29 results on '"Anil Karihaloo"'
Search Results
2. Prognostic imaging biomarkers for diabetic kidney disease (iBEAt): study protocol
- Author
-
Kim M. Gooding, Chrysta Lienczewski, Massimo Papale, Niina Koivuviita, Marlena Maziarz, Anna-Maria Dutius Andersson, Kanishka Sharma, Paola Pontrelli, Alberto Garcia Hernandez, Julie Bailey, Kay Tobin, Virva Saunavaara, Anna Zetterqvist, David Shelley, Irvin Teh, Claire Ball, Sapna Puppala, Mark Ibberson, Anil Karihaloo, Kaj Metsärinne, Rosamonde E. Banks, Peter S. Gilmour, Michael Mansfield, Mark Gilchrist, Dick de Zeeuw, Hiddo J. L. Heerspink, Pirjo Nuutila, Matthias Kretzler, Matthew Welberry Smith, Loreto Gesualdo, Dennis Andress, Nicolas Grenier, Angela C. Shore, Maria F. Gomez, Steven Sourbron, and for the BEAt-DKD consortium
- Subjects
Diabetic kidney disease ,Type 2 diabetes ,Magnetic resonance imaging ,Ultrasound ,Albuminuria ,Chronic kidney disease stages 1–3 ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Abstract Background Diabetic kidney disease (DKD) remains one of the leading causes of premature death in diabetes. DKD is classified on albuminuria and reduced kidney function (estimated glomerular filtration rate (eGFR)) but these have modest value for predicting future renal status. There is an unmet need for biomarkers that can be used in clinical settings which also improve prediction of renal decline on top of routinely available data, particularly in the early stages. The iBEAt study of the BEAt-DKD project aims to determine whether renal imaging biomarkers (magnetic resonance imaging (MRI) and ultrasound (US)) provide insight into the pathogenesis and heterogeneity of DKD (primary aim) and whether they have potential as prognostic biomarkers in DKD (secondary aim). Methods iBEAt is a prospective multi-centre observational cohort study recruiting 500 patients with type 2 diabetes (T2D) and eGFR ≥30 ml/min/1.73m2. At baseline, blood and urine will be collected, clinical examinations will be performed, and medical history will be obtained. These assessments will be repeated annually for 3 years. At baseline each participant will also undergo quantitative renal MRI and US with central processing of MRI images. Biological samples will be stored in a central laboratory for biomarker and validation studies, and data in a central data depository. Data analysis will explore the potential associations between imaging biomarkers and renal function, and whether the imaging biomarkers improve the prediction of DKD progression. Ancillary substudies will: (1) validate imaging biomarkers against renal histopathology; (2) validate MRI based renal blood flow measurements against H2O15 positron-emission tomography (PET); (3) validate methods for (semi-)automated processing of renal MRI; (4) examine longitudinal changes in imaging biomarkers; (5) examine whether glycocalyx and microvascular measures are associated with imaging biomarkers and eGFR decline; (6) explore whether the findings in T2D can be extrapolated to type 1 diabetes. Discussion iBEAt is the largest DKD imaging study to date and will provide valuable insights into the progression and heterogeneity of DKD. The results may contribute to a more personalised approach to DKD management in patients with T2D. Trial registration Clinicaltrials.gov ( NCT03716401 ).
- Published
- 2020
- Full Text
- View/download PDF
3. Unbiased Human Kidney Tissue Proteomics Identifies Matrix Metalloproteinase 7 as a Kidney Disease Biomarker
- Author
-
Daigoro Hirohama, Amin Abedini, Salina Moon, Aditya Surapaneni, Simon T. Dillon, Allison Vassalotti, Hongbo Liu, Tomohito Doke, Victor Martinez, Zaipul Md Dom, Anil Karihaloo, Matthew B. Palmer, Josef Coresh, Morgan E. Grams, Monika A. Niewczas, and Katalin Susztak
- Subjects
Nephrology ,General Medicine - Published
- 2023
- Full Text
- View/download PDF
4. Spatially resolved human kidney multi-omics single cell atlas highlights the key role of the fibrotic microenvironment in kidney disease progression
- Author
-
Amin Abedini, Ziyuan Ma, Julia Frederick, Poonam Dhillon, Michael S. Balzer, Rojesh Shrestha, Hongbo Liu, Steven Vitale, Kishor Devalaraja-Narashimha, Paola Grandi, Tanmoy Bhattacharyya, Erding Hu, Steven S. Pullen, Carine M Boustany-Kari, Paolo Guarnieri, Anil Karihaloo, Hanying Yan, Kyle Coleman, Matthew Palmer, Lea Sarov-Blat, Lori Morton, Christopher A. Hunter, Mingyao Li, and Katalin Susztak
- Abstract
Kidneys have one of the most complex three-dimensional cellular organizations in the body, but the spatial molecular principles of kidney health and disease are poorly understood. Here we generate high-quality single cell (sc), single nuclear (sn), spatial (sp) RNA expression and sn open chromatin datasets for 73 samples, capturing half a million cells from healthy, diabetic, and hypertensive diseased human kidneys. Combining the sn/sc and sp RNA information, we identify > 100 cell types and states and successfully map them back to their spatial locations. Computational deconvolution of spRNA-seq identifies glomerular/vascular, tubular, immune, and fibrotic spatial microenvironments (FMEs). Although injured proximal tubule cells appear to be the nidus of fibrosis, we reveal the complex, heterogenous cellular and spatial organization of human FMEs, including the highly intricate and organized immune environment. We demonstrate the clinical utility of the FME spatial gene signature for the classification of a large number of human kidneys for disease severity and prognosis. We provide a comprehensive spatially-resolved molecular roadmap for the human kidney and the fibrotic process and demonstrate the clinical utility of spatial transcriptomics.
- Published
- 2022
- Full Text
- View/download PDF
5. Multimodal single cell sequencing of human diabetic kidney disease implicates chromatin accessibility and genetic background in disease progression
- Author
-
Parker C. Wilson, Yoshiharu Muto, Haojia Wu, Anil Karihaloo, Sushrut S. Waikar, and Benjamin D. Humphreys
- Abstract
Multimodal single cell sequencing is a powerful tool for interrogating cell-specific changes in transcription and chromatin accessibility. We performed single nucleus RNA (snRNA-seq) and assay for transposase accessible chromatin sequencing (snATAC-seq) on human kidney cortex from donors with and without diabetic kidney disease (DKD) to identify altered signaling pathways and transcription factors associated with DKD. Both snRNA-seq and snATAC-seq had an increased proportion ofVCAM1+injured proximal tubule cells (PT_VCAM1) in DKD samples. PT_VCAM1 has a pro-inflammatory expression signature and transcription factor motif enrichment implicated NFkB signaling. We used stratified linkage disequilibrium score regression to partition heritability of kidney-function-related traits using publicly-available GWAS summary statistics. Cell-specific PT_VCAM1 peaks were enriched for heritability of chronic kidney disease (CKD), suggesting that genetic background may regulate chromatin accessibility and DKD progression. snATAC-seq found cell-specific differentially accessible regions (DAR) throughout the nephron that change accessibility in DKD and these regions were enriched for glucocorticoid receptor (GR) motifs. Changes in chromatin accessibility were associated with decreased expression of insulin receptor, increased gluconeogenesis, and decreased expression of the GR cytosolic chaperone,FKBP5, in the diabetic proximal tubule. Cleavage under targets and release using nuclease (CUT&RUN) profiling of GR binding in bulk kidney cortex and anin vitromodel of the proximal tubule (RPTEC) showed that DAR co-localize with GR binding sites. CRISPRi silencing of GR response elements (GRE) in theFKBP5gene body reducedFKBP5expression in RPTEC, suggesting that reducedFKBP5chromatin accessibility in DKD may alter cellular response to GR. We developed an open-source tool for single cell allele specific analysis (SALSA) to model the effect of genetic background on gene expression. Heterozygous germline single nucleotide variants (SNV) in proximal tubule ATAC peaks were associated with allele-specific chromatin accessibility and differential expression of target genes within cis-coaccessibility networks. Partitioned heritability of proximal tubule ATAC peaks with a predicted allele-specific effect was enriched for eGFR, suggesting that genetic background may modify DKD progression in a cell-specific manner.
- Published
- 2022
- Full Text
- View/download PDF
6. Renal Pre-Competitive Consortium (RPC2): discovering therapeutic targets together
- Author
-
Kevin L. Duffin, Barbara Mirel, Anil Karihaloo, Uptal D. Patel, Matthias Kretzler, Carol Moreno Quinn, Maria Chiara Magnone, Heather Ascani, and Mark Tomilo
- Subjects
0301 basic medicine ,Pharmacology ,medicine.medical_specialty ,business.industry ,Systems biology ,Disease progression ,030232 urology & nephrology ,MEDLINE ,Disease ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug development ,Drug Discovery ,medicine ,Identification (biology) ,Intensive care medicine ,business ,Kidney disease ,Pharmaceutical industry - Abstract
Despite significant effort, patients with kidney disease have not seen their outcomes improved significantly over the past two decades. This has motivated clinicians and researchers to consider alternative methods to identifying risk factors, disease progression markers, and effective therapies. Genome-scale data sets from patients with renal disease can be used to establish a platform to improve understanding of the molecular basis of disease; however, such studies require expertise and resources. To overcome these challenges, we formed an academic-industry consortium to share molecular target identification efforts and expertise across academia and the pharmaceutical industry. The Renal Pre-Competitive Consortium (RPC2) aims to accelerate novel drug development for kidney diseases through a systems biology approach. Here, we describe the rationale, philosophy, establishment, and initial results of this strategy.
- Published
- 2018
- Full Text
- View/download PDF
7. Chemokine receptor Cxcr4 contributes to kidney fibrosis via multiple effectors
- Author
-
Gilbert W. Moeckel, Anil Karihaloo, Amy Yuan, Yashang Lee, and Uimook Choi
- Subjects
Male ,Benzylamines ,Receptors, CXCR4 ,Physiology ,Bone Morphogenetic Protein 7 ,Biology ,Cyclams ,Kidney ,CXCR4 ,Transforming Growth Factor beta1 ,Chemokine receptor ,Downregulation and upregulation ,Heterocyclic Compounds ,Fibrosis ,medicine ,Animals ,Nephrosclerosis ,urogenital system ,Articles ,Macrophage Activation ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Bone morphogenetic protein 7 ,medicine.anatomical_structure ,Immunology ,Corrigendum ,Ureteral Obstruction ,Kidney disease - Abstract
Kidney fibrosis is the final common pathway for virtually every type of chronic kidney disease and is a consequence of a prolonged healing response that follows tissue inflammation. Chronic kidney inflammation ultimately leads to progressive tissue injury and scarring/fibrosis. Several pathways have been implicated in the progression of kidney fibrosis. In the present study, we demonstrate that G protein-coupled chemokine (C-X-C motif) receptor (CXCR)4 was significantly upregulated after renal injury and that sustained activation of Cxcr4 expression augmented the fibrotic response. We demonstrate that after unilateral ureteral obstruction (UUO), both gene and protein expression of Cxcr4 were highly upregulated in tubular cells of the nephron. The increased Cxcr4 expression in tubules correlated with their increased dedifferentiated state, leading to increased mRNA expression of platelet-derived growth factor (PDGF)-α, transforming growth factor (TGF)-β1, and concurrent loss of bone morphogenetic protein 7 (Bmp7). Ablation of tubular Cxcr4 attenuated UUO-mediated fibrotic responses, which correlated with a significant reduction in PDGF-α and TGF-β1 levels and preservation of Bmp7 expression after UUO. Furthermore, Cxcr4+ immune cells infiltrated the obstructed kidney and further upregulate their Cxcr4 expression. Genetic ablation of Cxcr4 from macrophages was protective against UUO-induced fibrosis. There was also reduced total kidney TGF-β1, which correlated with reduced Smad activation and α-smooth muscle actin levels. We conclude that chronic high Cxcr4 expression in multiple effector cell types can contribute to the pathogenesis of renal fibrosis by altering their biological profile. This study uncovered a novel cross-talk between Cxcr4-TGF-β1 and Bmp7 pathways and may provide novel targets for interrupting the progression of fibrosis.
- Published
- 2015
- Full Text
- View/download PDF
8. Renal Pre-Competitive Consortium (RPC
- Author
-
Mark, Tomilo, Heather, Ascani, Barbara, Mirel, Maria Chiara, Magnone, Carol Moreno, Quinn, Anil, Karihaloo, Kevin, Duffin, Uptal D, Patel, and Matthias, Kretzler
- Subjects
Drug Development ,Drug Industry ,Risk Factors ,Drug Design ,Systems Biology ,Disease Progression ,Animals ,Humans ,Kidney Diseases ,Molecular Targeted Therapy ,Biomarkers - Abstract
Despite significant effort, patients with kidney disease have not seen their outcomes improved significantly over the past two decades. This has motivated clinicians and researchers to consider alternative methods to identifying risk factors, disease progression markers, and effective therapies. Genome-scale data sets from patients with renal disease can be used to establish a platform to improve understanding of the molecular basis of disease; however, such studies require expertise and resources. To overcome these challenges, we formed an academic-industry consortium to share molecular target identification efforts and expertise across academia and the pharmaceutical industry. The Renal Pre-Competitive Consortium (RPC
- Published
- 2018
9. Semaphorin 7A in circulating regulatory T cells is increased in autosomal-dominant polycystic kidney disease and decreases with tolvaptan treatment
- Author
-
Jonathan Kaufman Scher, Madeline L. Droher, Katrina Lehmann Blount, Yashang Lee, Gilbert W. Moeckel, Erica L. Herzog, Feng Dai, Sherrie Bitterman, Siobhan Thompson, Neera K. Dahl, and Anil Karihaloo
- Subjects
0301 basic medicine ,Nephrology ,Male ,medicine.medical_specialty ,Physiology ,Tolvaptan ,Autosomal dominant polycystic kidney disease ,Semaphorins ,030204 cardiovascular system & hematology ,urologic and male genital diseases ,GPI-Linked Proteins ,Kidney ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigens, CD ,Physiology (medical) ,Internal medicine ,medicine ,Polycystic kidney disease ,Renal fibrosis ,Animals ,Humans ,IL-2 receptor ,urogenital system ,business.industry ,FOXP3 ,Middle Aged ,medicine.disease ,Polycystic Kidney, Autosomal Dominant ,female genital diseases and pregnancy complications ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Leukocytes, Mononuclear ,Female ,business ,Antidiuretic Hormone Receptor Antagonists ,medicine.drug - Abstract
Semaphorin 7A (SEMA7A) is an immunomodulating protein implicated in lung and liver fibrosis. In autosomal-dominant polycystic kidney disease (ADPKD), the progressive expansion of renal cysts, inflammation, and subsequent renal fibrosis leads to end-stage renal disease (ESRD). SEMA7A may play a role in renal fibrosis and in ADPKD. We evaluated Sema7a in a mouse model of renal fibrosis and determined the expression of SEMA7A in human ADPKD kidney. We analyzed SEMA7A expression on peripheral blood mononuclear cells (PBMCs), including CD45+ (leukocyte), CD14+(monocyte), CD4+ (T lymphocytes) and CD4+Foxp3+CD25+ [regulatory T lymphocytes (Tregs)] from 90 ADPKD patients (11 tolvaptan treated and 79 tolvaptan naive), and 21 healthy volunteers, using a Fluorescence-Activated Cell Sorting (FACS). Sema7a is required for renal fibrosis. SEMA7A shows robust expression in ADPKD kidneys, localizing to cysts derived from distal tubules. SEMA7A is higher in circulating monocytes, but unchanged in CD4+ lymphocytes in ADPKD patients. The SEMA7A increase was detected early (stage 1 CKD) and seemed more prominent in patients with smaller kidneys (p = 0.09). Compared to tolvaptan-naive ADPKD patients, those treated with tolvaptan showed reduced SEMA7A expression on monocytes, T lymphocytes, and Tregs, although the number of PBMCs was unchanged. After 1 month of tolvaptan treatment, SEMA7A expression on Tregs decreased. SEMA7A shows potential as both a therapeutic target in mammalian kidney fibrosis and as a marker of inflammation in ADPKD patients. SEMA7A expression was lower after tolvaptan treatment, which may reflect drug efficacy.
- Published
- 2017
10. Macrophages Promote Cyst Growth in Polycystic Kidney Disease
- Author
-
David Merrick, Yashang Lee, Lloyd G. Cantley, Michael J. Caplan, Stefan Somlo, Farrukh M. Koraishy, Anil Karihaloo, and Sarah C. Huen
- Subjects
Pathology ,medicine.medical_specialty ,Chemokine CXCL6 ,Inflammation ,Biology ,Cell Line ,Mice ,Cell Movement ,medicine ,Polycystic kidney disease ,Animals ,Antigens, Ly ,Cyst ,Chemokine CCL2 ,CXCL16 ,Cell Proliferation ,PKD1 ,Cell growth ,Macrophages ,Chemokine CXCL16 ,General Medicine ,Polycystic Kidney, Autosomal Dominant ,medicine.disease ,Mice, Inbred C57BL ,Nephrology ,Cell culture ,medicine.symptom ,Brief Communications ,Homing (hematopoietic) - Abstract
Polycystic kidney disease (PKD) exhibits an inflammatory component, but the contribution of inflammation to cyst progression is unknown. Macrophages promote the proliferation of tubular cells following ischemic injury, suggesting that they may have a role in cystogenesis. Furthermore, cultured Pkd1-deficient cells express the macrophage chemoattractants Mcp1 and Cxcl16 and stimulate macrophage migration. Here, in orthologous models of both PKD1 and PKD2, abnormally large numbers of alternatively activated macrophages surrounded the cysts. To determine whether pericystic macrophages contribute to the proliferation of cyst-lining cells, we depleted phagocytic cells from Pkd1(fl/fl);Pkhd1-Cre mice by treating with liposomal clodronate from postnatal day 10 until day 24. Compared with vehicle-treated controls, macrophage-depleted mice had a significantly lower cystic index, reduced proliferation of cyst-lining cells, better-preserved renal parenchyma, and improved renal function. In conclusion, these data suggest that macrophages home to cystic areas and contribute to cyst growth. Interruption of these homing and proliferative signals could have therapeutic potential for PKD.
- Published
- 2011
- Full Text
- View/download PDF
11. Activating AMP-activated protein kinase (AMPK) slows renal cystogenesis
- Author
-
Michael J. Caplan, Anil Karihaloo, Patricia Seo-Mayer, Hui Li, Stefan Somlo, Vinita Takiar, Saori Nishio, Kenneth R. Hallows, Li Zhang, and J Darwin King
- Subjects
medicine.medical_specialty ,Autosomal dominant polycystic kidney disease ,Cystic Fibrosis Transmembrane Conductance Regulator ,Mice, Transgenic ,AMP-Activated Protein Kinases ,Biology ,Cell Line ,Mice ,Dogs ,AMP-activated protein kinase ,Internal medicine ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Protein kinase A ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Multidisciplinary ,TOR Serine-Threonine Kinases ,AMPK ,Epithelial Cells ,Biological Sciences ,Polycystic Kidney, Autosomal Dominant ,medicine.disease ,Metformin ,Cystic fibrosis transmembrane conductance regulator ,Disease Models, Animal ,Endocrinology ,biology.protein ,Cancer research ,Chloride channel ,medicine.drug - Abstract
Renal cyst development and expansion in autosomal dominant polycystic kidney disease (ADPKD) involves both fluid secretion and abnormal proliferation of cyst-lining epithelial cells. The chloride channel of the cystic fibrosis transmembrane conductance regulator (CFTR) participates in secretion of cyst fluid, and the mammalian target of rapamycin (mTOR) pathway may drive proliferation of cyst epithelial cells. CFTR and mTOR are both negatively regulated by AMP-activated protein kinase (AMPK). Metformin, a drug in wide clinical use, is a pharmacological activator of AMPK. We find that metformin stimulates AMPK, resulting in inhibition of both CFTR and the mTOR pathways. Metformin induces significant arrest of cystic growth in both in vitro and ex vivo models of renal cystogenesis. In addition, metformin administration produces a significant decrease in the cystic index in two mouse models of ADPKD. Our results suggest a possible role for AMPK activation in slowing renal cystogenesis as well as the potential for therapeutic application of metformin in the context of ADPKD.
- Published
- 2011
- Full Text
- View/download PDF
12. Neutrophil gelatinase-associated lipocalin suppresses cyst growth by Pkd1 null cells in vitro and in vivo
- Author
-
Sekiya Shibazaki, Lloyd G. Cantley, Vikas P. Sukhatme, Pankaj Seth, Arnaud Marlier, Tong Wang, Feng Wei, Stefan Somlo, Anil Karihaloo, and Zhiheng Yu
- Subjects
Programmed cell death ,medicine.medical_specialty ,TRPP Cation Channels ,proliferation ,030232 urology & nephrology ,cysts ,Biology ,Lipocalin ,Protein Serine-Threonine Kinases ,Article ,Kidney Tubules, Proximal ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Lipocalin-2 ,Internal medicine ,Null cell ,medicine ,Polycystic kidney disease ,Animals ,Cyst ,NGAL ,030304 developmental biology ,Cell Proliferation ,Oncogene Proteins ,0303 health sciences ,Polycystic Kidney Diseases ,Cell growth ,Pkd ,apoptosis ,Pyruvate Dehydrogenase Acetyl-Transferring Kinase ,Epithelial Cells ,medicine.disease ,tubulogenesis ,Lipocalins ,Cell biology ,Endocrinology ,Cell culture ,Apoptosis ,Nephrology ,Acute-Phase Proteins - Abstract
Cyst growth in patients with autosomal dominant polycystic kidney disease is thought to be due to increased tubular cell proliferation. One model to explain this altered proliferation suggests that the polycystin proteins PC1 and PC2 localize to apical cilia and serve as an integral part of the flow-sensing pathway thus modulating the proliferative response. We measured proliferation and apoptosis in proximal tubule derived cell lines lacking PC1. These cells showed increased rates of proliferation, a decreased rate of apoptosis, compared to control heterozygous cell lines, and spontaneously formed cysts rather than tubules in an in vitro tubulogenesis assay. Addition of neutrophil gelatinase associated lipocalin (NGAL), a small secreted protein that binds diverse ligands, to the cells lacking PC1 inhibited proliferation and increased apoptosis leading to slower cyst growth in vitro. Sustained over-expression at low level of NGAL by an adenoviral delivery system suppressed cyst enlargement without improving renal function in the Pkd1 mutant mice. Our studies show that renal epithelial cells lacking PC1 have an inherent tendency to hyper-proliferate forming cysts in vitro independent of a flow stimulus. The potential benefit of attenuating cyst growth with NGAL remains to be determined.
- Published
- 2008
- Full Text
- View/download PDF
13. VEGF Attenuates Hyperoxic Injury through Decreased Apoptosis in Explanted Rat Embryonic Lung
- Author
-
Alia Bazzy-Asaad, Americo E. Esquibies, Anil Karihaloo, Lloyd G. Cantley, Hitoshi Nishio, and Farshid Ghassemi
- Subjects
Vascular Endothelial Growth Factor A ,Programmed cell death ,Time Factors ,Down-Regulation ,Apoptosis ,Hyperoxia ,Biology ,Rats, Sprague-Dawley ,Tissue Culture Techniques ,Andrology ,chemistry.chemical_compound ,In vivo ,Morphogenesis ,medicine ,Animals ,RNA, Messenger ,Lung ,Cell Proliferation ,Gene Expression Regulation, Developmental ,respiratory system ,Hypoxia (medical) ,Rats ,respiratory tract diseases ,Vascular endothelial growth factor ,medicine.anatomical_structure ,chemistry ,Pediatrics, Perinatology and Child Health ,Immunology ,Lung morphogenesis ,medicine.symptom - Abstract
Ambient oxygen concentration and vascular endothelial growth factor (VEGF)-A are vital in lung development. Since hypoxia stimulates VEGF-A production and hyperoxia reduces it, we hypothesized that VEGF-A down-regulation by exposure of airways to hyperoxia may result in abnormal lung development. An established model of in vitro rat lung development was used to examine the effects of hyperoxia on embryonic lung morphogenesis and VEGF-A expression. Under physiologic conditions, lung explant growth and branching is similar to that seen in vivo. However, in hyperoxia (50% O2) the number of terminal buds and branch length was significantly reduced after 4 d of culture. This effect correlated with a significant increase in cellular apoptosis and decrease in proliferation compared with culture under physiologic conditions. mRNA for Vegf164 and Vegf188 was reduced during hyperoxia and addition of VEGF165, but not VEGF121, to explants grown in 50% O2 resulted in partial reversal of the decrease in lung branching, correlating with a decrease in cell apoptosis. Thus, hyperoxia suppresses VEGF-A expression and inhibits airway growth and branching. The ability of exogenous VEGF165 to partially reverse apoptotic effects suggests this may be a potential approach for the prevention of hyperoxic injury.
- Published
- 2008
- Full Text
- View/download PDF
14. Polycystin-2 Regulates Proliferation and Branching Morphogenesis in Kidney Epithelial Cells
- Author
-
David H. Grimm, Yiqiang Cai, Anil Karihaloo, Michael J. Caplan, Stefan Somlo, and Lloyd G. Cantley
- Subjects
TRPP Cation Channels ,Mutant ,Morphogenesis ,Autosomal dominant polycystic kidney disease ,Gene Expression ,Biology ,urologic and male genital diseases ,Biochemistry ,Cell Line ,medicine ,Humans ,Extracellular Signal-Regulated MAP Kinases ,education ,Molecular Biology ,education.field_of_study ,Epidermal Growth Factor ,PKD1 ,Cysts ,Hepatocyte Growth Factor ,urogenital system ,Cell growth ,Kinase ,Membrane Proteins ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Polycystic Kidney, Autosomal Dominant ,medicine.disease ,female genital diseases and pregnancy complications ,Cell biology ,Kidney Tubules ,Polycystin 2 ,Cell culture ,embryonic structures ,Cell Division - Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation of multiple fluid-filled cysts that expand over time and destroy the renal architecture. Loss or mutation of polycystin-1 or polycystin-2, the respective proteins encoded by the ADPKD genes PKD1 and PKD2, is associated with most cases of ADPKD. Thus, the polycystin proteins likely play a role in cell proliferation and morphogenesis. Recent studies indicate that polycystin-1 is involved in these processes, but little is known about the role played by polycystin-2. To address this question, we created a number of related cell lines variable in their expression of polycystin-2. We show that the basal and epidermal growth factor-stimulated rate of cell proliferation is higher in cells that do not express polycystin-2 versus those that do, indicating that polycystin-2 acts as a negative regulator of cell growth. In addition, cells not expressing polycystin-2 exhibit significantly more branching morphogenesis and multicellular tubule formation under basal and hepatocyte growth factor-stimulated conditions than their polycystin-2-expressing counterparts, suggesting that polycystin-2 may also play an important role in the regulation of tubulogenesis. Cells expressing a channel mutant of polycystin-2 proliferated faster than those expressing the wild-type protein, but exhibited blunted tubule formation. Thus, the channel activity of polycystin-2 may be an important component of its regulatory machinery. Finally, we show that polycystin-2 regulation of cell proliferation appears to be dependent on its ability to prevent phosphorylated extracellular-related kinase from entering the nucleus. Our results indicate that polycystin-2 is necessary for the proper growth and differentiation of kidney epithelial cells and suggest a possible mechanism for the cyst formation seen in ADPKD2.
- Published
- 2006
- Full Text
- View/download PDF
15. Vascular Endothelial Growth Factor Induces Branching Morphogenesis/Tubulogenesis in Renal Epithelial Cells in a Neuropilin-Dependent Fashion
- Author
-
Sujata Kale, Lloyd G. Cantley, Shivalingappa Venkatesha, Anil Karihaloo, S. Ananth Karumanchi, and William L. Cantley
- Subjects
Vascular Endothelial Growth Factor A ,Morphogenesis ,Biology ,Kidney ,Endothelial cell differentiation ,Cell Line ,Protein kinase C signaling ,Mice ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Vasculogenesis ,Cell Movement ,Neuropilin ,Animals ,Humans ,Protein Isoforms ,Neuropilins ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Protein Kinase C ,Tube formation ,Wound Healing ,Epithelial Cells ,Cell Biology ,Cell biology ,Enzyme Activation ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Signal Transduction - Abstract
Vascular endothelial growth factor (VEGF) is well characterized for its role in endothelial cell differentiation and vascular tube formation. Alternate splicing of the VEGF gene in mice results in various VEGF-A isoforms, including VEGF-121 and VEGF-165. VEGF-165 is the most abundant isoform in the kidney and has been implicated in glomerulogenesis. However, its role in the tubular epithelium is not known. We demonstrate that VEGF-165 but not VEGF-121 induces single-cell branching morphogenesis and multicellular tubulogenesis in mouse renal tubular epithelial cells and that these morphogenic effects require activation of the phosphatidylinositol 3-kinase (PI 3-K) and, to a lesser degree, the extracellular signal-regulated kinase and protein kinase C signaling pathways. Further, VEGF-165-stimulated sheet migration is dependent only on PI 3-K signaling. These morphogenic effects of VEGF-165 require activation of both VEGF receptor 2 (VEGFR-2) and neuropilin-1 (Nrp-1), since neutralizing antibodies to either of these receptors or the addition of semaphorin 3A (which blocks VEGF-165 binding to Nrp-1) prevents the morphogenic response and the phosphorylation of VEGFR-2 along with the downstream signaling. We thus conclude that in addition to endothelial vasculogenesis, VEGF can induce renal epithelial cell morphogenesis in a Nrp-1-dependent fashion.
- Published
- 2005
- Full Text
- View/download PDF
16. Signals Which Build a Tubule
- Author
-
Christian H. Nickel, Lloyd G. Cantley, and Anil Karihaloo
- Subjects
Kidney ,Cell signaling ,Lung ,urogenital system ,Physiology ,Mammary gland ,Cell Communication ,General Medicine ,Biology ,Cell biology ,Kidney Tubules ,medicine.anatomical_structure ,Tubule ,Cell Movement ,Nephrology ,Branching morphogenesis ,Ureteric bud ,Genetics ,medicine ,Humans ,Growth Substances ,Process (anatomy) ,Cell Proliferation ,Signal Transduction - Abstract
The phenomenon of branching morphogenesis is a fundamental process critical for development of several tubular organs including lung, mammary gland, and kidney. In the case of kidney, the ureteric bud (UB) that extends out from a pre-existing epithelial tube, the Wolffian duct, gives rise to the branched collecting duct system while the surrounding metanephric mesenchyme undergoes mesenchymal-epithelial transition to form the proximal parts of the nephron. These events are mediated by several soluble factors that act in a cooperative fashion either as pro or anti tubulogenic factors. Among the growing list of such molecules are the members of the FGF, TGF-β, and Wnt families as well as GDNF, HGF, and EGF. Cells respond to these soluble factors by initiating signaling pathways that regulate cell proliferation, cell migration and cell morphogenesis. These signaling pathways are also regulated in parallel by cell-cell and cell-matrix interactions, leading to the complex events necessary for tubule formation. Recent in-vitro and in-vivo studies have begun to shed light on the overall regulation of this phenomenon while the specific subcellular mechanisms are only beginning to be understood. This review focuses on our understanding of the morphogenic responses that regulate in-vitro tubulogenesis and how they may help us to ultimately understand this process in vivo in the kidney.
- Published
- 2005
- Full Text
- View/download PDF
17. Bone marrow stem cells contribute to repair of the ischemically injured renal tubule
- Author
-
Paul R. Clark, Sujata Kale, Anil Karihaloo, Michael Kashgarian, Lloyd G. Cantley, and Diane S. Krause
- Subjects
Pathology ,medicine.medical_specialty ,Bone Marrow Cells ,Article ,Mice ,Bone Marrow Ablation ,Ischemia ,medicine ,Animals ,Renal stem cell ,Acute tubular necrosis ,Bone Marrow Transplantation ,Stem cell transplantation for articular cartilage repair ,business.industry ,Stem Cells ,Bone Marrow Stem Cell ,General Medicine ,Kidney Tubular Necrosis, Acute ,medicine.disease ,Mice, Inbred C57BL ,Kidney Tubules ,medicine.anatomical_structure ,Immunology ,Bone marrow ,Stem cell ,business ,Adult stem cell - Abstract
The paradigm for recovery of the renal tubule from acute tubular necrosis is that surviving cells from the areas bordering the injury must migrate into the regions of tubular denudation and proliferate to re-establish the normal tubular epithelium. However, therapies aimed at stimulating these events have failed to alter the course of acute renal failure in human trials. In the present study, we demonstrate that Lin-Sca-1+ cells from the adult mouse bone marrow are mobilized into the circulation by transient renal ischemia and home specifically to injured regions of the renal tubule. There they differentiate into renal tubular epithelial cells and appear to constitute the majority of the cells present in the previously necrotic tubules. Loss of stem cells following bone marrow ablation results in a greater rise in blood urea nitrogen after renal ischemia, while stem cell infusion after bone marrow ablation reverses this effect. Thus, therapies aimed at enhancing the mobilization, propagation, and/or delivery of bone marrow stem cells to the kidney hold potential as entirely new approaches for the treatment of acute tubular necrosis.
- Published
- 2003
- Full Text
- View/download PDF
18. Effect of Sorbinil and Ascorbic Acid on myo-Inositol Transport in Cultured Rat Schwann Cells Exposed to Elevated Extracellular Glucose
- Author
-
Kusum Joshi, Anil Karihaloo, and Jagjit S. Chopra
- Subjects
medicine.medical_specialty ,Myo-inositol transport ,Ascorbic Acid ,Biology ,Imidazolidines ,Biochemistry ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Aldehyde Reductase ,Internal medicine ,medicine ,Animals ,Inositol ,Enzyme Inhibitors ,Rats, Wistar ,Cells, Cultured ,Phospholipids ,Aldose reductase ,Imidazoles ,Biological Transport ,Metabolism ,Ascorbic acid ,Sciatic Nerve ,Aldose reductase inhibitor ,Rats ,carbohydrates (lipids) ,Kinetics ,Glucose ,Endocrinology ,Animals, Newborn ,chemistry ,Sorbitol ,Sorbinil ,Schwann Cells ,medicine.drug - Abstract
The effect of long-term (2 weeks) exposure to 0-50 mM glucose and 0-1 mM sorbitol on myo-inositol metabolism was studied in cultured rat Schwann cells. Experiments were carried out to determine the effect of sorbinil and ascorbic acid on myo-inositol uptake in rat Schwann cells cultured in the presence of increased extracellular glucose or sorbitol. myo-Inositol uptake and its incorporation into phospholipids decreased significantly when cells were grown in > or = 30 mM glucose for a period of 2 weeks. This inhibitory effect was partly blocked by sorbinil, an aldose reductase inhibitor, in a dose-dependent fashion. Significant prevention was achieved with 0.5 and 1 mM sorbinil. Ascorbic acid also prevented the reduction in myo-inositol uptake due to excess extracellular glucose, at 3 and 30 microM concentrations, but not at 300 microM. Neither sorbinil nor ascorbic acid could prevent the alterations in myo-inositol transport in cells exposed to high sorbitol levels for the same period of time. These data suggest that glucose-induced alteration of myo-inositol transport in Schwann cells is mediated, at least in part, via sorbitol accumulation. This myo-inositol transport impairment is prevented by sorbinil and also by ascorbic acid. Ascorbic acid may hold a fresh promise for the treatment/prevention of diabetic neuropathy/complications, at least as an adjunct therapy along with known aldose reductase inhibitors.
- Published
- 2002
- Full Text
- View/download PDF
19. MP071MITOCHONDRIAL FRAGMENTATION AND ELEVATED LEVELS OF MIOX EXPRESSION IS NOT OBSERVED IN HUMAN CKD
- Author
-
Magnus Söderberg, Mårten Hammar, Anette Ericsson, Birgitta Rosengren, Kjell Hultenby, Ann-Katrin Andersson, and Anil Karihaloo
- Subjects
Transplantation ,Nephrology ,business.industry ,Medicine ,Fragmentation (cell biology) ,business ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
20. Differential MAPK Pathways Utilized for HGF- and EGF-dependent Renal Epithelial Morphogenesis
- Author
-
Katherine Spokes, Dawn A. O’Rourke, Anil Karihaloo, Christian H. Nickel, and Lloyd G. Cantley
- Subjects
MAPK/ERK pathway ,Epidermal Growth Factor ,Hepatocyte Growth Factor ,Cell morphogenesis ,Morphogenesis ,Cell Biology ,Biology ,Mitogen-activated protein kinase kinase ,Kidney ,Biochemistry ,Molecular biology ,Cell biology ,Cell Movement ,Epidermal growth factor ,medicine ,Hepatocyte growth factor ,Mitogen-Activated Protein Kinases ,Signal transduction ,Protein kinase A ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Cell Line, Transformed ,medicine.drug - Abstract
Cells derived from the inner medullary collecting duct undergo in vitro branching tubulogenesis to both the c-met receptor ligand hepatocyte growth factor (HGF) as well as epidermal growth factor (EGF) receptor ligands. In contrast, many other cultured renal epithelial cells respond in this manner only to HGF, suggesting that these two receptors may use independent signaling pathways during morphogenesis. We have therefore compared the signaling pathways for mIMCD-3 cell morphogenesis in response to EGF and HGF. Inhibition of the p42/44 mitogen-activated protein kinase (MAPK) pathway with the mitogen-activated protein kinase kinase (MKK1) inhibitor PD98059 (50 μm) markedly inhibits HGF-induced cell migration with only partial inhibition of EGF-induced cell motility. Similarly, HGF-dependent, but not EGF-dependent, branching morphogenesis was more greatly inhibited by the MKK1 inhibitor. Examination of EGF-stimulated cells demonstrated that extracellular-regulated kinase 5 (ERK5) was activated in response to EGF but not HGF, and that activation of ERK5 was only 60% inhibited by 50 μm PD98059. In contrast, the MKK inhibitor U0126 markedly inhibited both ERK1/2 and ERK5 activation and completely prevented HGF- and EGF-dependent migration and branching process formation. Expression of dominant negative ERK5 (dnBMK1) likewise inhibited EGF-dependent branching process formation, but did not affect HGF-dependent branching process formation. Our results indicate that activation of the ERK1/ERK2 signaling pathway is critical for HGF-induced cell motility/morphogenesis in mIMCD-3 cells, whereas ERK5 appears to be required for EGF-dependent morphogenesis.
- Published
- 2001
- Full Text
- View/download PDF
21. Protein kinase and Ca2+ modulation of myo-inositol transport in cultured retinal pigment epithelial cells
- Author
-
Anil Karihaloo, Koichi Kato, Douglas A. Greene, and T. P. Thomas
- Subjects
Physiology ,Myo-inositol transport ,Biology ,Phosphatidylinositols ,Muscarinic agonist ,chemistry.chemical_compound ,Cytosol ,Humans ,Inositol ,Pigment Epithelium of Eye ,Protein kinase A ,Cells, Cultured ,Heat-Shock Proteins ,Protein Kinase C ,Protein kinase C ,Symporters ,Kinase ,Membrane Proteins ,Biological Transport ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Molecular biology ,Hypotonic Solutions ,Biochemistry ,chemistry ,Calcium ,Efflux ,Carrier Proteins ,Cotransporter - Abstract
The acute regulation of inwardly directed Na(+)-myo-inositol (MI) cotransporter activity and basal and volume-sensitive MI efflux by protein kinases C (PKC) and A (PKA), cytosolic Ca2+, and phosphoinositide (PI) turnover were characterized in cultured human retinal pigment epithelial cells using 2-[3H]MI and liquid scintillation spectrometry. Kinetic analysis revealed two distinct Na(+)-MI cotransporter components differing in apparent Michaelis constant and maximal velocity. Composite Na(+)-MI cotransport activity was stimulated by PKA activation, the muscarinic agonist carbachol, and the Ca2+ ionophore A-23187 and was inhibited by PKC activation. PKC activation also increased MI efflux, but only the volume-sensitive component, whereas PKA activation increased both basal and volume-sensitive MI efflux. These studies implicate PKC as a negative modulator of MI content through Na(+)-MI cotransport inhibition and potentiation of volume-sensitive MI efflux. PKA is a positive modulator of both Na(+)-MI cotransport and basal and volume-sensitive MI efflux. Cytosolic Ca2+ release through receptor-mediated PI hydrolysis may facilitate Na(+)-MI cotransport activity.
- Published
- 1997
- Full Text
- View/download PDF
22. Met and the epidermal growth factor receptor act cooperatively to regulate final nephron number and maintain collecting duct morphology
- Author
-
Shuta Ishibe, Snorri S. Thorgeirsson, Roland Schmitt, Hong Ma, Arnaud Marlier, Michael Kashgarian, Anil Karihaloo, Junhui Zhang, David S. Geller, Jan Czyczk, Akashi Togawa, Mitchihiro Mitobe, and Lloyd G. Cantley
- Subjects
Male ,medicine.medical_specialty ,Receptor expression ,Kidney development ,Nephron ,Biology ,Kidney ,Mice ,Pregnancy ,Internal medicine ,medicine ,Animals ,Epidermal growth factor receptor ,RNA, Messenger ,Renal Insufficiency ,Kidney Tubules, Collecting ,Receptor ,Molecular Biology ,DNA Primers ,Mice, Knockout ,Base Sequence ,urogenital system ,Nephrons ,Proto-Oncogene Proteins c-met ,Mice, Mutant Strains ,Cell biology ,ErbB Receptors ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Ureteric bud ,biology.protein ,Hepatocyte growth factor ,Female ,Development and Disease ,Ureter ,Developmental Biology ,medicine.drug ,Signal Transduction - Abstract
Ureteric bud (UB) branching during kidney development determines the final number of nephrons. Although hepatocyte growth factor and its receptor Met have been shown to stimulate branching morphogenesis in explanted embryonic kidneys, loss of Met expression is lethal during early embryogenesis without obvious kidney abnormalities. Metfl/fl;HoxB7-Cre mice,which lack Met expression selectively in the UB, were generated and found to have a reduction in final nephron number. These mice have increased Egf receptor expression in both the embryonic and adult kidney, and exogenous Egf can partially rescue the branching defect seen in kidney explants. Metfl/fl;HoxB7-Cre;wa-2/wa-2 mice, which lack normal Egfr and Met signaling, exhibit small kidneys with a marked decrease in UB branching at E14.5 as well as a reduction in final glomerular number. These mice developed progressive interstitial fibrosis surrounding collecting ducts with kidney failure and death by 3-4 weeks of age. Thus, in support of previous in vitro findings, Met and the Egf receptor can act cooperatively to regulate UB branching and mediate maintenance of the normal adult collecting duct.
- Published
- 2009
23. Vegf as an epithelial cell morphogen modulates branching morphogenesis of embryonic kidney by directly acting on the ureteric bud
- Author
-
Arnaud Marlier, Kai M. Schmidt-Ott, Anil Karihaloo, Jonathan Barasch, and Anna Rachel Gallagher
- Subjects
Vascular Endothelial Growth Factor A ,Embryology ,medicine.medical_specialty ,Mesenchyme ,Biology ,Kidney ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Mice ,Organ Culture Techniques ,Internal medicine ,medicine ,Morphogenesis ,Animals ,Laser capture microdissection ,Epithelial Cells ,Embryonic stem cell ,Vascular Endothelial Growth Factor Receptor-2 ,Epithelium ,Cell biology ,Rats ,Vascular endothelial growth factor ,Endocrinology ,medicine.anatomical_structure ,Kidney Tubules ,chemistry ,Ureteric bud ,Ureter ,Developmental Biology ,Morphogen - Abstract
There is growing evidence that vascular endothelial growth factor (Vegf), a well-recognized angiogenic factor, plays a regulatory role in non-endothelial tissues such as neurons and epithelial cells. In the kidney Vegf receptors have been detected in proximal tubule cells of the adult kidney and Vegf has been show to stimulate branching morphogenesis of the developing kidney. In this study, using laser-microdissection as well as manual separation of the UB, we demonstrate that Vegf receptors are present in the ureteric bud (UB). Furthermore, we determine that Vegf stimulates UB branching in whole kidney explant that is mediated directly by signaling through Vegfr2. In addition, Vegf also induced branching response in isolated UBs that are free of the surrounding mesenchyme. These responses seem to be strictly dependent on the dose of Vegf such that higher doses are inhibitory while lower dose are stimulatory. These data place Vegf in a unique position of being able to modulate vascular as well as epithelial development in the embryonic kidney.
- Published
- 2008
24. Vascular endothelial growth factor (VEGF) isoform attenuates hyperoxic injury of embryonic lung via a Flk1–neuropilin (Nrp)-1–protein kinase C (PKC) dependent pathway
- Author
-
Lloyd G. Cantley, Alia Bazzy-Asaad, Americo E. Esquibies, and Anil Karihaloo
- Subjects
Pulmonary and Respiratory Medicine ,Gene isoform ,Lung ,biology ,business.industry ,VEGF receptors ,Embryonic stem cell ,Cell biology ,Vascular endothelial growth factor ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Pediatrics, Perinatology and Child Health ,medicine ,Neuropilin ,biology.protein ,business ,Protein kinase C - Published
- 2013
- Full Text
- View/download PDF
25. Microarray analysis of in vitro pericyte differentiation reveals an angiogenic program of gene expression
- Author
-
Gary R. Grotendorst, Anil Karihaloo, Jun-ichi Hanai, Vikas P. Sukhatme, Barden Chan, Sujata Kale, and Lloyd G. Cantley
- Subjects
Cell type ,Umbilical Veins ,Angiogenesis ,Neovascularization, Physiologic ,Biology ,Biochemistry ,Cell Line ,Mice ,Vasculogenesis ,Gene expression ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Mice, Inbred C3H ,Microarray analysis techniques ,Gene Expression Profiling ,Multipotent Stem Cells ,Cell Differentiation ,Embryo, Mammalian ,Microarray Analysis ,In vitro ,Coculture Techniques ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Genes ,Normal blood ,Pericyte ,Pericytes ,Biotechnology - Abstract
The vasculature consists of endothelial cells (ECs) lined by pericyte/vascular smooth muscle cells (vSMCs). Pericyte/vSMCs provide support to the mature vasculature but are also essential for normal blood vessel development. To determine how pericyte-EC communication influences vascular development, we used the well-established in vitro model of TGFbeta-stimulated differentiation of 10T1/2 cells into pericyte/vSMCs. Microarray analysis was performed to identify genes that were differentially expressed by induced vs. uninduced 10T1/2 cells. We discovered that these cells show an angiogenic program of gene expression, with up-regulation of several genes previously implicated in angiogenesis, including VEGF, IL-6, VEGF-C, HB-EGF, CTGF, tenascin C, integrin alpha5, and Eph receptor A2. Up-regulation of some genes was validated by Western blots and immunocytochemistry. We also examined the functional significance of these gene expression changes. VEGF and IL-6 alone and in combination were important in 10T1/2 cell differentiation. Furthermore, we used a coculture system of 10T1/2 and human umbilical vein ECs (HUVECs), resulting in the formation of cordlike structures by the HUVECs. This cordlike structure formation was disrupted when neutralizing antibodies to VEGF or IL-6 were added to the coculture system. The results of these studies show that factors produced by pericytes may be responsible for recruiting ECs and promoting angiogenesis. Therefore, a further understanding of the genes involved in pericyte differentiation could provide a novel approach for developing anti-angiogenic therapies.
- Published
- 2004
26. Endostatin regulates branching morphogenesis of renal epithelial cells and ureteric bud
- Author
-
Jun Yang, Vikas P. Sukhatme, Anil Karihaloo, Lloyd G. Cantley, Silviu Grisaru, Jonathan Barasch, Vivekanand Jha, S. Ananth Karumanchi, Norman D. Rosenblum, Kevin T. Bush, Christian H. Nickel, and Sanjay K. Nigam
- Subjects
Epithelial cell morphogenesis ,Morphogenesis ,Biology ,Kidney ,Cell Line ,Mice ,Glypicans ,Epidermal growth factor ,Cell Movement ,medicine ,Animals ,Multidisciplinary ,Epidermal Growth Factor ,Hepatocyte Growth Factor ,Biological Sciences ,Peptide Fragments ,Cell biology ,Endostatins ,Rats ,Endothelial stem cell ,Cell culture ,Ureteric bud ,Hepatocyte growth factor ,Collagen ,Endostatin ,Ureter ,Heparan Sulfate Proteoglycans ,medicine.drug - Abstract
Endostatin (ES) inhibits endothelial cell migration and has been found to bind to glypicans (Gpcs) on both endothelial cells and renal epithelial cells. We examined the possibility that ES might regulate epithelial cell morphogenesis. The addition of ES to cultured epithelial cells causes an inhibition of both hepatocyte growth factor- and epidermal growth factor-dependent process formation and migration. In contrast, ES does not inhibit epidermal growth factor-dependent morphogenesis in renal epithelial cells derived from Gpc-3 −/mice, whereas expression of Gpc-1 in these cells reconstitutes ES responsiveness. Gpc-3 −/mice have been shown to display enhanced ureteric bud (UB) branching early in development, and cultured UB cells release ES into the media, suggesting that ES binding to Gpcs may regulate UB branching. The addition of ES inhibits branching of the explanted UB, whereas a neutralizing Ab to ES enhances UB outgrowth and branching. Thus, local expression of ES at the tips of the UB may play a role in the regulation of UB arborization.
- Published
- 2001
27. Cell surface glypicans are low-affinity endostatin receptors
- Author
-
Arthur D. Lander, Jun-ichi Hanai, Ganesh Venkataraman, Zachary Shriver, Yu Yamaguchi, S. Ananth Karumanchi, Mohanraj Dhanabal, Raghu Kalluri, Debabrata Mukhopadhyay, Leonidas Tsiokas, Anil Karihaloo, Kazuki Hagihara, Ram Sasisekharan, Lloyd G. Cantley, Robert L. Chen, Vikas P. Sukhatme, Huiyan Zeng, Ramani Ramchandran, Barden Chan, Vivekanand Jha, and Nishla Keiser
- Subjects
Glypican ,Type XVIII collagen ,Gene Expression ,Oligosaccharides ,macromolecular substances ,CHO Cells ,chemistry.chemical_compound ,Mice ,Cricetinae ,Collagen Type XVIII ,Animals ,Endothelium ,Cloning, Molecular ,Receptor ,Molecular Biology ,biology ,Heparin ,Sulfates ,Membrane Proteins ,Cell Biology ,Heparan sulfate ,3T3 Cells ,Molecular biology ,Peptide Fragments ,Endostatins ,Rats ,Endothelial stem cell ,Kidney Tubules ,Proteoglycan ,chemistry ,biology.protein ,cardiovascular system ,Collagen ,Endostatin ,Heparan Sulfate Proteoglycans ,Protein Binding - Abstract
Endostatin, a collagen XVIII fragment, is a potent anti-angiogenic protein. We sought to identify its endothelial cell surface receptor(s). Alkaline phosphatase- tagged endostatin bound endothelial cells revealing two binding affinities. Expression cloning identified glypican, a cell surface proteoglycan as the lower-affinity receptor. Biochemical and genetic studies indicated that glypicans' heparan sulfate glycosaminoglycans were critical for endostatin binding. Furthermore, endostatin selected a specific octasulfated hexasaccharide from a sequence in heparin. We have also demonstrated a role for endostatin in renal tubular cell branching morphogenesis and show that glypicans serve as low-affinity receptors for endostatin in these cells, as in endothelial cells. Finally, antisense experiments suggest the critical importance of glypicans in mediating endostatin activities.
- Published
- 2001
- Full Text
- View/download PDF
28. Alternate splicing in human Na+-MI cotransporter gene yields differentially regulated transport isoforms
- Author
-
Anil Karihaloo, Paul D. Killen, Francesca Porcellati, Masaki Togawa, Douglas A. Greene, Tommy Hlaing, Yoshiyuki Hosaka, Martin J. Stevens, and Dennis Larkin
- Subjects
Gene isoform ,Transcription, Genetic ,Physiology ,Xenopus ,Molecular Sequence Data ,Biology ,Open Reading Frames ,Dogs ,Gene expression ,Animals ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Protein kinase A ,Cells, Cultured ,Heat-Shock Proteins ,Base Sequence ,Symporters ,Reverse Transcriptase Polymerase Chain Reaction ,Alternative splicing ,Membrane Proteins ,Nucleic Acid Hybridization ,Cell Biology ,Exons ,Membrane transport ,Cyclic AMP-Dependent Protein Kinases ,Alternative Splicing ,Biochemistry ,Osmolyte ,Oocytes ,Cotransporter ,Carrier Proteins ,Intracellular - Abstract
myo-Inositol is a ubiquitous intracellular organic osmolyte and phosphoinositide precursor maintained at millimolar intracellular concentrations through the action of membrane-associated Na+- myo-inositol cotransporters (SMIT). Functional cloning and expression of a canine SMIT cDNA, which conferred SMIT activity in Xenopus oocytes, predicted a 718-amino acid peptide homologous to the Na+-glucose cotransporter with a potential protein kinase A phosphorylation site and multiple protein kinase C phosphorylation sites. A consistent ∼1.0- to 13.5-kb array of transcripts hybridizing with this cDNA are osmotically induced in a variety of mammalian cells and species, yet SMIT activity appears to vary among different tissues and species. An open reading frame on human chromosome 21 (SLC5A3) homologous to that of the canine cDNA (96.5%) is thought to comprise an intronless human SMIT gene. Recently, this laboratory ascribed multiply sized, osmotically induced SMIT transcripts in human retinal pigment epithelial cells to the alternate utilization of several 3′-untranslated SMIT exons. This article describes an alternate splice donor site within the coding region that extends the open reading frame into the otherwise untranslated 3′ exons, potentially generating novel SMIT isoforms. In these isoforms, the last putative transmembrane domain is replaced with intracellular carboxy termini containing a novel potential protein kinase A phosphorylation site and multiple protein kinase C phosphorylation sites, and this could explain the heterogeneity in the regulation and structure of the SMIT.
- Published
- 1999
29. Heparin binding VEGF isoforms attenuate hyperoxic embryonic lung growth retardation via a FLK1-neuropilin-1-PKC dependent pathway
- Author
-
Anil Karihaloo, Alia Bazzy-Asaad, Lloyd G. Cantley, Susan E. Quaggin, and Americo E. Esquibies
- Subjects
Vascular Endothelial Growth Factor A ,Pulmonary and Respiratory Medicine ,Cell ,Apoptosis ,Mice, Transgenic ,Hyperoxia ,Biology ,Mice ,Random Allocation ,chemistry.chemical_compound ,Organ Culture Techniques ,Pregnancy ,Neuropilin 1 ,Branching morphogenesis ,medicine ,Animals ,Protein Isoforms ,Lung ,Protein Kinase C ,Protein kinase C ,Heparin ,Research ,Kinase insert domain receptor ,Lung explant ,respiratory system ,VEGF ,Vascular Endothelial Growth Factor Receptor-2 ,Molecular biology ,Cell Hypoxia ,Neuropilin-1 ,respiratory tract diseases ,Cell biology ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,chemistry ,Female ,Signal transduction ,medicine.symptom ,Protein Binding ,Signal Transduction - Abstract
Background Previous work in our laboratory demonstrated that hyperoxia suppressed the expression of vascular endothelial growth factor (VEGF) by the embryonic lung, leading to increased epithelial cell apoptosis and failure of explant airway growth and branching that was rescued by the addition of Vegf165. The aims of this study were to determine protective pathways by which VEGF isoforms attenuate hyperoxic lung growth retardation and to identify the target cell for VEGF action. Methods Timed pregnant CD-1 or fetal liver kinase (FLK1)-eGFP lung explants cultured in 3% or 50% oxygen were treated ± Vegf121, VEGF164/Vegf165 or VEGF188 in the presence or absence of anti-rat neuropilin-1 (NRP1) antibody or GO6983 (protein kinase C (PKC) pan-inhibitor) and lung growth and branching quantified. Immunofluorescence studies were performed to determine apoptosis index and location of FLK1 phosphorylation and western blot studies of lung explants were performed to define the signaling pathways that mediate the protective effects of VEGF. Results Heparin-binding VEGF isoforms (VEGF164/Vegf165 and VEGF188) but not Vegf121 selectively reduced epithelial apoptosis and partially rescued lung bud branching and growth. These protective effects required NRP1-dependent FLK1 activation in endothelial cells. Analysis of downstream signaling pathways demonstrated that the VEGF-mediated anti-apoptotic effects were dependent on PKC activation. Conclusions Vegf165 activates FLK1-NRP1 signaling in endothelial cells, leading to a PKC-dependent paracrine signal that in turn inhibits epithelial cell apoptosis.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.