoA blocked the hypoxia-mediated reduction in CREB phosphprylation and inhibited the apoptosis induction, along with an ameliorated mitochondrial function in the hypoxia-treated C28/I2 cells. In conclusion, the present study confirmed the reduced CREB phosphorylation, along with the apoptosis induction and mitochondrial dysfunction in the hypoxia-treated chondrocyte cells. And the overexpression of RhoA ameliorated the hypoxia-induced mitochondrial dysfunction and apoptosis via blocking the hypoxia-mediated reduction in CREB phosphorylation.
Collapse
|
8
|
Bartram MP, Habbig S, Pahmeyer C, Höhne M, Weber LT, Thiele H, Altmüller J, Kottoor N, Wenzel A, Krueger M, Schermer B, Benzing T, Rinschen MM, Beck BB. Three-layered proteomic characterization of a novel ACTN4 mutation unravels its pathogenic potential in FSGS. Hum Mol Genet 2016; 25:1152-64. [PMID: 26740551 DOI: 10.1093/hmg/ddv638] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 12/31/2015] [Indexed: 01/09/2023] Open
Abstract
Genetic diseases constitute the most important cause for end-stage renal disease in children and adolescents. Mutations in the ACTN4 gene, encoding the actin-binding protein α-actinin-4, are a rare cause of autosomal dominant familial focal segmental glomerulosclerosis (FSGS). Here, we report the identification of a novel, disease-causing ACTN4 mutation (p.G195D, de novo) in a sporadic case of childhood FSGS using next generation sequencing. Proteome analysis by quantitative mass spectrometry (MS) of patient-derived urinary epithelial cells indicated that ACTN4 levels were significantly decreased when compared with healthy controls. By resolving the peptide bearing the mutated residue, we could proof that the mutant protein is less abundant when compared with the wild-type protein. Further analyses revealed that the decreased stability of p.G195D is associated with increased ubiquitylation in the vicinity of the mutation site. We next defined the ACTN4 interactome, which was predominantly composed of cytoskeletal modulators and LIM domain-containing proteins. Interestingly, this entire group of proteins, including several highly specific ACTN4 interactors, was globally decreased in the patient-derived cells. Taken together, these data suggest a mechanistic link between ACTN4 instability and proteome perturbations of the ACTN4 interactome. Our findings advance the understanding of dominant effects exerted by ACTN4 mutations in FSGS. This study illustrates the potential of genomics and complementary, high-resolution proteomics analyses to study the pathogenicity of rare gene variants.
Collapse
Affiliation(s)
- Malte P Bartram
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sandra Habbig
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany, Department of Pediatrics
| | - Caroline Pahmeyer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | | | - Marcus Krueger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | | |
Collapse
|
9
|
Martínez-García C, Izquierdo-Lahuerta A, Vivas Y, Velasco I, Yeo TK, Chen S, Medina-Gomez G. Renal Lipotoxicity-Associated Inflammation and Insulin Resistance Affects Actin Cytoskeleton Organization in Podocytes. PLoS One 2015; 10:e0142291. [PMID: 26545114 PMCID: PMC4636358 DOI: 10.1371/journal.pone.0142291] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/19/2015] [Indexed: 01/01/2023] Open
Abstract
In the last few decades a change in lifestyle has led to an alarming increase in the prevalence of obesity and obesity-associated complications. Obese patients are at increased risk of developing hypertension, heart disease, insulin resistance (IR), dyslipidemia, type 2 diabetes and renal disease. The excess calories are stored as triglycerides in adipose tissue, but also may accumulate ectopically in other organs, including the kidney, which contributes to the damage through a toxic process named lipotoxicity. Recently, the evidence suggests that renal lipid accumulation leads to glomerular damage and, more specifically, produces dysfunction in podocytes, key cells that compose and maintain the glomerular filtration barrier. Our aim was to analyze the early mechanisms underlying the development of renal disease associated with the process of lipotoxicity in podocytes. Our results show that treatment of podocytes with palmitic acid produced intracellular accumulation of lipid droplets and abnormal glucose and lipid metabolism. This was accompanied by the development of inflammation, oxidative stress and endoplasmic reticulum stress and insulin resistance. We found specific rearrangements of the actin cytoskeleton and slit diaphragm proteins (Nephrin, P-Cadherin, Vimentin) associated with this insulin resistance in palmitic-treated podocytes. We conclude that lipotoxicity accelerates glomerular disease through lipid accumulation and inflammation. Moreover, saturated fatty acids specifically promote insulin resistance by disturbing the cytoarchitecture of podocytes. These data suggest that renal lipid metabolism and cytoskeleton rearrangements may serve as a target for specific therapies aimed at slowing the progression of podocyte failure during metabolic syndrome.
Collapse
Affiliation(s)
- Cristina Martínez-García
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Genética Molecular. Universidad Rey Juan Carlos, Avda. de Atenas s/n, Alcorcón, Madrid, Spain
| | - Adriana Izquierdo-Lahuerta
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Genética Molecular. Universidad Rey Juan Carlos, Avda. de Atenas s/n, Alcorcón, Madrid, Spain
| | - Yurena Vivas
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Genética Molecular. Universidad Rey Juan Carlos, Avda. de Atenas s/n, Alcorcón, Madrid, Spain
| | - Ismael Velasco
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Genética Molecular. Universidad Rey Juan Carlos, Avda. de Atenas s/n, Alcorcón, Madrid, Spain
| | - Tet-Kin Yeo
- Division of Nephrology/Hypertension, Northwestern University, Chicago, Illinois, United States of America
| | - Sheldon Chen
- Division of Nephrology/Hypertension, Northwestern University, Chicago, Illinois, United States of America
| | - Gema Medina-Gomez
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Genética Molecular. Universidad Rey Juan Carlos, Avda. de Atenas s/n, Alcorcón, Madrid, Spain
| |
Collapse
|
10
|
Wood SJ, Goldufsky JW, Bello D, Masood S, Shafikhani SH. Pseudomonas aeruginosa ExoT Induces Mitochondrial Apoptosis in Target Host Cells in a Manner That Depends on Its GTPase-activating Protein (GAP) Domain Activity. J Biol Chem 2015; 290:29063-73. [PMID: 26451042 DOI: 10.1074/jbc.m115.689950] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Indexed: 12/17/2022] Open
Abstract
Pseudomonas aeruginosa is the most common cause of hospital-acquired pneumonia and a killer of immunocompromised patients. We and others have demonstrated that the type III secretion system (T3SS) effector protein ExoT plays a pivotal role in facilitating P. aeruginosa pathogenesis. ExoT possesses an N-terminal GTPase-activating protein (GAP) domain and a C-terminal ADP-ribosyltransferase (ADPRT) domain. Because it targets multiple non-overlapping cellular targets, ExoT performs several distinct virulence functions for P. aeruginosa, including induction of apoptosis in a variety of target host cells. Both the ADPRT and the GAP domain activities contribute to ExoT-induced apoptosis. The ADPRT domain of ExoT induces atypical anoikis by transforming an innocuous cellular protein, Crk, into a cytotoxin, which interferes with integrin survival signaling. However, the mechanism underlying the GAP-induced apoptosis remains unknown. In this study, we demonstrate that the GAP domain activity is both necessary and sufficient to induce mitochondrial (intrinsic) apoptosis. We show that intoxication with GAP domain results in: (i) JNK1/2 activation; (ii) substantial increases in the mitochondrial levels of activated pro-apoptotic proteins Bax and Bid, and to a lesser extent Bim; (iii) loss of mitochondrial membrane potential and cytochrome c release; and (iv) activation of initiator caspase-9 and executioner caspase-3. Further, GAP-induced apoptosis is partially mediated by JNK1/2, but it is completely dependent on caspase-9 activity. Together, the ADPRT and the GAP domains make ExoT into a highly versatile and potent cytotoxin, capable of inducing multiple forms of apoptosis in target host cells.
Collapse
Affiliation(s)
| | | | | | - Sara Masood
- From the Department of Immunology/Microbiology
| | - Sasha H Shafikhani
- From the Department of Immunology/Microbiology, Department of Internal Medicine, and Cancer Center, Rush University Medical Center, Chicago, Illinois 60612
| |
Collapse
|
11
|
Yin CP, Guan SH, Zhang B, Wang XX, Yue SW. Upregulation of HIF-1α protects neuroblastoma cells from hypoxia-induced apoptosis in a RhoA-dependent manner. Mol Med Rep 2015; 12:7123-31. [PMID: 26323527 DOI: 10.3892/mmr.2015.4267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
Hypoxic conditions regulate several metabolic enzymes and transcription factors that are involved in cancer, ischemia and pulmonary diseases. The Ras homolog (Rho) family, including Rho member A (RhoA), is involved in reorganization of the actin cytoskeleton, cell migration and in the regulation of apoptosis and gene transcription. The aim of the present study was to investigate the expression of hypoxia‑inducible factor (HIF)‑α and the activity of RhoA in PC12 neuroblastoma cells under hypoxic conditions. The upregulation of HIF‑α and RhoA by hypoxia was determined using reverse transcription‑quantitative polymerase chain reaction and western blot assays, cell apoptosis was analyzed using flow cytometry, and the activity of caspase 3 was examined using a western blot assay and caspase 3 activity assay kit. The PC12 cells were induced to apoptosis following exposure to hypoxia, and exhibited increased expression of HIF‑α and increased mRNA and protein expression levels of RhoA. The overexpression of HIF‑α attenuated the hypoxia‑induced apoptosis of the PC12 cells. In addition, RhoA knockdown using small interfering RNA abrogated the antagonism of HIF‑1α towards hypoxia‑induced apoptosis. The results of the present study confirmed the protective role of HIF‑1α and RhoA in hypoxia‑induced PC12 cell apoptosis, and that the upregulation of HIF‑1α by hypoxia is RhoA‑dependent.
Collapse
Affiliation(s)
- Cui-Ping Yin
- Department of Physical Medicine and Rehabilitation, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shang-Hui Guan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bo Zhang
- Department of Physical Medicine and Rehabilitation, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Xue-Xin Wang
- Department of Physical Medicine and Rehabilitation, Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Shou-Wei Yue
- Department of Physical Medicine and Rehabilitation, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
12
|
Dai R, Lin Y, Liu H, Rao J, Zhai Y, Zha X, Fang X, Xu H. A vital role for Angptl3 in the PAN-induced podocyte loss by affecting detachment and apoptosis in vitro. BMC Nephrol 2015; 16:38. [PMID: 25884163 PMCID: PMC4383073 DOI: 10.1186/s12882-015-0034-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/17/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Podocyte detachment and apoptosis are two risk factors causing podocyte loss, F-actin rearrangement is involved in detachment and apoptosis. However, the nature of events that promote detachment and apoptosis of podocytes and whether detachment occurred simultaneously with apoptosis are still unclear. Previously, it was found that angiopoietin-like3 (Angptl3) induces F-actin rearrangement in podocytes. In this study we investigate whether Angptl3 influences podocyte loss (detachment and apoptosis) and the process through which Angptl3 exactly influenced the podocyte loss. METHODS In conditionally immortalized mice podocytes, recombinant mice Angptl3 protein (rm-Angptl3) was used to mimic Angptl3 overexpression model and transfection with small interfering RNA (siRNA) to knockdown the expression of Angptl3. Both flow cytometry analysis and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay were used to detect apoptosis. Rearrangement of F-actin was assessed using confocal microscopy. Western blot assay was used to measure levels of Angptl3, integrin α3β1, integrin-linked kinase (ILK), p53, caspase 3, and phosphorylation of integrin β1. RESULTS In a puromycin aminonucleoside (PAN)-induced podocyte injury model, rm-Angptl3 accelerated the loss of podocytes, both detachment and apoptosis occurred, and F-actin rearrangement is involved in the process. However, knockdown of Angptl3 by siRNA markedly ameliorated these injuries. Observed effects were partially correlated with the altered integrin α3β1, ILK and p53, rather than caspase 3. CONCLUSIONS Angptl3 is a novel factor involved in the PAN-induced podocyte loss by affecting detachment and apoptosis in vitro. This study helps to deepen the understanding of the mechanisms of podocyte loss and lays the foundation for developing a new successful therapy for podocyte injury via lower expression of Angptl3.
Collapse
Affiliation(s)
- Rufeng Dai
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Yi Lin
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Shandong, 266003, China.
| | - Haimei Liu
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Jia Rao
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Yihui Zhai
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Xiliang Zha
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xiaoyan Fang
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Hong Xu
- Department of Nephrology and Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
13
|
Drozdova T, Papillon J, Cybulsky AV. Nephrin missense mutations: induction of endoplasmic reticulum stress and cell surface rescue by reduction in chaperone interactions. Physiol Rep 2013; 1:e00086. [PMID: 24303155 PMCID: PMC3831901 DOI: 10.1002/phy2.86] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/16/2013] [Accepted: 08/19/2013] [Indexed: 12/12/2022] Open
Abstract
Nephrin, an important component of the podocyte filtration slit diaphragm, plays a key role in the maintenance of glomerular permselectivity. Mutations in nephrin lead to proteinuria and congenital nephrotic syndrome. Nephrin undergoes posttranslational modifications in the endoplasmic reticulum (ER) prior to export to the plasma membrane. We examined the effects of human nephrin disease-associated missense mutations on nephrin folding in the ER and on cellular trafficking in cultured cells. Compared with wild-type (WT) nephrin, the mutants showed impaired glycosylation and enhanced association with the ER chaperone, calnexin, as well as accumulation in the ER. Nephrin mutants demonstrated enhanced ubiquitination, and they underwent ER-associated degradation. Certain nephrin mutants did not traffic to the plasma membrane. Expression of nephrin mutants resulted in the stimulation of the activating transcription factor-6 pathway of the unfolded protein response, and an increase in the ER chaperone, Grp94. We treated cells with castanospermine (an inhibitor of glucosidase I) in order to decrease the association of nephrin mutants with calnexin. Castanospermine increased plasma membrane expression of nephrin mutants; however, full glycosylation and signaling activity of the mutants were not restored. Modulation of ER quality control mechanisms represents a potential new approach to development of therapies for proteinuric kidney disease, including congenital nephrotic syndrome.
Collapse
Affiliation(s)
- Tetyana Drozdova
- Department of Medicine, McGill University Health Centre, McGill University Montreal, Quebec, Canada
| | | | | |
Collapse
|
14
|
Belmonte SL, Ram R, Mickelsen DM, Gertler FB, Blaxall BC. Cardiac overexpression of Mammalian enabled (Mena) exacerbates heart failure in mice. Am J Physiol Heart Circ Physiol 2013; 305:H875-84. [PMID: 23832697 DOI: 10.1152/ajpheart.00342.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mammalian enabled (Mena) is a key regulator of cytoskeletal actin dynamics, which has been implicated in heart failure (HF). We have previously demonstrated that cardiac Mena deletion produced cardiac dysfunction with conduction abnormalities and hypertrophy. Moreover, elevated Mena expression correlates with HF in human and animal models, yet the precise role of Mena in cardiac pathophysiology is unclear. In these studies, we evaluated mice with cardiac myocyte-specific Mena overexpression (TTA/TgTetMena) comparable to that observed in cardiac pathology. We found that the hearts of TTA/TgTetMena mice were functionally and morphologically comparable to wild-type littermates, except for mildly increased heart mass in the transgenic mice. Interestingly, TTA/TgTetMena mice were particularly susceptible to cardiac injury, as these animals experienced pronounced decreases in ejection fraction and fractional shortening as well as heart dilatation and hypertrophy after transverse aortic constriction (TAC). By "turning off" Mena overexpression in TTA/TgTetMena mice either immediately prior to or immediately after TAC surgery, we discovered that normalizing Mena levels eliminated cardiac hypertrophy in TTA/TgTetMena animals but did not preclude post-TAC cardiac functional deterioration. These findings indicate that hearts with increased levels of Mena fare worse when subjected to cardiac injury and suggest that Mena contributes to HF pathophysiology.
Collapse
Affiliation(s)
- Stephen L Belmonte
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | | | | | | | | |
Collapse
|
15
|
The Expression of Intermediate Filament Protein Nestin and Its Association With Cyclin-dependent Kinase 5 in the Glomeruli of Rats With Diabetic Nephropathy. Am J Med Sci 2013; 345:470-7. [DOI: 10.1097/maj.0b013e3182648459] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Chiluiza D, Krishna S, Schumacher VA, Schlöndorff J. Gain-of-function mutations in transient receptor potential C6 (TRPC6) activate extracellular signal-regulated kinases 1/2 (ERK1/2). J Biol Chem 2013; 288:18407-20. [PMID: 23645677 DOI: 10.1074/jbc.m113.463059] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Gain-of-function mutations in the canonical transient receptor potential 6 (TRPC6) gene are a cause of autosomal dominant focal segmental glomerulosclerosis (FSGS). The mechanisms whereby abnormal TRPC6 activity results in proteinuria remain unknown. The ERK1/2 MAPKs are activated in glomeruli and podocytes in several proteinuric disease models. We therefore examined whether FSGS-associated mutations in TRPC6 result in activation of these kinases. In 293T cells and cultured podocytes, overexpression of gain-of-function TRPC6 mutants resulted in increased ERK1/2 phosphorylation, an effect dependent upon channel function. Pharmacologic inhibitor studies implicated several signaling mediators, including calmodulin and calcineurin, supporting the importance of TRPC6-mediated calcium influx in this process. Through medium transfer experiments, we uncovered two distinct mechanisms for ERK activation by mutant TRPC6, a cell-autonomous, EGF receptor-independent mechanism and a non-cell-autonomous mechanism involving metalloprotease-mediated release of a presumed EGF receptor ligand. The inhibitors KN-92 and H89 were able to block both pathways in mutant TRPC6 expressing cells as well as the prolonged elevation of intracellular calcium levels upon carbachol stimulation seen in these cells. However, these effects appear to be independent of their effects on calcium/calmodulin-dependent protein kinase II and PKA, respectively. Phosphorylation of Thr-70, Ser-282, and Tyr-31/285 were not necessary for ERK activation by mutant TRPC6, although a phosphomimetic TRPC6 S282E mutant was capable of ERK activation. Taken together, these results identify two pathways downstream of mutant TRPC6 leading to ERK activation that may play a role in the development of FSGS.
Collapse
Affiliation(s)
- David Chiluiza
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
17
|
Krtil J, Pláteník J, Čuřík N, Brima W, Tesař V, Zima T. The protective effects of erythropoietin on rat glomerular podocytes in culture are modulated by extracellular matrix proteins. Kidney Blood Press Res 2013; 38:142-55. [PMID: 24685986 DOI: 10.1159/000355762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Podocytes are typically cultured on collagen I; however, collagen I is absent from healthy glomerular basement membranes. Erythropoietin (EPO) is thought to protect podocytes in vivo. Here, we studied how various types of extracellular matrix (ECM) proteins and EPO affect podocytes in culture. METHODS Primary rat podocytes were replated on collagen I, collagen IV, whole ECM extract, laminin, or bare plastic. Cellular adhesion (8 hours after plating), proliferation (5 days, 10 % serum), and resistance to serum deprivation (3 days, 0.5 % serum) were assessed. BrdU incorporation and expression of podocyte-specific markers were employed as measures of cellular proliferation and differentiation, respectively. qPCR was used to verify expression of EPO receptor in cultured podocytes. RESULTS Cellular adhesion was similar on all ECM proteins and unaffected by EPO. Proliferation was accelerated by laminin and the ECM extract, but the final cell density was similar on all ECM surfaces. Collagen IV supported the serum-deprived cells better than the other ECM proteins. EPO (2-20 ng/ml) improved viability of serum-deprived podocytes on collagen I, collagen IV, and ECM, but not on laminin or bare plastic. The cells expressed mRNA for EPO receptor. CONCLUSION The physiological ECM proteins are more supportive of primary podocytic cultures compared with collagen I. The protective effects of EPO during serum deprivation are modulated by the cultivation surface.
Collapse
Affiliation(s)
- Jan Krtil
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | | | | | | | | | | |
Collapse
|
18
|
Liu W, Zhang Y, Hao J, Liu S, Liu Q, Zhao S, Shi Y, Duan H. Nestin protects mouse podocytes against high glucose-induced apoptosis by a Cdk5-dependent mechanism. J Cell Biochem 2012; 113:3186-96. [DOI: 10.1002/jcb.24195] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
19
|
Xie P, Kondeti VK, Lin S, Haruna Y, Raparia K, Kanwar YS. Role of extracellular matrix renal tubulo-interstitial nephritis antigen (TINag) in cell survival utilizing integrin (alpha)vbeta3/focal adhesion kinase (FAK)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B-serine/threonine kinase (AKT) signaling pathway. J Biol Chem 2011; 286:34131-46. [PMID: 21795690 PMCID: PMC3190785 DOI: 10.1074/jbc.m111.241778] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Revised: 07/19/2011] [Indexed: 11/06/2022] Open
Abstract
Tubulo-interstitial nephritis antigen (TINag) is an extracellular matrix protein expressed in tubular basement membranes. Combined mutations in TINag and nephrocystin-1 genes lead to nephronophthisis with reduced cell survival. Because certain extracellular matrix proteins are known to modulate cell survival, studies were initiated in Lewis rats lacking TINag to assess if they are more susceptible to cisplatin-induced injury. Cisplatin induced a higher degree of tubular cell damage and apoptosis in regions where TINag is expressed in a parental Wistar strain. This was accompanied by an accentuated increase in serum creatinine and Kim-1 RNA and renal expression of Bax, p53, and its nuclear accumulation, mtDNA fragmentation, and a decrease of Bcl-2. Cisplatin induced fulminant apoptosis of HK-2 cells with increased caspase3/7 activity, mtDNA fragmentation, and a reduced cell survival. These effects were partially reversed in cells maintained on TINag substratum. Far Western/solid phase assays established TINag binding with integrin αvβ3 comparable with vitronectin. Transfection of cells with αv-siRNA accentuated cisplatin-induced apoptosis, aberrant translocation of cytochrome c and Bax, and reduced cell survival. The αv-siRNA decreased expression of integrin-recruited focal adhesion kinase (FAK) and p-FAK, while increasing the expression of p53 and p-p53. Similarly, p-AKT was reduced although ILK was unaffected. Inhibition of PI3K had similar adverse cellular effects. These effects were ameliorated in cells on TINag substratum. In vivo, a higher degree of decrease in the expression of p-FAK and pAKT was observed in Lewis rats following cisplatin treatment. These in vivo and in vitro studies demonstrate an essential role of TINag in cellular survival to maintain proper tubular homeostasis utilizing integrin αvβ3 and downstream effectors.
Collapse
Affiliation(s)
- Ping Xie
- From the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| | - Vinay K. Kondeti
- From the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| | - Sun Lin
- the Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha 410011, China, and
| | - Yoshisuke Haruna
- the Department of Medicine, Kawasaki Medical School, Kurashiki, Okayama 7010192, Japan
| | - Kirtee Raparia
- From the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yashpal S. Kanwar
- From the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
20
|
Wen D, You L, Zhang Q, Zhang L, Gu Y, Hao CM, Chen J. Upregulation of nestin protects podocytes from apoptosis induced by puromycin aminonucleoside. Am J Nephrol 2011; 34:423-34. [PMID: 21952051 DOI: 10.1159/000331701] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/12/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND Nestin is an intermediate filament protein widely used as a marker of stem cells or progenitor cells. Nestin is also highly expressed in the glomerular podocyte, a type of terminally differentiated epithelial cell. Little is known about the significance of nestin in podocytes. METHODS Puromycin aminonucleoside (PAN) was injected into the rats to produce a PAN nephrosis model. Transmission electronic microscopy and terminal dUTP nick end-labeling assay were used to examine the podocyte foot process (FP) effacement and apoptosis, respectively. A mouse podocyte cell line was cultured and incubated with PAN. Immunoblot was used to examine the level of nestin expression both in vivo and in vitro. Enhanced green fluorescence protein-tagged plasmids containing nestin shRNA were transfected into the cultured podocytes to silence nestin expression. F-actin arrangement within cultured podocytes was investigated by immunofluorescence, while the apoptosis rate was examined by both Hoechst stain and flow cytometry. RESULTS In the PAN-induced rat nephrosis model, podocyte nestin expression was increased in the absence of apparent podocyte apoptosis, even though the FP was significantly effaced. In the cultured mouse podocytes, PAN upregulated nestin expression in a time-dependent manner within 24 h of treatment. Notably, no significant apoptosis occurred, however knocking down nestin expression resulted in a remarkable derangement of actin cytoskeleton and an increase in apoptosis in the cultured podocytes 24 h after being incubated with PAN. CONCLUSIONS Upregulation of nestin expression during PAN nephrosis could protect podocytes from apoptosis and that this process is mediated by maintaining the regular arrangement of actin cytoskeleton.
Collapse
Affiliation(s)
- Donghai Wen
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Podocyte Injury Associated with Mutant α-Actinin-4. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:563128. [PMID: 21808733 PMCID: PMC3144672 DOI: 10.1155/2011/563128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 05/08/2011] [Indexed: 11/17/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) is an important cause of proteinuria and nephrotic syndrome in humans. The pathogenesis of FSGS may be associated with glomerular visceral epithelial cell (GEC; podocyte) injury, leading to apoptosis, detachment, and "podocytopenia", followed by glomerulosclerosis. Mutations in α-actinin-4 are associated with FSGS in humans. In cultured GECs, α-actinin-4 mediates adhesion and cytoskeletal dynamics. FSGS-associated α-actinin-4 mutants show increased binding to actin filaments, compared with the wild-type protein. Expression of an α-actinin-4 mutant in mouse podocytes in vivo resulted in proteinuric FSGS. GECs that express mutant α-actinin-4 show defective spreading and motility, and such abnormalities could alter the mechanical properties of the podocyte, contribute to cytoskeletal disruption, and lead to injury. The potential for mutant α-actinin-4 to injure podocytes is also suggested by the characteristics of this mutant protein to form microaggregates, undergo ubiquitination, impair the ubiquitin-proteasome system, enhance endoplasmic reticulum stress, and exacerbate apoptosis.
Collapse
|
22
|
Cybulsky AV, Takano T, Papillon J, Kitzler TM, Bijian K. Endoplasmic reticulum stress in glomerular epithelial cell injury. Am J Physiol Renal Physiol 2010; 301:F496-508. [PMID: 21159733 DOI: 10.1152/ajprenal.00728.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) may be associated with glomerular epithelial cell (GEC; podocyte) apoptosis due to acquired injury or mutations in specific podocyte proteins. This study addresses mediation of GEC injury, focusing on endoplasmic reticulum (ER) stress. We studied signaling in cultured GECs in the presence or absence of the extracellular matrix (ECM). Adhesion to collagen supports cell survival, but adhesion to plastic (loss of contact with ECM) leads to apoptosis. Compared with collagen-adherent cells, GECs on plastic showed increased protein misfolding in the ER, and an adaptive-protective ER stress response, including increased expression of ER chaperones, increased phosphorylation of eukaryotic translation initiation factor-2α (eIF2α), and a reduction in protein synthesis. Activation of these ER stress pathways counteracted apoptosis. However, tunicamycin (a potent stimulator of ER stress) changed the ER stress response from protective to cytotoxic, as tunicamycin induced the proapoptotic ER stress gene, C/EBP homologous protein-10, and exacerbated apoptosis in GECs adherent to plastic, but not collagen. In GECs adherent to plastic, adaptive ER stress was associated with an increase in polyubiquitinated proteins and "choking" of the proteasome. Furthermore, pharmacological inhibition of the proteasome induced ER stress in GECs. Finally, we show that ER stress (induction of ER chaperones and eIF2α phosphorylation) was evident in experimental FSGS in vivo. Thus interactions of GECs with ECM may regulate protein folding and induction of the ER stress response. FSGS is associated with induction of ER stress. Enhancing protective aspects of the ER stress response may reduce apoptosis and possibly glomerulosclerosis.
Collapse
Affiliation(s)
- Andrey V Cybulsky
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | | | | | | | | |
Collapse
|
23
|
Sanchez-Niño MD, Sanz AB, Lorz C, Gnirke A, Rastaldi MP, Nair V, Egido J, Ruiz-Ortega M, Kretzler M, Ortiz A. BASP1 promotes apoptosis in diabetic nephropathy. J Am Soc Nephrol 2010; 21:610-21. [PMID: 20110383 DOI: 10.1681/asn.2009020227] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Apoptosis contributes to the development of diabetic nephropathy (DN), but the mechanisms that lead to diabetes-induced cell death are not fully understood. Here, we combined a functional genomics screen for cDNAs that induce apoptosis in vitro with transcriptional profiling of renal biopsies from patients with DN. Twelve of the 138 full-length cDNAs that induced cell death in human embryonic kidney cells matched upregulated mRNA transcripts in tissue from human DN. Confirmatory screens identified induction of BASP1 in tubular cross sections of human DN tissue. In vitro, apoptosis-inducing conditions such as serum deprivation, high concentrations of glucose, and proinflammatory cytokines increased BASP1 mRNA and protein in human tubular epithelial cells. In normal cells, BASP1 localized to the cytoplasm, but in apoptotic cells, it colocalized with actin in the periphery. Overexpression of BASP1 induced cell death with features of apoptosis; conversely, small interfering RNA (siRNA)-mediated knockdown of BASP1 protected tubular cells from apoptosis. Supporting possible involvement of BASP1 in renal disease other than DN, we also observed significant upregulation of renal BASP1 in spontaneously hypertensive rats and a trend toward increased tubulointerstitial BASP1 mRNA in human hypertensive nephropathy. In summary, a combined functional genomics approach identified BASP1 as a proapoptotic factor in DN and possibly also in hypertensive nephropathy.
Collapse
Affiliation(s)
- Maria Dolores Sanchez-Niño
- Nefrología, Fundación Jiménez Díaz, Universidad Autonoma de Madrid and Instituto Reina Sofia de Investigaciones Nefrológicas-IRSIN, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Su B, Bu Y, Engelberg D, Gelman IH. SSeCKS/Gravin/AKAP12 inhibits cancer cell invasiveness and chemotaxis by suppressing a protein kinase C- Raf/MEK/ERK pathway. J Biol Chem 2009; 285:4578-86. [PMID: 20018890 DOI: 10.1074/jbc.m109.073494] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
SSeCKS/Gravin/AKAP12 ("SSeCKS") encodes a cytoskeletal protein that regulates G(1) --> S progression by scaffolding cyclins, protein kinase C (PKC) and PKA. SSeCKS is down-regulated in many tumor types including prostate, and when re-expressed in MAT-LyLu (MLL) prostate cancer cells, SSeCKS selectively inhibits metastasis by suppressing neovascularization at distal sites, correlating with its ability to down-regulate proangiogenic genes including Vegfa. However, the forced re-expression of VEGF only rescues partial lung metastasis formation. Here, we show that SSeCKS potently inhibits chemotaxis and Matrigel invasion, motility parameters contributing to metastasis formation. SSeCKS suppressed serum-induced activation of the Raf/MEK/ERK pathway, resulting in down-regulation of matrix metalloproteinase-2 expression. In contrast, SSeCKS had no effect on serum-induced phosphorylation of the Src substrate, Shc, in agreement with our previous data that SSeCKS does not inhibit Src kinase activity in cells. Invasiveness and chemotaxis could be restored by the forced expression of constitutively active MEK1, MEK2, ERK1, or PKCalpha. SSeCKS suppressed phorbol ester-induced ERK1/2 activity only if it encoded its PKC binding domain (amino acids 553-900), suggesting that SSeCKS attenuates ERK activation through a direct scaffolding of conventional and/or novel PKC isozymes. Finally, control of MLL invasiveness by SSeCKS is influenced by the actin cytoskeleton: the ability of SSeCKS to inhibit podosome formation is unaffected by cytochalasin D or jasplakinolide, whereas its ability to inhibit MEK1/2 and ERK1/2 activation is nullified by jasplakinolide. Our findings suggest that SSeCKS suppresses metastatic motility by disengaging activated Src and then inhibiting the PKC-Raf/MEK/ERK pathways controlling matrix metalloproteinase-2 expression and podosome formation.
Collapse
Affiliation(s)
- Bing Su
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | |
Collapse
|
25
|
Karoubi G, Ormiston ML, Stewart DJ, Courtman DW. Single-cell hydrogel encapsulation for enhanced survival of human marrow stromal cells. Biomaterials 2009; 30:5445-55. [PMID: 19595454 DOI: 10.1016/j.biomaterials.2009.06.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 06/21/2009] [Indexed: 02/08/2023]
Abstract
Inadequate extracellular matrix cues and subsequent apoptotic cell death are among crucial factors currently limiting cell viability and organ retention in cell-based therapeutic strategies for vascular regeneration. Here we describe the use of a single-cell hydrogel capsule to provide enhanced cell survival of adherent cells in transient suspension culture. Human marrow stromal cells (hMSCs) were singularly encapsulated in agarose capsules containing the immobilized matrix molecules, fibronectin and fibrinogen to ameliorate cell-matrix survival signals. MSCs in the enriched capsules demonstrated increased viability, greater metabolic activity and enhanced cell-cytoskeletal patterning. Increased cell viability resulted from the re-induction of cell-matrix interactions likely via integrin clustering and subsequent activation of the extracellular signal regulated MAPK (ERK)/mitogen activated protein kinase (MAPK) signaling cascade. Proof of principle in-vivo studies, investigating autologous MSC delivery into Fisher 344 rat hindlimb, depicted a significant increase in the number of engrafted cells using the single-cell encapsulation system. Incorporation of immobilized adhesion molecules compensates, at least in part, for the missing cell-matrix cues, thereby attenuating the initial anoikis stimuli and providing protection from subsequent apoptosis. Thus, this single-cell encapsulation strategy may markedly enhance therapeutic cell survival in targeted tissues.
Collapse
Affiliation(s)
- Golnaz Karoubi
- University of Toronto and St. Michael's Hospital, Toronto, ON, Canada
| | | | | | | |
Collapse
|
26
|
Cybulsky AV, Takano T, Papillon J, Bijian K, Guillemette J, Kennedy CRJ. Glomerular epithelial cell injury associated with mutant α-actinin-4. Am J Physiol Renal Physiol 2009; 297:F987-95. [PMID: 19640905 DOI: 10.1152/ajprenal.00055.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) may be associated with glomerular epithelial cell (GEC; podocyte) apoptosis due to acquired injury or mutations in α-actinin-4. This study addresses how FSGS-associated mutant α-actinin-4 may induce GEC injury, focusing on endoplasmic reticulum (ER) stress and metabolism of mutant α-actinin-4 via the ubiquitin-proteasome system. In a model of experimental FSGS induced by expression of an α-actinin-4 K256E transgene in podocytes, we show induction of ER stress, including upregulation of ER chaperones (bip, grp94), phosphorylation of the eukaryotic translation initiation factor-2α subunit, and induction of the proapop totic gene C/EBP homologous protein-10 (CHOP). To address mechanisms of ER stress, we studied signaling in cultured GEC and COS cells expressing α-actinin-4 K256E. Previously, we showed that expression of this α-actinin-4 mutant in GEC increased apoptosis. In the present study, we show that α-actinin-4 K256E upregulates grp94 and CHOP expression in COS cells and significantly exacerbates induction of bip and CHOP in GEC in the presence of tunicamycin. ER stress was associated with aggregation and ubiquitination of α-actinin-4 K256E and impairment of the ubiquitin-proteasome system. In addition, α-actinin-4 K256E exacerbated apoptosis in the context of mild proteasome inhibition. Thus α-actinin-4 K256E triggers several metabolic abnormalities, which may lead to GEC injury and glomerulosclerosis.
Collapse
Affiliation(s)
- Andrey V. Cybulsky
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec; and
| | - Tomoko Takano
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec; and
| | - Joan Papillon
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec; and
| | - Krikor Bijian
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec; and
| | - Julie Guillemette
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec; and
| | - Chris R. J. Kennedy
- Kidney Research Centre, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
27
|
Liu S, Ding J, Fan Q, Zhang H. The activation of extracellular signal-regulated kinase is responsible for podocyte injury. Mol Biol Rep 2009; 37:2477-84. [PMID: 19728154 DOI: 10.1007/s11033-009-9761-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 08/11/2009] [Indexed: 01/02/2023]
Abstract
Podocyte and its slit diaphragm play an important role in maintaining normal glomerular filtration barrier function and structure. Podocyte apoptosis and slit diaphragm injury leads to proteinuria and glomerulosclerosis. However, the molecular mechanism of podocyte injury remains poorly understood. The family of mitogen-activated protein kinases including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase, and p38 signal pathways, are implicated in the progression of various glomerulopathies. However, the role of the activated signal pathway(s) in podocyte injury is elusive. This study examined phosphorylation of ERK in rat puromycin aminonucleoside (PAN) nephropathy as well as conditionally immortalized mouse podocyte treated with PAN in vitro. The effect of treatment with U0126, an inhibitor of ERK, was also investigated. In PAN nephropathy, the phosphorylation of ERK was marked. In podocyte injury, the marked and sustained activation of ERK pathway was also observed before the appearance of significant podocyte apoptosis. Pretreatment with U0126 to podocyte completely inhibited ERK activation, with complete suppression podocyte apoptosis and ameliorated nephrin protein expression along with the phosphorylation of nephrin in podocyte injury. In cultured podocyte, PAN induced actin recorganition, and U0126 inhibited such change. However, U0126 did not recovery the phosphorylation change of neph1 in podocyte injury. We concluded that the sustained activation of ERK along with the phosphorylation of neph1 might be necessary for podocyte injury. The study here suggested that ERK might become a potential target for therapeutic intervention to prevent podocytes from injury which will result in proteinuria.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pediatrics, Peking University First Hospital, No.1, Xi An Men Da Jie, 100034, Beijing, People's Republic of China.
| | | | | | | |
Collapse
|
28
|
Huynh C, Ren G, Papillon J, Guillemette J, Takano T, Cybulsky AV. The cytoprotective role of Ras in complement-mediated glomerular epithelial cell injury. Clin Immunol 2009; 131:343-53. [DOI: 10.1016/j.clim.2008.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 11/24/2008] [Accepted: 11/24/2008] [Indexed: 11/16/2022]
|
29
|
Zhu B, Chen N, Wang ZH, Pan XX, Ren H, Zhang W, Wang WM. Identification and functional analysis of a novel TRPC6 mutation associated with late onset familial focal segmental glomerulosclerosis in Chinese patients. Mutat Res 2008; 664:84-90. [PMID: 19124028 DOI: 10.1016/j.mrfmmm.2008.11.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 11/14/2008] [Accepted: 11/21/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Mutations in the TRPC6 gene are responsible for a late onset form of familial focal segmental glomerulosclerosis (FSGS). However, the role of TRPC6 variation in Chinese patients with late onset familial FSGS remains unclear. Here, we screened 31 Chinese pedigrees with late onset familial FSGS for changes in TRPC6 by DNA sequence analysis. METHODS Genomic DNA was extracted from peripheral leukocytes. We PCR-amplified each of 13 exons of TRPC6 for sequence analysis. When a novel nucleotide change seemed likely to cause FSGS, we carried out an in vitro research to determine the effects of the mutation on TRPC6 function. HEK 293 cells were transfected stably with vectors containing mutant or wild type TRPC6 cDNA. We then compared the expression of mutant TRPC6 to wild type TRPC6 using Western blot. For the observation of the function of mutant TRPC6 channel compared with wild type TRPC6 channel, Intracellular Ca(2+) concentration was detected using fluorescent indicator Fluo-3 among different groups of cells upon stimulation with 1-oleoyl-2-acetyl sn-glycerol (OAG). RESULTS All the 31 pedigrees with late onset familial FSGS were collected in our department from September 1997 to October 2007. A novel TRPC6 mutation (cytosine 2664 adenine resulting in Glutamine 889 Lysine substitution, Q889K) was identified in one of these pedigrees. Mutant TRPC6 (TRPC6(Q889K)) or wild type TRPC6 was stably expressed in HEK293 cells by Western blot. The mutant TRPC6 expression was a little increased without significant difference compared with wild type TRPC6 expression, whereas the intracellular Ca(2+) level in cells expressing mutant TRPC6 was significantly increased compared with that in the cells expressing wild TRPC6 upon stimulation. CONCLUSION We identified a novel TRPC6 mutation Q889K associated with late onset FFSGS in Chinese pedigrees and this mutation was demonstrated to be "gain of function" by an in vitro functional research.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Nephrology, Ruijin Hospital, School of medicine, Shanghai Jiaotong University, Shanghai 200025, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Cytotoxicity of Mononuclear Cells as Induced by Peritoneal Dialysis Fluids: Insight into Mechanisms that Regulate Osmotic Stress-Related Apoptosis. Perit Dial Int 2008. [DOI: 10.1177/089686080802800619] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective High glucose content of peritoneal dialysis fluids (PDFs) has been shown to contribute to loss of peritoneal function during long-term peritoneal dialysis. However, hyperosmolality and hypertonicity of PDF are usually seen as similar stress events inducing osmotic stress-induced programmed cell death. In this study, we examined the impact of various osmotic agents on apoptosis induced by hyperosmolar PDFs, focusing on the mechanisms underlying the lethal effects of PDFs on peripheral blood mono-nuclear cells (PBMCs). Methods We assessed apoptosis and necrosis by annexin V–propidium iodide (PI) labeling, and caspase-3 activity by fluorescence assay. F-actin remodeling was measured using fluorescent phalloidin labeling. Results Hyperosmolality does not cause the cytotoxicity observed with PDF, but exposure to agents incapable of permeating cell membranes results in a significant increase in the percentage of apoptotic PBMCs by annexin V–PI labeling, which is confirmed by the increase in caspase-3 activity. Interestingly, inhibition of caspase-3 by Z-VAD-FMK did not suppress apoptosis. Extracellular hypertonicity produced polymerization of filamentous actin and cell shrinkage, which displayed similar time courses. Cell shrinkage was blocked by cytochalasin D, indicating an active role for actin cytoskeleton in hypertonicity-induced cell shrinkage. F-actin polymerization was related to an increase in intra-cellular ionic strength. Finally, we excluded a direct role for actin remodeling in osmotic stress-induced programmed cell death. Conclusions Exposure to osmolytes that cannot penetrate cell membranes results in a hypertonicity-induced apoptosis that cannot be blocked by the broad-spectrum caspase inhibitor Z-VAD-FMK. In addition, extracellular hypertonicity induced by impermeant solutes produces F-actin polymerization through an increase in intracellular ionic strength. The remodeling of the cytoskeleton does not modulate apoptosis but participates in cell shrinkage.
Collapse
|
31
|
Goldwich A, Scholz M, Tamm ER. Myocilin promotes substrate adhesion, spreading and formation of focal contacts in podocytes and mesangial cells. Histochem Cell Biol 2008; 131:167-80. [PMID: 18855004 DOI: 10.1007/s00418-008-0518-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2008] [Indexed: 11/30/2022]
Abstract
Myocilin, a secreted glycoprotein of the olfactomedin family, is constitutively expressed in podocytes of the rat kidney and induced in mesangial cells during mesangioproliferative glomerulonephritis. As myocilin has been found to be associated with fibrillar components of the extracellular matrix, and adhesive properties have been shown for other members of the olfactomedin family, we hypothesized that myocilin might play a role in cell-matrix interactions in the glomerulus. To elucidate functional properties of myocilin, recombinant myocilin was expressed in 293 EBNA cells and purified by Ni-chelate and heparin chromatography. Culture plates were coated with myocilin, and primary rat mesangial cells and cells from an immortal murine podocyte cell line were seeded onto the plates in serum free conditions. Both cell types showed concentration-dependant attachment to myocilin, an effect that was statistically significant and could be blocked with specific antibodies. When compared to equal amounts of fibronectin or collagen 1, myocilin was less effective in promoting substrate adhesion. Synergistic effects in substrate adhesion were observed when myocilin was added to low concentrations of fibronectin. Twenty-five percent of cells that had attached to myocilin substrates showed spreading and expressed focal contacts which were labeled by vinculin/phalloidin staining. Comparable findings were observed when human or murine trabecular meshwork cells were seeded on myocilin substrates. Adhesive properties of myocilin required multimer formation, and were not observed when culture plates were coated with a C-terminal fragment of myocilin, containing the olfactomedin domain. We conclude that myocilin promotes substrate adhesion of podocytes and mesangial cells, and might contribute to cell-matrix adhesion of both cell types in vivo.
Collapse
Affiliation(s)
- Andreas Goldwich
- Institute of Anatomy, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
32
|
Ung CY, Li H, Ma XH, Jia J, Li BW, Low BC, Chen YZ. Simulation of the regulation of EGFR endocytosis and EGFR-ERK signaling by endophilin-mediated RhoA-EGFR crosstalk. FEBS Lett 2008; 582:2283-90. [PMID: 18505685 DOI: 10.1016/j.febslet.2008.05.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 04/28/2008] [Accepted: 05/16/2008] [Indexed: 12/24/2022]
Abstract
Deregulations of EGFR endocytosis in EGFR-ERK signaling are known to cause cancers and developmental disorders. Mutations that impaired c-Cbl-EGFR association delay EGFR endocytosis and produce higher mitogenic signals in lung cancer. ROCK, an effector of small GTPase RhoA was shown to negatively regulate EGFR endocytosis via endophilin A1. A mathematical model was developed to study how RhoA and ROCK regulate EGFR endocytosis. Our study suggested that over-expressing RhoA as well as ROCK prolonged ERK activation partly by reducing EGFR endocytosis. Overall, our study hypothesized an alternative role of RhoA in tumorigenesis in addition to its regulation of cytoskeleton and cell motility.
Collapse
Affiliation(s)
- Choong Yong Ung
- Bioinformatics and Drug Design Group, Department of Pharmacy, National University of Singapore, 3 Science Drive 2, Singapore 117543, Singapore
| | | | | | | | | | | | | |
Collapse
|
33
|
Zhu S, Korzh V, Gong Z, Low BC. RhoA prevents apoptosis during zebrafish embryogenesis through activation of Mek/Erk pathway. Oncogene 2007; 27:1580-9. [PMID: 17873909 DOI: 10.1038/sj.onc.1210790] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
RhoA small GTPase, as a key regulator for actin cytoskeletal rearrangement, plays pivotal roles during morphogenesis, cytokinesis, phagocytosis and cell migration, but little is known about its signaling mechanism that controls cell survival in vivo. Using zebrafish as a model, we show that non-overlapping antisense morpholinos that block either translation or splicing of rhoA lead to extensive apoptosis during embryogenesis, resulting in overall reduction of body size and body length. These defects are associated with reduced activation of growth-promoting Erk and decreased expression of anti-apoptotic bcl-2. Moreover, ectopic expression of rhoA, Mek or BCL-2 mRNA rescues such phenotypes. Consistently, combined suppression of RhoA and Mek/Erk or Bcl-2 pathways by sub-optimal dose of rhoA morpholino and pharmacological inhibitors for either Mek (U0126) or Bcl-2 (HA 14-1) can induce developmental abnormalities and enhanced apoptosis, similar to those caused by effective RhoA knockdown. Furthermore, U0126 abrogates the rescue by RhoA and MEK but not BCL-2. In contrast, HA 14-1 effectively abolishes all functional rescues by RhoA, MEK or BCL-2, supporting that RhoA prevents apoptosis by activation of Mek/Erk pathway and requiring Bcl-2. These findings reveal an important genetic and functional relationship between RhoA with Mek/Erk and Bcl-2 for cell survival control during embryogenesis.
Collapse
Affiliation(s)
- S Zhu
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
34
|
Bijian K, Zhang L, Shen SH. Collagen-mediated survival signaling is modulated by CD45 in Jurkat T cells. Mol Immunol 2007; 44:3682-90. [PMID: 17524482 DOI: 10.1016/j.molimm.2007.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 04/03/2007] [Accepted: 04/06/2007] [Indexed: 11/20/2022]
Abstract
T cell activation is a critical step in the development of a proper immune response to infection and inflammation. This dynamic process requires efficient T cell receptor signaling, which in turn is modulated by integrin receptor activation and the actin cytoskeleton. CD45 is a key player in T cell receptor mediated signal transduction. However, its exact role in integrin mediated signaling in T cells remains to be elucidated. The present study addresses the relationship between CD45 and beta1-integrin mediated survival signaling in the human T leukemic cell line Jurkat, in which collagen receptors alpha1 beta1 and alpha2 beta1 integrins are localized. Wild type (WT)-Jurkat T cells treated with collagen demonstrated increased cell proliferation and survival. Monitoring the intracellular signaling pathways activated by collagen in WT-Jurkat cells revealed increased focal adhesion kinase (FAK) and extracellular signal-regulated kinase (ERK) activation. Moreover, examination of the actin cytoskeleton of WT-Jurkat T cells treated with collagen demonstrated the presence of an organized cortical actin structure, reminiscent of the survival phenotype. This is in contrast to CD45-deficient J45.01 T cells, where collagen treatment failed to enhance cell proliferation/survival and was unable to stimulate FAK and ERK activity. In addition, the actin cytoskeleton of collagen treated J45.01 T cells was disorganized with cortical actin aggregates present throughout. The importance of an organized actin cytoskeleton to proper cell signaling and survival was further demonstrated by the inability of collagen treated WT-Jurkat cells to activate the FAK and ERK survival pathway in the presence of cytochalasin D, a cytoskeleton-disrupting drug. Consistently, addition of the CD45 specific inhibitor abolished collagen-stimulated FAK and ERK activation in WT-Jurkat cells, further depicting CD45 as the key mediator. Furthermore, collagen-mediated T cell signaling alone was able to activate IL-2 gene transcription devoid of concomitant T cell receptor activation. Taken together, these results are the first to demonstrate that CD45 is important in promoting cell survival by modulating integrin-mediated FAK/ERK signaling in Jurkat T cells and is involved in a distinct signal transduction pathway, separate from T cell receptor signaling, influencing T cell immune responses. Hence, this study will help further our knowledge about beta1-integrin mediated signaling in T cells, which may prove to be essential for the regulation of various T cell mediated immune responses.
Collapse
Affiliation(s)
- Krikor Bijian
- Mammalian Cell Genetics Group, Biotechnology Research Institute, National Research Council of Canada, 6100 Royalmount av., Montreal, Quebec H4P 2R2, Canada
| | | | | |
Collapse
|