201
|
Molecular make-up of the glomerular filtration barrier. Biochem Biophys Res Commun 2010; 396:164-9. [PMID: 20494132 DOI: 10.1016/j.bbrc.2010.04.069] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 04/09/2010] [Indexed: 12/23/2022]
Abstract
The glomerular filtration barrier is composed of glomerular endothelial cells, the glomerulus basement membrane and the podocyte cell layer. The filtration barrier is a target of injury in several systemic and renal diseases, and this often leads to progressive renal disease and kidney failure. Therefore, it is essential to understand the molecular biology of the glomerulus. During the last two decades, a lot of new information about molecular components of the glomerulus filtration barrier has been generated. Many of the key discoveries have been obtained through studies on the genetic background of inherited glomerular diseases. These studies have emphasized the role of podocytes in the filtration barrier function. During the last decade, the use of knockout mouse technology has become more available and given important new insights into the functional significance of glomerular components. Large-scale approaches, such as microarray profiling, have also given data about molecules involved in the biology and pathology of the glomerulus. In the coming decade, the use of global expression profiling platforms, transgenic mouse lines, and other in vivo gene delivery methods will rapidly expand our understanding of biology and pathology of the glomerular filtration barrier, and hopefully expose novel target molecules for therapy in progressive renal diseases.
Collapse
|
202
|
Dix L, Roth-Kleiner M, Osterheld MC. Placental vascular obstructive lesions: risk factor for developing necrotizing enterocolitis. PATHOLOGY RESEARCH INTERNATIONAL 2010; 2010:838917. [PMID: 21151528 PMCID: PMC2989861 DOI: 10.4061/2010/838917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 02/16/2010] [Accepted: 02/18/2010] [Indexed: 11/20/2022]
Abstract
Necrotizing enterocolitis (NEC) is a severe neonatal disease affecting particularly preterm infants. Its exact pathogenesis still remains unknown. In this study, we have compared the prevalence of vascular obstructive lesions in placentae of premature newborns which developed NEC and of a control group. We further compared separately the findings of placentae of infants of less than 30 weeks of gestation, the age group in which NEC occurs most frequently. We found signs of fetal vascular obstructive lesions in 65% of the placentae of preterm patients developing NEC, compared to only 17% of the placentae of preterm patients in the control group. In the age groups below 30 weeks of gestation, 58.5% of placentae of later NEC patients presented such lesions compared to 24.5% in the control group. The significant difference between NEC and control group suggests a strong association between fetal vascular obstructive lesions and NEC. Therefore, we propose that fetal vascular obstructive lesions might be considered as a risk factor for the development of NEC in premature infants.
Collapse
Affiliation(s)
- Laure Dix
- Institute of Pathology, University of Lausanne, CH-1011 Lausanne, Switzerland
| | | | | |
Collapse
|
203
|
Wen T, Zhang Z, Yu Y, Qu H, Koch M, Aumailley M. Integrin alpha3 subunit regulates events linked to epithelial repair, including keratinocyte migration and protein expression. Wound Repair Regen 2010; 18:325-34. [PMID: 20412552 DOI: 10.1111/j.1524-475x.2010.00590.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two integrins, alpha3beta1 and alpha6beta4, are high-affinity receptors for laminin 332, the major laminin isoform of the dermal-epidermal junction, although they are thought to have different functions. Biological and genetic studies have firmly established that the alpha6beta4 integrin is indispensable for the stable anchorage of the epidermis to the underlying dermis. In contrast, the alpha3beta1 integrin is thought to be important for cell migration, although the issue is controversial, and both positive and negative effects have been reported. To address the function of alpha3beta1 integrin, we used small interfering RNA to down-regulate the alpha3 subunit in human keratinocytes. The resulting phenotype indicates that lack of alpha3beta1 integrin compromises intercellular adhesion and collective migration, while it enhances single cell migration with a concomitant increase of both focal adhesion kinase and extracellular signal-regulated kinase. In addition, down-regulation of integrin alpha3 subunit results in an increased expression of fibronectin and precursor laminin 332, two extracellular matrix proteins known to be up-regulated during wound healing. Thus, down-regulation of alpha3beta1 integrin recapitulates crucial events governing keratinocyte migration associated with wound healing and tissue repair.
Collapse
|
204
|
Immunopathologies linked to integrin signalling. Semin Immunopathol 2010; 32:173-82. [DOI: 10.1007/s00281-010-0202-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 02/08/2010] [Indexed: 02/07/2023]
|
205
|
Zou MS, Yu J, Zhou JH, Nie GM, Ding DS, Luo LM, Xu HT, He WS. 1,25-Dihydroxyvitamin D3 ameliorates podocytopenia in rats with adriamycin-induced nephropathy. Intern Med 2010; 49:2677-86. [PMID: 21173542 DOI: 10.2169/internalmedicine.49.4174] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To investigate the role of α3β1 integrin and α/β-dystroglycan in protective effects of 1,25(OH)2D3 on podocytes in rats with adriamycin-induced nephropathy. METHODS Sprague-Dawley rats were randomly divided into three groups: control group (NC), nephropathy group (NE), and nephropathy+1,25(OH)2D3 group (ND). Rats in NE and ND group were injected intravenously with adriamycin (0.1 mg/10 g body weight) to induce nephropathy, and those in ND group were then subcutaneously treated with 1,25(OH)2D3 for 8 weeks. Urinary protein level, number of urine podocytes, foot process width and glomerulosclerotic index were determined. Nephrin and podocin mRNA and protein expressions were determined by RT-PCR and western blot, respectively. Podocyte density and expressions of α3β1 integrin and α/β-dystroglycan (DG) were analyzed by immunohistochemistry and western blot, respectively. RESULTS The increase in proteinuria, podocyturia and width of foot process in NE group were ameliorated after treatment with 1,25(OH)2D3 for 8 weeks. The glomerulosclerotic index was significantly decreased in ND group when compared with NE group. The podocyte density in ND group (10.3±1.64 cells/glomerulus) was significantly higher than that in NE group (8.43±1.75 cells/glomerulus) (p=0.008). 1,25(OH)2D3 treatment could significantly up-regulate the mRNA and protein expressions of nephrin and podocin, and the protein expressions of α3β1 integrin and α/β-DG. CONCLUSION The expressions of nephrin, podocin, α3β1 integrin and α/β-DG were decreased in rats with nephropathy. However, 1,25(OH)2D3 treatment could significantly up-regulate the expressions of nephrin, podocin, α3β1 integrin and α/β-DG proteins which might suppress podocyte detachment and podocytopenia.
Collapse
Affiliation(s)
- Min-shu Zou
- Department of Pediatrics, Wuhan General Hospital of Guangzhou Command, and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
206
|
|
207
|
Caccavari F, Valdembri D, Sandri C, Bussolino F, Serini G. Integrin signaling and lung cancer. Cell Adh Migr 2010; 4:124-9. [PMID: 20139694 PMCID: PMC2852569 DOI: 10.4161/cam.4.1.10976] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 12/16/2009] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of most non small cell lung carcinomas is due to their ability to efficiently invade surrounding tissues and blood vessels, finally metastasizing to distant organs. Integrin mediated adhesive interaction with the surrounding extracellular matrix is a key limiting step in the regulation of the invasive properties of several cancer cell types. Here, we examine the rising evidences about the role that integrins can play in the physiopathology of non small cell lung carcinomas by regulating cell adhesion as well as the activation of growth factors and the traffic of their cognate receptors. Modulation of the signaling pathways controlled by integrins in lung cancer cells might offer the opportunity to design and develop new drugs that might be successfully combined with conventional chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Francesca Caccavari
- Laboratory of Cell Signaling; Division of Vascular Biology; Institute for Cancer Research and Treatment and Department of Oncological Sciences; University of Torino School of Medicine; Candiolo (TO), Italy
| | - Donatella Valdembri
- Laboratory of Cell Signaling; Division of Vascular Biology; Institute for Cancer Research and Treatment and Department of Oncological Sciences; University of Torino School of Medicine; Candiolo (TO), Italy
| | - Chiara Sandri
- Laboratory of Cell Signaling; Division of Vascular Biology; Institute for Cancer Research and Treatment and Department of Oncological Sciences; University of Torino School of Medicine; Candiolo (TO), Italy
| | - Federico Bussolino
- Laboratory of Angiogenesis; Division of Vascular Biology; Institute for Cancer Research and Treatment and Department of Oncological Sciences; University of Torino School of Medicine; Candiolo (TO), Italy
| | - Guido Serini
- Laboratory of Cell Signaling; Division of Vascular Biology; Institute for Cancer Research and Treatment and Department of Oncological Sciences; University of Torino School of Medicine; Candiolo (TO), Italy
| |
Collapse
|
208
|
Warburton D, El-Hashash A, Carraro G, Tiozzo C, Sala F, Rogers O, De Langhe S, Kemp PJ, Riccardi D, Torday J, Bellusci S, Shi W, Lubkin SR, Jesudason E. Lung organogenesis. Curr Top Dev Biol 2010; 90:73-158. [PMID: 20691848 PMCID: PMC3340128 DOI: 10.1016/s0070-2153(10)90003-3] [Citation(s) in RCA: 303] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Developmental lung biology is a field that has the potential for significant human impact: lung disease at the extremes of age continues to cause major morbidity and mortality worldwide. Understanding how the lung develops holds the promise that investigators can use this knowledge to aid lung repair and regeneration. In the decade since the "molecular embryology" of the lung was first comprehensively reviewed, new challenges have emerged-and it is on these that we focus the current review. Firstly, there is a critical need to understand the progenitor cell biology of the lung in order to exploit the potential of stem cells for the treatment of lung disease. Secondly, the current familiar descriptions of lung morphogenesis governed by growth and transcription factors need to be elaborated upon with the reinclusion and reconsideration of other factors, such as mechanics, in lung growth. Thirdly, efforts to parse the finer detail of lung bud signaling may need to be combined with broader consideration of overarching mechanisms that may be therapeutically easier to target: in this arena, we advance the proposal that looking at the lung in general (and branching in particular) in terms of clocks may yield unexpected benefits.
Collapse
Affiliation(s)
- David Warburton
- The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Ivaska J, Heino J. Interplay between cell adhesion and growth factor receptors: from the plasma membrane to the endosomes. Cell Tissue Res 2010; 339:111-20. [PMID: 19722108 PMCID: PMC2784865 DOI: 10.1007/s00441-009-0857-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 08/03/2009] [Indexed: 11/24/2022]
Abstract
The emergence of multicellular animals could only take place once evolution had produced molecular mechanisms for cell adhesion and communication. Today, all metazoans express integrin-type adhesion receptors and receptors for growth factors. Integrins recognize extracellular matrix proteins and respective receptors on other cells and, following ligand binding, can activate the same cellular signaling pathways that are regulated by growth factor receptors. Recent reports have indicated that the two receptor systems also collaborate in many other ways. Here, we review the present information concerning the role of integrins as assisting growth factor receptors and the interplay between the receptors in cell signaling and in the orchestration of receptor recycling.
Collapse
Affiliation(s)
- Johanna Ivaska
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, FI-20520 Finland
- Centre for Biotechnology, Turku University, Turku, FI-20520 Finland
- Department of Biochemistry and Food Chemistry, University of Turku, Turku, FI-20014 Finland
| | - Jyrki Heino
- Department of Biochemistry and Food Chemistry, University of Turku, Turku, FI-20014 Finland
| |
Collapse
|
210
|
Reidy KJ, Villegas G, Teichman J, Veron D, Shen W, Jimenez J, Thomas D, Tufro A. Semaphorin3a regulates endothelial cell number and podocyte differentiation during glomerular development. Development 2009; 136:3979-89. [PMID: 19906865 DOI: 10.1242/dev.037267] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Semaphorin3a (Sema3a), a chemorepellant guidance protein, plays crucial roles in neural, cardiac and peripheral vascular patterning. Sema3a is expressed in the developing nephron, mature podocytes and collecting tubules. Sema3a acts as a negative regulator of ureteric bud branching, but its function in glomerular development has not been examined. Here we tested the hypothesis that Sema3a regulates glomerular vascular development using loss- and gain-of-function mouse models. Sema3a deletion resulted in defects in renal vascular patterning, excess endothelial cells within glomerular capillaries, effaced podocytes with extremely wide foot processes and albuminuria. Podocyte Sema3a overexpression during organogenesis resulted in glomerular hypoplasia, characterized by glomerular endothelial cell apoptosis, delayed and abnormal podocyte foot process development, a complete absence of slit diaphragms and congenital proteinuria. Nephrin, WT1 and VEGFR2 were downregulated in Sema3a-overexpressing kidneys. We conclude that Sema3a is an essential negative regulator of endothelial cell survival in developing glomeruli and plays a crucial role in podocyte differentiation in vivo. Hence, a tight regulation of Sema3a dosage is required for the establishment of a normal glomerular filtration barrier.
Collapse
Affiliation(s)
- Kimberly J Reidy
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Abstract
Proteinuria is a major health-care problem that affects several hundred million people worldwide. Proteinuria is a cardinal sign and a prognostic marker of kidney disease, and also an independent risk factor for cardiovascular morbidity and mortality. Microalbuminuria is the earliest cue of renal complications of diabetes, obesity, and the metabolic syndrome. It can often progress to overt proteinuria that in 10-50% of patients is associated with the development of chronic kidney disease, ultimately requiring dialysis or transplantation. Therefore, reduction or prevention of proteinuria is highly desirable. Here we review recent novel insights into the pathogenesis and treatment of proteinuria, with a special emphasis on the emerging concept that proteinuria can result from enzymatic cleavage of essential regulators of podocyte actin dynamics by cytosolic cathepsin L (CatL), resulting in a motile podocyte phenotype. Finally, we describe signaling pathways controlling the podocyte actin cytoskeleton and motility and how these pathways can be manipulated for therapeutic benefit.
Collapse
Affiliation(s)
- Peter Mundel
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA.
| | | |
Collapse
|
212
|
Meehan DT, Delimont D, Cheung L, Zallocchi M, Sansom SC, Holzclaw JD, Rao V, Cosgrove D. Biomechanical strain causes maladaptive gene regulation, contributing to Alport glomerular disease. Kidney Int 2009; 76:968-76. [PMID: 19710627 PMCID: PMC2780007 DOI: 10.1038/ki.2009.324] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Patients with Alport's syndrome develop a number of pro-inflammatory cytokine and matrix metalloproteinase (MMP) abnormalities that contribute to progressive renal failure. Changes in the composition and structure of the glomerular basement membranes likely alter the biomechanics of cell adhesion and signaling in these patients. To test if enhanced strain on the capillary tuft due to these structural changes contributes to altered gene regulation, we subjected cultured podocytes to cyclic biomechanical strain. There was robust induction of interleukin (IL)-6, along with MMP-3, -9, -10, and -14, but not MMP-2 or -12 by increased strain. Neutralizing antibodies against IL-6 attenuated the strain-mediated induction of MMP-3 and -10. Alport mice treated with a general inhibitor of nitric oxide synthase (L-NAME) developed significant hypertension and increased IL-6 and MMP-3 and -10 in their glomeruli relative to those of normotensive Alport mice. These hypertensive Alport mice also had elevated proteinuria along with more advanced histological and ultrastructural glomerular basement membrane damage. We suggest that MMP and cytokine dysregulation may constitute a maladaptive response to biomechanical strain in the podocytes of Alport patients, thus contributing to glomerular disease initiation and progression.
Collapse
MESH Headings
- Adaptation, Physiological/genetics
- Animals
- Blood Pressure
- Cells, Cultured
- Cytoskeleton/metabolism
- Disease Models, Animal
- Gene Expression Regulation
- Glomerular Basement Membrane/metabolism
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/physiopathology
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Kidney Glomerulus/metabolism
- Kidney Glomerulus/physiopathology
- Matrix Metalloproteinase 10/genetics
- Matrix Metalloproteinase 14/genetics
- Matrix Metalloproteinase 3/genetics
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinases/genetics
- Matrix Metalloproteinases/metabolism
- Mice
- Mice, Inbred C57BL
- NG-Nitroarginine Methyl Ester
- Nephritis, Hereditary/genetics
- Nephritis, Hereditary/metabolism
- Nephritis, Hereditary/physiopathology
- Podocytes/metabolism
- Proteinuria/chemically induced
- Proteinuria/genetics
- Proteinuria/physiopathology
- RNA, Messenger/metabolism
- Sodium Chloride, Dietary
- Stress, Mechanical
- Time Factors
Collapse
Affiliation(s)
| | | | - Linda Cheung
- Boys Town National Research Hospital, Omaha Nebraska
| | | | - Steven C. Sansom
- University of Nebraska Medical Center, Department of Cellular and Integrative Physiology, Omaha, NE
| | - J. David Holzclaw
- University of Nebraska Medical Center, Department of Cellular and Integrative Physiology, Omaha, NE
| | - Velidi Rao
- Boys Town National Research Hospital, Omaha Nebraska
| | | |
Collapse
|
213
|
Benjamin JT, Gaston DC, Halloran BA, Schnapp LM, Zent R, Prince LS. The role of integrin alpha8beta1 in fetal lung morphogenesis and injury. Dev Biol 2009; 335:407-17. [PMID: 19769957 DOI: 10.1016/j.ydbio.2009.09.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 09/11/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
Prenatal inflammation prevents normal lung morphogenesis and leads to bronchopulmonary dysplasia (BPD), a common complication of preterm birth. We previously demonstrated in a bacterial endotoxin mouse model of BPD that disrupting fibronectin localization in the fetal lung mesenchyme causes arrested saccular airway branching. In this study we show that expression of the fibronectin receptor, integrin alpha8beta1 is decreased in the lung mesenchyme in the same inflammation model suggesting it is required for normal lung development. We verified a role for integrin alpha8beta1 in lung development using integrin alpha8-null mice, which develop fusion of the medial and caudal lobes as well as abnormalities in airway division. We further show in vivo and in vitro that alpha8-null fetal lung mesenchymal cells fail to form stable adhesions and have increased migration. Thus we propose that integrin alpha8beta1 plays a critical role in lung morphogenesis by regulating mesenchymal cell adhesion and migration. Furthermore, our data suggest that disruption of the interactions between extracellular matrix and integrin alpha8beta1 may contribute to the pathogenesis of BPD.
Collapse
Affiliation(s)
- John T Benjamin
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232-0493, USA
| | | | | | | | | | | |
Collapse
|
214
|
Trivedi S, Zeier M, Reiser J. Role of podocytes in lupus nephritis. Nephrol Dial Transplant 2009; 24:3607-12. [DOI: 10.1093/ndt/gfp427] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
215
|
Patrakka J, Tryggvason K. New insights into the role of podocytes in proteinuria. Nat Rev Nephrol 2009; 5:463-8. [DOI: 10.1038/nrneph.2009.108] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
216
|
Fukasawa H, Bornheimer S, Kudlicka K, Farquhar MG. Slit diaphragms contain tight junction proteins. J Am Soc Nephrol 2009; 20:1491-503. [PMID: 19478094 DOI: 10.1681/asn.2008101117] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Slit diaphragms are essential components of the glomerular filtration apparatus, as changes in these junctions are the hallmark of proteinuric diseases. Slit diaphragms, considered specialized adherens junctions, contain both unique membrane proteins (e.g., nephrin, podocin, and Neph1) and typical adherens junction proteins (e.g., P-cadherin, FAT, and catenins). Whether slit diaphragms also contain tight junction proteins is unknown. Here, immunofluorescence, immunogold labeling, and cell fractionation demonstrated that rat slit diaphragms contain the tight junction proteins JAM-A (junctional adhesion molecule A), occludin, and cingulin. We found these proteins in the same protein complexes as nephrin, podocin, CD2AP, ZO-1, and Neph1 by cosedimentation, coimmunoprecipitation, and pull-down assays. PAN nephrosis increased the protein levels of JAM-A, occludin, cingulin, and ZO-1 several-fold in glomeruli and loosened their attachment to the actin cytoskeleton. These data extend current information about the molecular composition of slit diaphragms by demonstrating the presence of tight junction proteins, although slit diaphragms lack the characteristic morphologic features of tight junctions. The contribution of these proteins to the assembly of slit diaphragms and potential signaling cascades requires further investigation.
Collapse
Affiliation(s)
- Hirotaka Fukasawa
- Department of Cellular and Molecular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0651, USA
| | | | | | | |
Collapse
|
217
|
Takabatake Y, Sugiyama T, Kohara H, Matsusaka T, Kurihara H, Koni PA, Nagasawa Y, Hamano T, Matsui I, Kawada N, Imai E, Nagasawa T, Rakugi H, Isaka Y. The CXCL12 (SDF-1)/CXCR4 axis is essential for the development of renal vasculature. J Am Soc Nephrol 2009; 20:1714-23. [PMID: 19443644 DOI: 10.1681/asn.2008060640] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
CXC chemokine ligand 12 (CXCL12; stromal cell-derived factor 1) is a unique homeostatic chemokine that signals through its cognate receptor, CXCR4. CXCL12/CXCR4 signaling is essential for the formation of blood vessels in the gastrointestinal tract during development, but its contribution to renal development remains unclear. Here, we found that CXCL12-secreting stromal cells surround CXCR4-positive epithelial components of early nephrons and blood vessels in the embryonic kidney. In glomeruli, we observed CXCL12-secreting podocytes in close proximity to CXCR4-positive endothelial cells. Both CXCL12- and CXCR4-deficient kidneys exhibited identical phenotypes; there were no apparent abnormalities in early nephrogenesis or in differentiation of podocytes and tubules, but there was defective formation of blood vessels, including ballooning of the developing glomerular tuft and disorganized patterning of the renal vasculature. To clarify the relative importance of different cellular defects resulting from ablation of CXCL12 and CXCR4, we established endothelial cell-specific CXCR4-deficient mice, which recapitulated the renal phenotypes of conventional CXCR4-deficient mice. We conclude that CXCL12 secreted from stromal cells or podocytes acts on endothelial cells to regulate vascular development in the kidney. These findings suggest new potential therapeutic targets for remodeling the injured kidney.
Collapse
Affiliation(s)
- Yoshitsugu Takabatake
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine (B6), Suita, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Wu W, Kitamura S, Truong DM, Rieg T, Vallon V, Sakurai H, Bush KT, Vera DR, Ross RS, Nigam SK. Beta1-integrin is required for kidney collecting duct morphogenesis and maintenance of renal function. Am J Physiol Renal Physiol 2009; 297:F210-7. [PMID: 19439520 DOI: 10.1152/ajprenal.90260.2008] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Deletion of integrin-beta1 (Itgb1) in the kidney collecting system led to progressive renal dysfunction and polyuria. The defect in the concentrating ability of the kidney was concomitant with decreased medullary collecting duct expression of aquaporin-2 and arginine vasopressin receptor 2, while histological examination revealed hypoplastic renal medullary collecting ducts characterized by increased apoptosis, ectasia and cyst formation. In addition, a range of defects from small kidneys with cysts and dilated tubules to bilateral renal agenesis was observed. This was likely due to altered growth and branching morphogenesis of the ureteric bud (the progenitor tissue of the renal collecting system), despite the apparent ability of the ureteric bud-derived cells to induce differentiation of the metanephric mesenchyme. These data not only support a role for Itgb1 in the development of the renal collecting system but also raise the possibility that Itgb1 links morphogenesis to terminal differentiation and ultimately collecting duct function and/or maintenance.
Collapse
Affiliation(s)
- Wei Wu
- Department of Pediatrics, University of California, La Jolla, California 92093-0693, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Dessapt C, Baradez MO, Hayward A, Dei Cas A, Thomas SM, Viberti G, Gnudi L. Mechanical forces and TGFbeta1 reduce podocyte adhesion through alpha3beta1 integrin downregulation. Nephrol Dial Transplant 2009; 24:2645-55. [PMID: 19420102 DOI: 10.1093/ndt/gfp204] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Podocyturia is a marker of diabetic nephr- opathy, a possible determinant of its progression and a powerful risk factor for cardiovascular disease. A reduction in podocyte adhesion to the glomerular basement membrane (GBM) via downregulation of alpha3beta1 integrin expression, the main podocyte anchoring dimer to the GBM, may represent one of the mechanisms of podocyturia in glomerular disease. This study investigated the role of mechanical forces and transforming growth factor beta1 (TGFbeta1) in podocyte adhesion and integrin expression. METHODS Conditionally immortalized murine podocytes were exposed to mechanical stretch and/or TGFbeta1 for 48 h. Podocyte adhesion, apoptosis and alpha3beta1 integrin expression were assessed. RESULTS Stretch and TGFbeta1 significantly reduced podocyte adhesion and alpha3beta1 integrin expression, events paralleled by increased apoptosis. Blockade of beta1 integrin, with a specific antibody, demonstrated a reduced podocyte adhesion indicating that beta1 integrin downregulation was required for the loss of podocyte adhesion. This was linked to an increase in podocyte apoptosis. The role of apoptosis in podocyte adhesion was further investigated using caspase-3 inhibitors. Podocyte apoptosis inhibition did not affect stretch- and TGFbeta1-mediated integrin downregulation and the loss of podocyte adhesion, suggesting that alpha3beta1 integrin downregulation is sufficient to alter cell adhesion. Although stretch significantly increased podocyte TGFbeta type I, II and III receptors but not podocyte TGFbeta1 secretion, the combination of stretch and TGFbeta1 did not show any additive or synergistic effects on podocyte adhesion and alpha3beta1 integrin expression. CONCLUSIONS These results suggest that downregulation of alpha3beta1 integrin expression, by mechanical forces or TGFbeta1, is per se sufficient to reduce podocyte adhesion. Apoptosis may represent a parallel important determinant of the podocyte loss from the GBM.
Collapse
|
220
|
Volpe MV, Chung E, Ulm JP, Gilchrist BF, Ralston S, Wang KT, Nielsen HC. Aberrant cell adhesion molecule expression in human bronchopulmonary sequestration and congenital cystic adenomatoid malformation. Am J Physiol Lung Cell Mol Physiol 2009; 297:L143-52. [PMID: 19411307 DOI: 10.1152/ajplung.90618.2008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In many organs, integrins and cadherins are partly regulated by Hox genes, but their interactions in airway morphogenesis and congenital lung diseases are unknown. We previously showed that the Hox protein HoxB5 is abnormally increased in bronchopulmonary sequestration (BPS) and congenital cystic adenomatoid malformation (CCAM), congenital lung lesions with abnormal airway branching. We now report on alpha(2)-, alpha(3)-, and beta(1)-integrin and E-cadherin expression in normal human lung and in BPS and CCAM tissue previously shown to have abnormal HoxB5 expression and on the relationship of cell adhesion molecule expression to Hoxb5 regulation. alpha(2)-, alpha(3)-, and beta(1)-integrins and E-cadherin expression in normal human lung and BPS and CCAM were evaluated using Western blot and immunohistochemistry. Fetal mouse lung fibroblasts with Hoxb5-specific siRNA downregulation were evaluated for alpha(2)-integrin protein levels by Western blot. Compared with normal human lung, a previously undetected alpha(2)-integrin isoform potentially lacking essential cytoplasmic sequences was significantly increased in BPS and CCAM, and alpha(2)-integrin spatial and cellular expression was more intense. E-cadherin protein levels were also significantly increased, whereas alpha(3) increased in CCAM compared with canalicular, but not with alveolar, stage lung. beta(1)-integrin levels were unchanged. We conclude that in BPS and CCAM, altered alpha(2)-integrin cytoplasmic signaling contributes to abnormal cellular behavior in these lung lesions. Aberrant cell adhesion molecule and Hox protein regulation are likely part of the mechanism involved in the development of BPS and CCAM.
Collapse
Affiliation(s)
- Maryann V Volpe
- Department of Pediatrics, Division of Newborn Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts 02111, USA.
| | | | | | | | | | | | | |
Collapse
|
221
|
Bruggeman SWM, Hulsman D, van Lohuizen M. Bmi1 deficient neural stem cells have increased integrin dependent adhesion to self-secreted matrix. Biochim Biophys Acta Gen Subj 2009; 1790:351-60. [PMID: 19298843 DOI: 10.1016/j.bbagen.2009.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 02/25/2009] [Accepted: 03/02/2009] [Indexed: 10/21/2022]
Abstract
BACKGROUND Neural cells deficient for Polycomb group (PcG) protein Bmi1 are impaired in the formation and differentiation of high grade glioma, an incurable cancer of the brain. It was shown that mechanisms involved in cell adhesion and migration were specifically affected in these tumors. METHODS Using biochemical and cell biological approaches, we investigated the adhesive capacities of Bmi1;Ink4a/Arf deficient primary neural stem cells (NSCs). RESULTS Bmi1;Ink4a/Arf deficient NSCs have altered expression of Collagen-related genes, secrete increased amounts of extracellular matrix, and exhibit enhanced cell-matrix binding through the Beta-1 Integrin receptor. These traits are independent from the well described role of Bmi1 as repressor of the Ink4a/Arf tumor suppressor locus. CONCLUSION In addition to proliferative processes, Bmi1 controls the adhesive capacities of primary NSCs by modulating extracellular matrix secretion. GENERAL SIGNIFICANCE Since PcG protein Bmi1 is important for both normal development and tumorigenesis, it is vital to understand the complete network in which this protein acts. Whereas it is clear that control of Ink4a/Arf is a major Bmi1 function, there is evidence that other downstream mechanisms exist. Hence, our novel finding that Bmi1 also governs cell adhesion significantly contributes to our understanding of the PcG proteins.
Collapse
Affiliation(s)
- Sophia W M Bruggeman
- The Netherlands Cancer Institute, Division of Molecular Genetics, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | | | | |
Collapse
|
222
|
Turgeon B, Meloche S. Interpreting neonatal lethal phenotypes in mouse mutants: insights into gene function and human diseases. Physiol Rev 2009; 89:1-26. [PMID: 19126753 DOI: 10.1152/physrev.00040.2007] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The mouse represents the model of choice to study the biological function of mammalian genes through mutation of its genome. However, the biggest challenge of mouse geneticists remains the phenotypic analysis of mouse mutants. A survey of mouse mutant databases reveals a surprisingly high number of gene mutations leading to neonatal death. These genetically modified mouse mutants have been instrumental in elucidating gene function and have become important models of congenital human diseases. The main complication when phenotyping mutant mice dying during the neonatal period is the large spectrum of physiological systems whose defects can challenge neonatal survival. Here, we present a comprehensive review of gene mutations leading to neonatal lethality and discuss the impact of these mutations on the major physiological processes critical to mouse newborn survival: parturition, breathing, suckling, and homeostasis. Selected examples of mouse mutants are highlighted to illustrate how the precise identification of the timing and cause of death associated with these physiological processes allows for a more profound understanding of the underlying cellular and molecular defects. This review provides a guide for the analysis of neonatal lethal phenotypes in mutant mice that will be helpful for dissecting out the function of specific genes during mouse development.
Collapse
Affiliation(s)
- Benjamin Turgeon
- Department of Pharmacology and Molecular Biology, Université de Montréal, Montreal, Quebec, Canada
| | | |
Collapse
|
223
|
Hemler ME. Targeting of tetraspanin proteins--potential benefits and strategies. Nat Rev Drug Discov 2009; 7:747-58. [PMID: 18758472 DOI: 10.1038/nrd2659] [Citation(s) in RCA: 274] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tetraspanin transmembrane proteins have emerged as key players in malignancy, the immune system, during fertilization and infectious disease processes. Tetraspanins engage in a wide range of specific molecular interactions, occurring through the formation of tetraspanin-enriched microdomains (TEMs). TEMs therefore serve as a starting point for understanding how tetraspanins affect cell signalling, adhesion, morphology, motility, fusion and virus infection. An abundance of recent evidence suggests that targeting tetraspanins, for example, by monoclonal antibodies, soluble large-loop proteins or RNAi technology, should be therapeutically beneficial.
Collapse
Affiliation(s)
- Martin E Hemler
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, Massachussetts 02115, USA.
| |
Collapse
|
224
|
Barisoni L, Schnaper HW, Kopp JB. Advances in the biology and genetics of the podocytopathies: implications for diagnosis and therapy. Arch Pathol Lab Med 2009; 133:201-16. [PMID: 19195964 PMCID: PMC3118840 DOI: 10.5858/133.2.201] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT Etiologic factors and pathways leading to altered podocyte phenotype are clearly numerous and involve the activity of different cellular function. OBJECTIVE To focus on recent discoveries in podocyte biology and genetics and their relevance to these human glomerular diseases, named podocytopathies. DATA SOURCES Genetic mutations in genes encoding for proteins in the nucleus, slit diaphragm, podocyte cytoplasm, and cell membrane are responsible for podocyte phenotype and functional abnormalities. Podocyte injury may also derive from secondary stimuli, such as mechanical stress, infections, or use of certain medications. Podocytes can respond to injury in a limited number of ways, which include (1) effacement, (2) apoptosis, (3) arrest of development, and (4) dedifferentiation. Each of these pathways results in a specific glomerular morphology: minimal change nephropathy, focal segmental glomerulosclerosis, diffuse mesangial sclerosis, and collapsing glomerulopathy. CONCLUSIONS Based on current knowledge of podocyte biology, we organized etiologic factors and morphologic features in a taxonomy of podocytopathies, which provides a novel approach to the classification of these diseases. Current and experimental therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Laura Barisoni
- Department of Pathology and Medicine, Division of Nephrology, New York University School of Medicine, New York, NY 10017, USA.
| | | | | |
Collapse
|
225
|
Walker J, Menko AS. Integrins in lens development and disease. Exp Eye Res 2009; 88:216-25. [PMID: 18671967 PMCID: PMC2698936 DOI: 10.1016/j.exer.2008.06.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 06/19/2008] [Accepted: 06/23/2008] [Indexed: 11/26/2022]
Abstract
Integrins are the major cell surface receptors for proteins in the extracellular matrix. These receptors form major cell signaling centers that are bidirectional, communicating messages between the cell and its environment. They are a large receptor family, with members well-known to regulate cellular processes essential to both development and disease. In this review we examine the literature regarding integrins in the lens. Here we cover integrin function in lens cell differentiation, in the development of the lens and in protection of the lens epithelial cell phenotype. In addition, we analyze the role of integrins in the progression of lens fibrotic diseases, focusing particularly on integrin regulation of TGFbeta signaling pathways in posterior capsule opacification (PCO) and anterior subcapsular cataract (ASC).
Collapse
Affiliation(s)
- Janice Walker
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
226
|
Kim Y, Kugler MC, Wei Y, Kim KK, Li X, Brumwell AN, Chapman HA. Integrin alpha3beta1-dependent beta-catenin phosphorylation links epithelial Smad signaling to cell contacts. J Cell Biol 2009; 184:309-22. [PMID: 19171760 PMCID: PMC2654298 DOI: 10.1083/jcb.200806067] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 12/19/2008] [Indexed: 12/26/2022] Open
Abstract
Injury-initiated epithelial to mesenchymal transition (EMT) depends on contextual signals from the extracellular matrix, suggesting a role for integrin signaling. Primary epithelial cells deficient in their prominent laminin receptor, alpha3beta1, were found to have a markedly blunted EMT response to TGF-beta1. A mechanism for this defect was explored in alpha3-null cells reconstituted with wild-type (wt) alpha3 or point mutants unable to engage laminin 5 (G163A) or epithelial cadherin (E-cadherin; H245A). After TGF-beta1 stimulation, wt epithelial cells but not cells expressing the H245A mutant internalize complexes of E-cadherin and TGF-beta1 receptors, generate phospho-Smad2 (p-Smad2)-pY654-beta-catenin complexes, and up-regulate mesenchymal target genes. Although Smad2 phosphorylation is normal, p-Smad2-pY654-beta-catenin complexes do not form in the absence of alpha3 or when alpha3beta1 is mainly engaged on laminin 5 or E-cadherin in adherens junctions, leading to attenuated EMT. These findings demonstrate that alpha3beta1 coordinates cross talk between beta-catenin and Smad signaling pathways as a function of extracellular contact cues and thereby regulates responses to TGF-beta1 activation.
Collapse
Affiliation(s)
- Young Kim
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | |
Collapse
|
227
|
Martín‐Belmonte F, Rodríguez‐Fraticelli AE. Chapter 3 Acquisition of Membrane Polarity in Epithelial Tube Formation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 274:129-82. [DOI: 10.1016/s1937-6448(08)02003-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
228
|
Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML, Frank JA, Brumwell AN, Wheeler SE, Kreidberg JA, Chapman HA. Epithelial cell alpha3beta1 integrin links beta-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J Clin Invest 2009; 119:213-24. [PMID: 19104148 PMCID: PMC2613463 DOI: 10.1172/jci36940] [Citation(s) in RCA: 234] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 10/22/2008] [Indexed: 12/25/2022] Open
Abstract
Pulmonary fibrosis, in particular idiopathic pulmonary fibrosis (IPF), results from aberrant wound healing and scarification. One population of fibroblasts involved in the fibrotic process is thought to originate from lung epithelial cells via epithelial-mesenchymal transition (EMT). Indeed, alveolar epithelial cells (AECs) undergo EMT in vivo during experimental fibrosis and ex vivo in response to TGF-beta1. As the ECM critically regulates AEC responses to TGF-beta1, we explored the role of the prominent epithelial integrin alpha3beta1 in experimental fibrosis by generating mice with lung epithelial cell-specific loss of alpha3 integrin expression. These mice had a normal acute response to bleomycin injury, but they exhibited markedly decreased accumulation of lung myofibroblasts and type I collagen and did not progress to fibrosis. Signaling through beta-catenin has been implicated in EMT; we found that in primary AECs, alpha3 integrin was required for beta-catenin phosphorylation at tyrosine residue 654 (Y654), formation of the pY654-beta-catenin/pSmad2 complex, and initiation of EMT, both in vitro and in vivo during the fibrotic phase following bleomycin injury. Finally, analysis of lung tissue from IPF patients revealed the presence of pY654-beta-catenin/pSmad2 complexes and showed accumulation of pY654-beta-catenin in myofibroblasts. These findings demonstrate epithelial integrin-dependent profibrotic crosstalk between beta-catenin and Smad signaling and support the hypothesis that EMT is an important contributor to pathologic fibrosis.
Collapse
Affiliation(s)
- Kevin K. Kim
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Ying Wei
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Charles Szekeres
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Matthias C. Kugler
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Paul J. Wolters
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Marla L. Hill
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - James A. Frank
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Alexis N. Brumwell
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Sarah E. Wheeler
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Jordan A. Kreidberg
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| | - Harold A. Chapman
- Pulmonary and Critical Care Division, Department of Medicine, and Cardiovascular Research Institute, UCSF, San Francisco, California, USA.
Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Pulmonary and Critical Care Division, Department of Medicine, and San Francisco VA Medical Center, San Francisco, California, USA
| |
Collapse
|
229
|
Abrahamson DR. Development of kidney glomerular endothelial cells and their role in basement membrane assembly. Organogenesis 2009; 5:275-87. [PMID: 19568349 PMCID: PMC2659369 DOI: 10.4161/org.7577] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 12/03/2008] [Indexed: 01/07/2023] Open
Abstract
Data showing that the embryonic day 12 (E12) mouse kidney contains its own pool of endothelial progenitor cells is presented. Mechanisms that regulate metanephric endothelial recruitment and differentiation, including the hypoxia-inducible transcription factors and vascular endothelial growth factor/vascular endothelial growth factor receptor signaling system, are also discussed. Finally, evidence that glomerular endothelial cells contribute importantly to assembly of the glomerular basement membrane (GBM), especially the laminin component, is reviewed. Together, this forum offers insights on blood vessel development in general, and formation of the glomerular capillary in particular, which inarguably is among the most unique vascular structures in the body.
Collapse
Affiliation(s)
- Dale R Abrahamson
- Department of Anatomy and Cell Biology; University of Kansas Medical Center; Kansas City, Kansas USA
| |
Collapse
|
230
|
Shi S, Yu L, Chiu C, Sun Y, Chen J, Khitrov G, Merkenschlager M, Holzman LB, Zhang W, Mundel P, Bottinger EP. Podocyte-selective deletion of dicer induces proteinuria and glomerulosclerosis. J Am Soc Nephrol 2008; 19:2159-69. [PMID: 18776119 PMCID: PMC2573016 DOI: 10.1681/asn.2008030312] [Citation(s) in RCA: 288] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/06/2008] [Indexed: 11/03/2022] Open
Abstract
Dicer is an enzyme that generates microRNA (miRNA), which are small, noncoding RNA that function as important regulators of gene and protein expression. For exploration of the functional roles of miRNA in glomerular biology, Dicer was inactivated selectively in mouse podocytes. Mutant mice developed proteinuria 4 to 5 weeks after birth and died several weeks later, presumably from kidney failure. Multiple abnormalities were observed in glomeruli of mutant mice, including foot process effacement, irregular and split areas of the glomerular basement membrane, podocyte apoptosis and depletion, mesangial expansion, capillary dilation, and glomerulosclerosis. Gene profiling revealed upregulation of 190 genes in glomeruli isolated from mutant mice at the onset of proteinuria compared with control littermates. Target sequences for 16 miRNA were significantly enriched in the 3'-untranslated regions of the 190 upregulated genes. Further suggesting validity of the in silico analysis, six of the eight top-candidate miRNA were identified in miRNA libraries generated from podocyte cultures; these included four members of the mir-30 miRNA family, which are known to degrade target transcripts directly. Among 15 upregulated target genes of the mir-30 miRNA, four genes known to be expressed and/or functional in podocytes were identified, including receptor for advanced glycation end product, vimentin, heat-shock protein 20, and immediate early response 3. Receptor for advanced glycation end product and immediate early response 3 are known to mediate podocyte apoptosis, whereas vimentin and heat-shock protein-20 are involved in cytoskeletal structure. Taken together, these results provide a knowledge base for ongoing investigations to validate functional roles for the mir-30 miRNA family in podocyte homeostasis and podocytopathies.
Collapse
Affiliation(s)
- Shaolin Shi
- Division of Nephrology, Department of Medicine, Mount Sinai School of Medicine, One Gustave L Levy Place, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Tesař V, Zima T. Recent Progress in the Pathogenesis of Nephrotic Proteinuria. Crit Rev Clin Lab Sci 2008; 45:139-220. [DOI: 10.1080/10408360801934865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
232
|
Abstract
Angiogenesis, the formation of new blood vessels from preexisting vasculature, contributes to the pathogenesis of many disorders, including ischemic diseases and cancer. Integrins are cell adhesion molecules that are expressed on the surface of endothelial cells and pericytes, making them potential targets for antiangiogenic therapy. Here we review the contribution of endothelial and mural cell integrins to angiogenesis and highlight their potential as antiangiogenesis targets.
Collapse
Affiliation(s)
- Rita Silva
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Gabriela D'Amico
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Kairbaan M. Hodivala-Dilke
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Louise E. Reynolds
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| |
Collapse
|
233
|
Analysis of genes encoding laminin beta2 and related proteins in patients with Galloway-Mowat syndrome. Pediatr Nephrol 2008; 23:1779-86. [PMID: 18594871 DOI: 10.1007/s00467-008-0880-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 01/15/2023]
Abstract
Galloway-Mowat syndrome (GMS) is a rare autosomal recessive disorder characterized by early onset nephrotic syndrome and microcephaly with various anomalies of the central nervous system. GMS likely represents a heterogeneous group of disorders with hitherto unknown genetic etiology. The clinical phenotype to some extent overlaps that of Pierson syndrome (PS), which comprises congenital nephrotic syndrome and distinct ocular abnormalities but which may also include neurodevelopmental deficits and microcephaly. PS is caused by mutations of LAMB2, the gene encoding laminin beta2. We hypothesized that GMS might be allelic to PS or be caused by defects in proteins that interact with laminin beta2. In a cohort of 18 patients with GMS or a GMS-like phenotype we therefore analyzed the genes encoding laminin beta2 (LAMB2), laminin alpha5 (LAMA5), alpha3-integrin (ITGA3), beta1-integrin (ITGB1) and alpha-actinin-4 (ACTN4), but we failed to find causative mutations in these genes. We inferred that LAMA5, ITGA3, ITGB1, and ACTN4 are not directly involved in the pathogenesis of GMS. We excluded LAMB2 as a candidate gene for GMS. Further studies are required, including linkage analysis in families with GMS to identify genes underlying this disease.
Collapse
|
234
|
Lamar JM, Pumiglia KM, DiPersio CM. An immortalization-dependent switch in integrin function up-regulates MMP-9 to enhance tumor cell invasion. Cancer Res 2008; 68:7371-9. [PMID: 18794124 PMCID: PMC2782775 DOI: 10.1158/0008-5472.can-08-1080] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Integrins, the major receptors for cell adhesion to the extracellular matrix, play important roles during tumor progression. However, it is still unclear whether genetic lesions that occur during carcinoma development can lead to altered integrin function, and how changes in integrin function contribute to subsequent carcinoma progression. Loss-of-function mutations in p53 and activating mutations in H-Ras, which immortalize and transform epithelial cells, respectively, are common causal events in squamous cell carcinoma (SCC). Phenotypes resulting from these two genetic lesions promote SCC progression and are, therefore, potential targets for anticancer therapies. We developed a model system of keratinocyte transformation that has allowed us to investigate the individual roles of p53 mutation and oncogenic Ras mutation in the acquisition of integrin alpha3beta1-regulated phenotypes that promote SCC progression. Using this model, we show that keratinocyte immortalization by p53-null mutation causes a switch in alpha3beta1 function that induces matrix metalloproteinase (MMP)-9 gene expression in tumorigenic cells. This acquired alpha3beta1-dependent regulation of MMP-9 was maintained during subsequent transformation by oncogenic Ras, and it promoted invasion of tumorigenic keratinocytes. Our results show that loss of p53 function leads to changes in integrin-mediated gene regulation that occur during SCC progression and play a critical role in tumor cell invasion.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Line, Transformed
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- Gene Expression
- Humans
- Integrin alpha3beta1/metabolism
- Keratinocytes/enzymology
- Keratinocytes/metabolism
- Keratinocytes/pathology
- Matrix Metalloproteinase 9/biosynthesis
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- John M Lamar
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208-3479, USA
| | | | | |
Collapse
|
235
|
Pitera JE, Scambler PJ, Woolf AS. Fras1, a basement membrane-associated protein mutated in Fraser syndrome, mediates both the initiation of the mammalian kidney and the integrity of renal glomeruli. Hum Mol Genet 2008; 17:3953-64. [PMID: 18787044 PMCID: PMC2638576 DOI: 10.1093/hmg/ddn297] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
FRAS1 is mutated in some individuals with Fraser syndrome (FS) and the encoded protein is expressed in embryonic epidermal cells, localizing in their basement membrane (BM). Syndactyly and cryptophthalmos in FS are sequelae of skin fragility but the bases for associated kidney malformations are unclear. We demonstrate that Fras1 is expressed in the branching ureteric bud (UB), and that renal agenesis occurs in homozygous Fras1 null mutant blebbed (bl) mice on a C57BL6J background. In vivo, the bl/bl bud fails to invade metanephric mesenchyme which undergoes involution, events replicated in organ culture. The expression of glial cell line-derived neurotrophic factor and growth-differentiation factor 11 was defective in bl/bl renal primordia in vivo, whereas, in culture, the addition of either growth factor restored bud invasion into the mesenchyme. Mutant primordia also showed deficient expression of Hoxd11 and Six2 transcription factors, whereas the activity of bone morphogenetic protein 4, an anti-branching molecule, was upregulated. In wild types, Fras1 was also expressed by nascent nephrons. Foetal glomerular podocytes expressed Fras1 transcripts and Fras1 immunolocalized in a glomerular BM-like pattern. On a mixed background, bl mutants, and also compound mutants for bl and my, another bleb strain, sometimes survive into adulthood. These mice have two kidneys, which contain subsets of glomeruli with perturbed nephrin, podocin, integrin α3 and fibronectin expression. Thus, Fras1 protein coats branching UB epithelia and is strikingly upregulated in the nephron lineage after mesenchymal/epithelial transition. Fras1 deficiency causes defective interactions between the bud and mesenchyme, correlating with disturbed expression of key nephrogenic molecules. Furthermore, Fras1 may also be required for the formation of normal glomeruli.
Collapse
Affiliation(s)
- Jolanta E Pitera
- Nephro-Urology Unit, UCL Institute of Child Health, London WC1 N 1EH, UK
| | | | | |
Collapse
|
236
|
Baleato RM, Guthrie PL, Gubler MC, Ashman LK, Roselli S. Deletion of CD151 results in a strain-dependent glomerular disease due to severe alterations of the glomerular basement membrane. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:927-37. [PMID: 18787104 DOI: 10.2353/ajpath.2008.071149] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alterations in CD151 have been associated with primary glomerular disease in both humans and mice, implicating CD151 as a key component of the glomerular filtration barrier. CD151 belongs to the tetraspanin family and associates with cell-matrix adhesion complexes such as alpha3beta1-integrin. Here we show that Cd151-deficient mice develop severe kidney disease on an FVB background but are healthy on a B6 background, providing a new and unique tool for the identification of genes that modulate the onset of proteinuria. To better understand the function of CD151 in the kidney, we studied its expression pattern and characterized early ultrastructural defects in Cd151-null kidneys. CD151 is expressed in podocytes of the mouse kidney and co-localizes with alpha3-integrin at the base of podocyte foot processes, at the site of anchorage to the glomerular basement membrane (GBM). Interestingly, the first ultrastructural lesions seen at the onset of proteinuria in Cd151-null kidneys were severe alterations of the GBM, reminiscent of Alport syndrome and consisting of massive thickening and splitting of the GBM. These lesions are associated with increased expression of GBM components. Podocyte abnormalities, effacement of foot processes, and podocyte loss appear to occur consequently to the GBM damage. In conclusion, CD151 appears to be involved in the establishment, maturation, and/or maintenance of the GBM structure in addition to its role in integrin-mediated adhesion strengthening.
Collapse
Affiliation(s)
- Rosa M Baleato
- School of Biomedical Sciences and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | | | | | | | | |
Collapse
|
237
|
Yamada M, Sumida Y, Fujibayashi A, Fukaguchi K, Sanzen N, Nishiuchi R, Sekiguchi K. The tetraspanin CD151 regulates cell morphology and intracellular signaling on laminin-511. FEBS J 2008; 275:3335-51. [DOI: 10.1111/j.1742-4658.2008.06481.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
238
|
Tanjore H, Zeisberg EM, Gerami-Naini B, Kalluri R. Beta1 integrin expression on endothelial cells is required for angiogenesis but not for vasculogenesis. Dev Dyn 2008; 237:75-82. [PMID: 18058911 DOI: 10.1002/dvdy.21385] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Integrins are a family of cell adhesion receptors that are involved in cell-matrix and cell-cell communications. They facilitate cell proliferation, migration, and survival. Using the Cre-Lox system, we deleted beta1 integrin on Tie2-positive (Tie2-cre beta1 Int (fl/fl)) vascular endothelial cells. Deletion of beta1 integrin on vascular endothelial cells results in embryonic lethality. Blood vessel defects are encountered in the Tie2-Cre beta1 Int (fl/fl) embryos at embryonic age (E9.5), and embryos die before reaching E10.5. The embryos exhibit growth retardation and both histological evaluation and PECAM-1 staining of E9.5 embryos revealed defects in angiogenic sprouting and vascular branching morphogenesis. Large and medium-size vessel formation is not affected in these embryos. Angiogenic defects were observed in several regions of the embryo and yolk sacs. These results indicate that beta1 integrin expression on vascular endothelial cells is crucial for embryonic angiogenesis but dispensable for vasculogenesis.
Collapse
Affiliation(s)
- Harikrishna Tanjore
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
239
|
Di-Poï N, Zákány J, Duboule D. Distinct roles and regulations for HoxD genes in metanephric kidney development. PLoS Genet 2008; 3:e232. [PMID: 18159948 PMCID: PMC2151092 DOI: 10.1371/journal.pgen.0030232] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2007] [Accepted: 11/14/2007] [Indexed: 11/24/2022] Open
Abstract
Hox genes encode homeodomain-containing proteins that control embryonic development in multiple contexts. Up to 30 Hox genes, distributed among all four clusters, are expressed during mammalian kidney morphogenesis, but functional redundancy between them has made a detailed functional account difficult to achieve. We have investigated the role of the HoxD cluster through comparative molecular embryological analysis of a set of mouse strains carrying targeted genomic rearrangements such as deletions, duplications, and inversions. This analysis allowed us to uncover and genetically dissect the complex role of the HoxD cluster. Regulation of metanephric mesenchyme-ureteric bud interactions and maintenance of structural integrity of tubular epithelia are differentially controlled by some Hoxd genes during renal development, consistent with their specific expression profiles. We also provide evidence for a kidney-specific form of colinearity that underlies the differential expression of two distinct sets of genes located on both sides and overlapping at the Hoxd9 locus. These insights further our knowledge of the genetic control of kidney morphogenesis and may contribute to understanding certain congenital kidney malformations, including polycystic kidney disease and renal hypoplasia. Hox genes encode proteins that control embryonic development along the head-to-tail axis and in multiple organs. Here, we show that several members of this gene family are necessary for the normal development of the mammalian kidneys. These genes are clustered in one site on the chromosome and their respective positions within the group determine which component of the kidneys they will contribute to. Using a large collection of engineered mutations in this system, we show that these genes are required both for the growth of the kidneys and for their proper organization, such that mutations in some genes reduce the size of the organs, whereas mutations in others induce polycystic kidneys. Our set of genetic rearrangements also allowed us to localize the position of regulatory sequences, which control the expression of these genes during kidney development.
Collapse
Affiliation(s)
- Nicolas Di-Poï
- Department of Zoology and Animal Biology, University of Geneva, Geneva, Swizerland
| | - József Zákány
- Department of Zoology and Animal Biology, University of Geneva, Geneva, Swizerland
| | - Denis Duboule
- Department of Zoology and Animal Biology, University of Geneva, Geneva, Swizerland
- School of Life Sciences, Ecole Polytechnique Fédérale Lausanne, Lausanne, Switzerland
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
240
|
Wallgard E, Larsson E, He L, Hellström M, Armulik A, Nisancioglu MH, Genove G, Lindahl P, Betsholtz C. Identification of a core set of 58 gene transcripts with broad and specific expression in the microvasculature. Arterioscler Thromb Vasc Biol 2008; 28:1469-76. [PMID: 18483404 DOI: 10.1161/atvbaha.108.165738] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Pathological angiogenesis is an integral component of many diseases. Antiangiogenesis and vascular targeting are therefore promising new therapeutic principles. However, few endothelial-specific putative drug targets have been identified, and information is still limited about endothelial-specific molecular processes. Here we aimed at determining the endothelial cell-specific core transcriptome in vivo. METHODS AND RESULTS Analysis of publicly available microarray data identified a mixed vascular/lung cluster of 132 genes that correlated with known endothelial markers. Filtering against kidney glomerular/nonglomerular and brain vascular/nonvascular microarray profiles separated contaminating lung markers, leaving 58 genes with broad and specific microvascular expression. More than half of these have not previously been linked to endothelial functions or studied in detail before. The endothelial cell-specific expression of a selected subset of these, Eltd1, Gpr116, Ramp2, Slc9a3r2, Slc43a3, Rasip1, and NM_023516, was confirmed by real-time quantitative polymerase chain reaction and/or immunohistochemistry. CONCLUSIONS We have used a combination of publicly available and own microarray data to identify 58 gene transcripts with broad yet specific expression in microvascular endothelium. Most of these have unknown functions, but many of them are predicted to be cell surface expressed or implicated in cell signaling processes and should therefore be explored as putative microvascular drug targets.
Collapse
Affiliation(s)
- Elisabet Wallgard
- Department of Medical Biochemistry and Biophysics, KarolinskaInstitutet, Scheeles väg 2, A3, floor 4, SE-171 77, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Pozzi A, Jarad G, Moeckel GW, Coffa S, Zhang X, Gewin L, Eremina V, Hudson BG, Borza DB, Harris RC, Holzman LB, Phillips CL, Fassler R, Quaggin SE, Miner JH, Zent R. Beta1 integrin expression by podocytes is required to maintain glomerular structural integrity. Dev Biol 2008; 316:288-301. [PMID: 18328474 PMCID: PMC2396524 DOI: 10.1016/j.ydbio.2008.01.022] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 01/09/2008] [Accepted: 01/15/2008] [Indexed: 11/22/2022]
Abstract
Integrins are transmembrane heteromeric receptors that mediate interactions between cells and extracellular matrix (ECM). beta1, the most abundantly expressed integrin subunit, binds at least 12 alpha subunits. beta1 containing integrins are highly expressed in the glomerulus of the kidney; however their role in glomerular morphogenesis and maintenance of glomerular filtration barrier integrity is poorly understood. To study these questions we selectively deleted beta1 integrin in the podocyte by crossing beta1(flox/flox) mice with podocyte specific podocin-cre mice (pod-Cre), which express cre at the time of glomerular capillary formation. We demonstrate that podocyte abnormalities are visualized during glomerulogenesis of the pod-Cre;beta1(flox/flox) mice and proteinuria is present at birth, despite a grossly normal glomerular basement membrane. Following the advent of glomerular filtration there is progressive podocyte loss and the mice develop capillary loop and mesangium degeneration with little evidence of glomerulosclerosis. By 3 weeks of age the mice develop severe end stage renal failure characterized by both tubulointerstitial and glomerular pathology. Thus, expression of beta1 containing integrins by the podocyte is critical for maintaining the structural integrity of the glomerulus.
Collapse
Affiliation(s)
- Ambra Pozzi
- Department of Medicine, Veterans Affairs Hospital, Vanderbilt University Medical Center, Nashville, 37232
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, 37232
| | - George Jarad
- Renal Division, Washington University School of Medicine, St. Louis, MO 63110
| | - Gilbert W. Moeckel
- Department of Pathology, Vanderbilt University Medical Center, Nashville, 37232
| | - Sergio Coffa
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
| | - Xi Zhang
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, 37232
| | - Leslie Gewin
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
| | - Vera Eremina
- Department of Maternal and Fetal Health, The Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Billy G. Hudson
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
| | - Dorin-Bogdan Borza
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
| | - Raymond C. Harris
- Department of Medicine, Veterans Affairs Hospital, Vanderbilt University Medical Center, Nashville, 37232
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
- Departments of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, 37232
| | - Lawrence B. Holzman
- Division of Nephrology, University of Michigan Medical School, Ann Arbor, Mich., USA
| | - Carrie L. Phillips
- Division of Nephrology and Department of Pathology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reinhard Fassler
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, 82152 Martinsried, Germany
| | - Susan E. Quaggin
- Department of Maternal and Fetal Health, The Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey H. Miner
- Renal Division, Washington University School of Medicine, St. Louis, MO 63110
| | - Roy Zent
- Department of Medicine, Veterans Affairs Hospital, Vanderbilt University Medical Center, Nashville, 37232
- Division of Nephrology, Departments of Medicine, Vanderbilt University Medical Center, Nashville, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, 37232
- Departments of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, 37232
| |
Collapse
|
242
|
Kreidberg JA. Integrins and matrix in the glomerulus: old mysteries and new insights. J Am Soc Nephrol 2008; 19:650-1. [PMID: 18322154 DOI: 10.1681/asn.2008020160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
243
|
Vaughan MR, Quaggin SE. How do mesangial and endothelial cells form the glomerular tuft? J Am Soc Nephrol 2008; 19:24-33. [PMID: 18178797 DOI: 10.1681/asn.2007040471] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The glomerular capillary tuft is a highly intricate and specialized microvascular bed that filters plasma water and solute to form urine. The mature glomerulus contains four cell types: Parietal epithelial cells that form Bowman's capsule, podocytes that cover the outermost layer of the glomerular filtration barrier, glycocalyx-coated fenestrated endothelial cells that are in direct contact with blood, and mesangial cells that sit between the capillary loops. Filtration begins only after the influx and organization of endothelial and mesangial cells in the developing glomerulus. Tightly coordinated movement and cross-talk between these cell types is required for the formation of a functional glomerular filtration barrier, and disruption of these processes has devastating consequences for early life. Current concepts of the role of mesangial and endothelial cells in formation of the capillary tuft are reviewed here.
Collapse
Affiliation(s)
- Michael R Vaughan
- Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | | |
Collapse
|
244
|
Bridgewater DJ, Dionne JM, Butt MJ, Pin CL, Matsell DG. The role of the type I insulin-like growth factor receptor (IGF-IR) in glomerular integrity. Growth Horm IGF Res 2008; 18:26-37. [PMID: 17689124 DOI: 10.1016/j.ghir.2007.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 06/15/2007] [Accepted: 06/18/2007] [Indexed: 10/23/2022]
Abstract
Insulin-like growth factors (IGFs) have been implicated in normal mammalian kidney development. To confirm a role for the IGF system in podocyte and glomerular integrity, we generated a transgenic mouse that expresses a dominant-negative type 1 IGF receptor (IGF-IR) and determined the structural and functional consequences. Using a 4.25kb fragment of the murine nephrin promoter, the dominant-negative construct was expressed exclusively in the kidney, confirmed by Southern blot and RT-PCR analysis. IGF-Ir486(FLAGstop) protein localized specifically to the glomerular podocyte based on FLAG immunohistochemistry and on co-localization with nephrin and podocin. Wild type and transgenic glomeruli expressed both the alpha- and beta-subunits of the endogenous IGF-IR, with normal expression of both nephrin and podocin. Although the animals were viable and phenotypically normal, histological analysis of the kidneys revealed abnormal and small glomeruli with dilated glomerular capillaries and condensed podocyte nuclei, while ultra-structural examination revealed diffuse but segmental podocyte foot process broadening, fusion, and effacement. Explanted glomeruli from transgenic animals demonstrated a significant inhibition of podocyte cell outgrowth when compared to controls. These studies suggest that IGF signaling is essential for maintaining the integrity of the podocyte and that alterations of IGF signaling may play a role in progressive glomerular disease.
Collapse
Affiliation(s)
- Darren J Bridgewater
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
245
|
Kanasaki K, Kanda Y, Palmsten K, Tanjore H, Lee SB, Lebleu VS, Gattone VH, Kalluri R. Integrin beta1-mediated matrix assembly and signaling are critical for the normal development and function of the kidney glomerulus. Dev Biol 2008; 313:584-93. [PMID: 18082680 PMCID: PMC3947521 DOI: 10.1016/j.ydbio.2007.10.047] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 10/08/2007] [Accepted: 10/29/2007] [Indexed: 12/01/2022]
Abstract
The human kidneys filter 180 l of blood every day via about 2.5 million glomeruli. The three layers of the glomerular filtration apparatus consist of fenestrated endothelium, specialized extracellular matrix known as the glomerular basement membrane (GBM) and the podocyte foot processes with their modified adherens junctions known as the slit diaphragm (SD). In this study we explored the contribution of podocyte beta1 integrin signaling for normal glomerular function. Mice with podocyte specific deletion of integrin beta1 (podocin-Cre beta1-fl/fl mice) are born normal but cannot complete postnatal renal development. They exhibit detectable proteinuria on day 1 and die within a week. The kidneys of podocin-Cre beta1-fl/fl mice exhibit normal glomerular endothelium but show severe GBM defects with multilaminations and splitting including podocyte foot process effacement. The integrin linked kinase (ILK) is a downstream mediator of integrin beta1 activity in epithelial cells. To further explore whether integrin beta1-mediated signaling facilitates proper glomerular filtration, we generated mice deficient of ILK in the podocytes (podocin-Cre ILK-fl/fl mice). These mice develop normally but exhibit postnatal proteinuria at birth and die within 15 weeks of age due to renal failure. Collectively, our studies demonstrate that podocyte beta1 integrin and ILK signaling is critical for postnatal development and function of the glomerular filtration apparatus.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Basement Membrane/pathology
- Basement Membrane/ultrastructure
- Carrier Proteins/urine
- Cell Cycle Proteins
- Crosses, Genetic
- DNA-Binding Proteins/urine
- Embryo, Mammalian
- Endothelium/metabolism
- Endothelium/ultrastructure
- Epithelial Cells/metabolism
- Epithelial Cells/ultrastructure
- Extracellular Matrix/metabolism
- Fluorescent Dyes/metabolism
- Gene Deletion
- Genes, Reporter
- Glomerulosclerosis, Focal Segmental/etiology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Indoles/metabolism
- Integrases/genetics
- Integrases/metabolism
- Integrin beta3/metabolism
- Intracellular Signaling Peptides and Proteins/urine
- Kidney Glomerulus/embryology
- Kidney Glomerulus/metabolism
- Kidney Glomerulus/ultrastructure
- Luminescent Proteins/metabolism
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/urine
- Mice
- Mice, Mutant Strains
- Mice, Transgenic
- Nuclear Proteins/urine
- Podocytes/enzymology
- Podocytes/metabolism
- Podocytes/ultrastructure
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proteinuria/etiology
- Proteinuria/physiopathology
- RNA Splicing Factors
- Renal Insufficiency/etiology
- Renal Insufficiency/physiopathology
- Rhodamines/metabolism
- Signal Transduction
- Tetraspanin 29
- Time Factors
- Transgenes
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- Keizo Kanasaki
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Yoshiko Kanda
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Kristin Palmsten
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Harikrishna Tanjore
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Soo Bong Lee
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Valerie S. Lebleu
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Vincent H. Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Raghu Kalluri
- Division of Matrix Biology and the Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA
| |
Collapse
|
246
|
Wei C, Möller CC, Altintas MM, Li J, Schwarz K, Zacchigna S, Xie L, Henger A, Schmid H, Rastaldi MP, Cowan P, Kretzler M, Parrilla R, Bendayan M, Gupta V, Nikolic B, Kalluri R, Carmeliet P, Mundel P, Reiser J. Modification of kidney barrier function by the urokinase receptor. Nat Med 2008; 14:55-63. [PMID: 18084301 DOI: 10.1038/nm1696] [Citation(s) in RCA: 422] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 11/20/2007] [Indexed: 12/12/2022]
Abstract
Podocyte dysfunction, represented by foot process effacement and proteinuria, is often the starting point for progressive kidney disease. Therapies aimed at the cellular level of the disease are currently not available. Here we show that induction of urokinase receptor (uPAR) signaling in podocytes leads to foot process effacement and urinary protein loss via a mechanism that includes lipid-dependent activation of alphavbeta3 integrin. Mice lacking uPAR (Plaur-/-) are protected from lipopolysaccharide (LPS)-mediated proteinuria but develop disease after expression of a constitutively active beta3 integrin. Gene transfer studies reveal a prerequisite for uPAR expression in podocytes, but not in endothelial cells, for the development of LPS-mediated proteinuria. Mechanistically, uPAR is required to activate alphavbeta3 integrin in podocytes, promoting cell motility and activation of the small GTPases Cdc42 and Rac1. Blockade of alphavbeta3 integrin reduces podocyte motility in vitro and lowers proteinuria in mice. Our findings show a physiological role for uPAR signaling in the regulation of kidney permeability.
Collapse
Affiliation(s)
- Changli Wei
- Nephrology Division and Program in Glomerular Disease, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Abstract
Polycystic kidney disease (PKD) is a diverse group of human monogenic lethal conditions inherited as autosomal dominant (AD) or recessive (AR) traits. Recent development of genetically engineered mouse models of ADPKD, ARPKD, and nephronophthisis/medullary cystic disease (NPHP) are providing additional insights into the molecular mechanisms governing of these disease processes as well as the developmental differentiation of the normal kidney. Genotypic and phenotypic mouse models are discussed and provide evidence for the fundamental involvement of cell-matrix, cell-cell, and primary cilia-lumen interactions, as well as epithelial proliferation, apoptosis, and polarization. Structure/function relationships between the PKD1, PKD2, PKHD1, and NPHP genes and proteins support the notion of a regulatory multiprotein cystic complex with a mechanosensory function that integrates signals from the extracellular environment. The plethora of intracellular signaling cascades that can impact renal cystic development suggest an exquisitely sensitive requirement for integrated downstream transduction and provide potential targets for therapeutic intervention. Appropriate genocopy models that faithfully recapitulate the phenotypic characteristics of the disease will be invaluable tools to analyze the effects of modifier genes and small molecule inhibitor therapies.
Collapse
|
248
|
Rahuel C, Filipe A, Ritie L, El Nemer W, Patey-Mariaud N, Eladari D, Cartron JP, Simon-Assmann P, Le Van Kim C, Colin Y. Genetic inactivation of the laminin alpha5 chain receptor Lu/BCAM leads to kidney and intestinal abnormalities in the mouse. Am J Physiol Renal Physiol 2007; 294:F393-406. [PMID: 18032551 DOI: 10.1152/ajprenal.00315.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lutheran blood group and basal cell adhesion molecule (Lu/BCAM) has been recognized as a unique receptor for laminin alpha5 chain in human red blood cells and as a coreceptor in epithelial, endothelial, and smooth muscle cells. Because limited information is available regarding the function of this adhesion glycoprotein in vivo, we generated Lu/BCAM-null mice and looked for abnormalities in red blood cells as well as in kidney and intestine, two tissues showing alteration in laminin alpha5 chain-deficient mice. We first showed that, in contrast to humans, wild-type murine red blood cells failed to express Lu/BCAM. Lu/BCAM-null mice were healthy and developed normally. However, although no alteration of the renal function was evidenced, up to 90% of the glomeruli from mutant kidneys exhibited abnormalities characterized by a reduced number of visible capillary lumens and irregular thickening of the glomerular basement membrane. Similarly, intestine analysis of mutant mice revealed smooth muscle coat thickening and disorganization. Because glomerular basement membrane and smooth muscle coat express laminin alpha5 chain and are in contact with cell types expressing Lu/BCAM in wild-type mice, these results provide evidence that Lu/BCAM, as a laminin receptor, is involved in vivo in the maintenance of normal basement membrane organization in the kidney and intestine.
Collapse
Affiliation(s)
- Cécile Rahuel
- Institut National de la Santé et de la Recherche Médicale, Unité 665, Institut National de la Transfusion Sanguine, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
D'Agati VD. Podocyte injury in focal segmental glomerulosclerosis: Lessons from animal models (a play in five acts). Kidney Int 2007; 73:399-406. [PMID: 17989648 DOI: 10.1038/sj.ki.5002655] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Genetic engineering in the mouse has ushered in a new era of disease modeling that has advanced our understanding of podocyte injury in the pathogenesis of focal segmental glomerulosclerosis. Historically, the major animal models of focal segmental glomerulosclerosis involve direct podocyte injury (exemplified by toxin models) and indirect podocyte injury due to adaptive responses (exemplified by renal ablation models). In both paradigms, recent evidence indicates that podocyte depletion is a major pathomechanism mediating proteinuria and glomerulosclerosis. Podocyte-specific toxin models support that podocyte loss is sufficient to cause focal segmental glomerulosclerosis in a dose-dependent manner. Knockout and transgenic models have provided proof of concept that mutations in specific podocyte proteins mediate genetic forms of focal segmental glomerulosclerosis. Transgenic models of HIV-associated nephropathy have helped to elucidate the role of direct viral infection and podocyte expression of viral gene products in the pathogenesis of this form of collapsing glomerulopathy. Taken together, emerging data support that injury directed to or inherent within the podocyte constitutes the critical event in diverse pathways to glomerulosclerosis.
Collapse
Affiliation(s)
- V D D'Agati
- Department of Pathology, Columbia University, College of Physicians & Surgeons, New York, New York 10032, USA.
| |
Collapse
|
250
|
Endothelial expression of beta1 integrin is required for embryonic vascular patterning and postnatal vascular remodeling. Mol Cell Biol 2007; 28:794-802. [PMID: 17984225 DOI: 10.1128/mcb.00443-07] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The largest subgroup of integrins is that containing the beta1 subunit. beta1 integrins have been implicated in a wide array of biological processes ranging from adhesion to cell growth, organogenesis, and mechanotransduction. Global deletion of beta1 integrin expression results in embryonic death at ca. embryonic day 5 (E5), a developmental time point too early to determine the effects of this integrin on vascular development. To elucidate the specific role of beta1 integrin in the vasculature, we conditionally deleted the beta1 gene in the endothelium. Homozygous deletion of beta1 integrins in the endothelium resulted in failure of normal vascular patterning, severe fetal growth retardation, and embryonic death at E9.5 to 10, although there were no overt effects on vasculogenesis. Heterozygous endothelial beta1 gene deletion did not diminish fetal or postnatal survival, but it reduced beta1 subunit expression in endothelial cells from adult mice by approximately 40%. These mice demonstrated abnormal vascular remodeling in response to experimentally altered in vivo blood flow and diminished vascularization in healing wounds. These data demonstrate that endothelial expression of beta1 integrin is required for developmental vascular patterning and that endothelial beta1 gene dosing has significant functional effects on vascular remodeling in the adult. Understanding how beta1 integrin expression is modulated may have significant clinical importance.
Collapse
|