Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Pathol ; 262(3): 296-309, 2024 03.
Article in English | MEDLINE | ID: mdl-38129319

ABSTRACT

The standard of care for patients with Alport syndrome (AS) is angiotensin-converting enzyme (ACE) inhibitors. In autosomal recessive Alport (ARAS) mice, ACE inhibitors double lifespan. We previously showed that deletion of Itga1 in Alport mice [double-knockout (DKO) mice] increased lifespan by 50%. This effect seemed dependent on the prevention of laminin 211-mediated podocyte injury. Here, we treated DKO mice with vehicle or ramipril starting at 4 weeks of age. Proteinuria and glomerular filtration rates were measured at 5-week intervals. Glomeruli were analyzed for laminin 211 deposition in the glomerular basement membrane (GBM) and GBM ultrastructure was analyzed using transmission electron microscopy (TEM). RNA sequencing (RNA-seq) was performed on isolated glomeruli at all time points and the results were compared with cultured podocytes overlaid (or not) with recombinant laminin 211. Glomerular filtration rate declined in ramipril-treated DKO mice between 30 and 35 weeks. Proteinuria followed these same patterns with normalization of foot process architecture in ramipril-treated DKO mice. RNA-seq revealed a decline in the expression of Foxc2, nephrin (Nphs1), and podocin (Nphs2) mRNAs, which was delayed in the ramipril-treated DKO mice. GBM accumulation of laminin 211 was delayed in ramipril-treated DKO mice, likely due to a role for α1ß1 integrin in CDC42 activation in Alport mesangial cells, which is required for mesangial filopodial invasion of the subendothelial spaces of the glomerular capillary loops. Ramipril synergized with Itga1 knockout, tripling lifespan compared with untreated ARAS mice. © 2023 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Nephritis, Hereditary , Podocytes , Humans , Mice , Animals , Integrin alpha1/genetics , Integrin alpha1/metabolism , Ramipril/pharmacology , Ramipril/metabolism , Longevity , Glomerular Basement Membrane/metabolism , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Podocytes/metabolism , Laminin/genetics , Laminin/metabolism , Mice, Knockout , Proteinuria/drug therapy , Proteinuria/genetics , Proteinuria/metabolism , Sequence Analysis, RNA
2.
J Pathol ; 258(1): 26-37, 2022 09.
Article in English | MEDLINE | ID: mdl-35607980

ABSTRACT

In Alport mice, activation of the endothelin A receptor (ETA R) in mesangial cells results in sub-endothelial invasion of glomerular capillaries by mesangial filopodia. Filopodia deposit mesangial matrix in the glomerular basement membrane (GBM), including laminin 211 which activates NF-κB, resulting in induction of inflammatory cytokines. Herein we show that collagen α1(III) is also deposited in the GBM. Collagen α1(III) localized to the mesangium in wild-type mice and was found in both the mesangium and the GBM in Alport mice. We show that collagen α1(III) activates discoidin domain receptor family, member 1 (DDR1) receptors both in vitro and in vivo. To elucidate whether collagen α1(III) might cause podocyte injury, cultured murine Alport podocytes were overlaid with recombinant collagen α1(III), or not, for 24 h and RNA was analyzed by RNA sequencing (RNA-seq). These same cells were subjected to siRNA knockdown for integrin α2 or DDR1 and the RNA was analyzed by RNA-seq. Results were validated in vivo using RNA-seq from RNA isolated from wild-type and Alport mouse glomeruli. Numerous genes associated with podocyte injury were up- or down-regulated in both Alport glomeruli and cultured podocytes treated with collagen α1(III), 18 of which have been associated previously with podocyte injury or glomerulonephritis. The data indicate α2ß1 integrin/DDR1 co-receptor signaling as the dominant regulatory mechanism. This may explain earlier studies where deletion of either DDR1 or α2ß1 integrin in Alport mice ameliorates renal pathology. © 2022 Boys Town National Research Hospital. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Nephritis, Hereditary , Podocytes , Animals , Basement Membrane/pathology , Collagen Type III , Collagen Type IV/genetics , Discoidin Domain Receptor 1/genetics , Glomerular Basement Membrane/pathology , Humans , Integrin alpha2beta1 , Mice , Mice, Knockout , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Podocytes/pathology , Pseudopodia/pathology , RNA
3.
PLoS One ; 15(8): e0237907, 2020.
Article in English | MEDLINE | ID: mdl-32822386

ABSTRACT

Previous work demonstrates that the hearing loss in Alport mice is caused by defects in the stria vascularis. As the animals age, progressive thickening of strial capillary basement membranes (SCBMs) occurs associated with elevated levels of extracellular matrix expression and hypoxia-related gene and protein expression. These conditions render the animals susceptible to noise-induced hearing loss. In an effort to develop a more comprehensive understanding of how the underlying mutation in the COL4A3 gene influences homeostasis in the stria vascularis, we performed vascular permeability studies combined with RNA-seq analysis using isolated stria vascularis from 7-week old wild-type and Alport mice on the 129 Sv background. Alport SCBMs were found to be less permeable than wild-type littermates. RNA-seq and bioinformatics analysis revealed 68 genes were induced and 61 genes suppressed in the stria from Alport mice relative to wild-type using a cut-off of 2-fold. These included pathways involving transcription factors associated with the regulation of pro-inflammatory responses as well as cytokines, chemokines, and chemokine receptors that are up- or down-regulated. Canonical pathways included modulation of genes associated with glucose and glucose-1-PO4 degradation, NAD biosynthesis, histidine degradation, calcium signaling, and glutamate receptor signaling (among others). In all, the data point to the Alport stria being in an inflammatory state with disruption in numerous metabolic pathways indicative of metabolic stress, a likely cause for the susceptibility of Alport mice to noise-induced hearing loss under conditions that do not cause permanent hearing loss in age/strain-matched wild-type mice. The work lays the foundation for studies aimed at understanding the nature of strial pathology in Alport mice. The modulation of these genes under conditions of therapeutic intervention may provide important pre-clinical data to justify trials in humans afflicted with the disease.


Subject(s)
Gene Expression Regulation/genetics , Hearing Loss, Noise-Induced/metabolism , Nephritis, Hereditary/metabolism , Stria Vascularis/metabolism , Animals , Autoantigens/genetics , Autoantigens/metabolism , Basement Membrane/metabolism , Chemokines/genetics , Chemokines/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Extracellular Matrix/metabolism , Female , Glucose/genetics , Glucose/metabolism , Hearing Loss, Noise-Induced/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , RNA-Seq , Signal Transduction/genetics , Stria Vascularis/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Up-Regulation
4.
Hear Res ; 390: 107935, 2020 05.
Article in English | MEDLINE | ID: mdl-32234583

ABSTRACT

In 129 Sv autosomal Alport mice, the strial capillary basement membranes (SCBMs) progressively thicken between 5 and 9 weeks of age resulting in a hypoxic microenvironment with metabolic stress and induction of pro-inflammatory cytokines and chemokines. These events occur concomitant with a drop in endocochlear potential and a susceptibility to noise-induced hearing loss under conditions that do not permanently affect age/strain-matched littermates. Here we aimed to gain an understanding of events that occur before the onset of SCBM thickening. Alport stria has normal thickness and shows levels of extracellular matrix (ECM) molecules in the SCBMs commensurate with wild-type mice. Hearing thresholds in the 3-week Alport mice do not differ from those of wild-type mice. We performed RNAseq analysis using RNA from stria vascularis isolated from 3-week Alport mice and wild type littermates. Data was processed using Ingenuity Pathway Analysis software and further distilled using manual procedures. RNAseq analysis revealed significant dysregulation of genes involved in cell adhesion, cell migration, formation of protrusions, and both actin and tubulin cytoskeletal dynamics. Overall, the data suggested changes in the cellular architecture of the stria might be apparent. To test this notion, we performed dual immunofluorescence analysis on whole mounts of the stria vascularis from these same animals stained with anti-isolectin gs-ib4 (endothelial cell marker) and anti-desmin (pericyte marker) antibodies. The results showed evidence of pericyte detachment and migration as well as the formation of membrane ruffling on pericytes in z-stacked confocal images from Alport mice compared to wild type littermates. This was confirmed by TEM analysis. Earlier work from our lab showed that endothelin A receptor blockade prevents SCBM thickening and ECM accumulation in the SCBMs. Treating cultured pericytes with endothelin-1 induced actin cytoskeletal rearrangement, increasing the ratio of filamentous to globular actin. Collectively, these findings suggest that the change in type IV collagen composition in the Alport SCBMs results in cellular insult to the pericyte compartment, activating detachment and altered cytoskeletal dynamics. These events precede SCBM thickening and hearing loss in Alport mice, and thus constitute the earliest event so far recognized in Alport strial pathology.


Subject(s)
Actin Cytoskeleton/ultrastructure , Basement Membrane/ultrastructure , Nephritis, Hereditary/pathology , Pericytes/ultrastructure , Stria Vascularis/ultrastructure , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Basement Membrane/drug effects , Basement Membrane/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Endothelin-1/pharmacology , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Male , Mice, 129 Strain , Microscopy, Confocal , Microscopy, Electron, Transmission , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Pericytes/drug effects , Pericytes/metabolism , RNA-Seq , Receptor, Endothelin A/agonists , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Signal Transduction , Stria Vascularis/drug effects , Stria Vascularis/metabolism
5.
Kidney Int ; 94(2): 303-314, 2018 08.
Article in English | MEDLINE | ID: mdl-29759420

ABSTRACT

Lysyl oxidase like-2 (LOXL2) is an amine oxidase with both intracellular and extracellular functions. Extracellularly, LOXL2 promotes collagen and elastin crosslinking, whereas intracellularly, LOXL2 has been reported to modify histone H3, stabilize SNAIL, and reduce cell polarity. Although LOXL2 promotes liver and lung fibrosis, little is known regarding its role in renal fibrosis. Here we determine whether LOXL2 influences kidney disease in COL4A3 (-/-) Alport mice. These mice were treated with a small molecule inhibitor selective for LOXL2 or with vehicle and assessed for glomerular sclerosis and fibrosis, albuminuria, blood urea nitrogen, lifespan, pro-fibrotic gene expression and ultrastructure of the glomerular basement membrane. Laminin α2 deposition in the glomerular basement membrane and mesangial filopodial invasion of the glomerular capillaries were also assessed. LOXL2 inhibition significantly reduced interstitial fibrosis and mRNA expression of MMP-2, MMP-9, TGF-ß1, and TNF-α. LOXL2 inhibitor treatment also reduced glomerulosclerosis, expression of MMP-10, MMP-12, and MCP-1 mRNA in glomeruli, and decreased albuminuria and blood urea nitrogen. Mesangial filopodial invasion of the capillary tufts was blunted, as was laminin α2 deposition in the glomerular basement membrane, and glomerular basement membrane ultrastructure was normalized. There was no effect on lifespan. Thus, LOXL2 plays an important role in promoting both glomerular and interstitial pathogenesis associated with Alport syndrome in mice. Other etiologies of chronic kidney disease are implicated with our observations.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Enzyme Inhibitors/therapeutic use , Glomerular Basement Membrane/pathology , Glomerular Mesangium/pathology , Nephritis, Hereditary/pathology , Amino Acid Oxidoreductases/antagonists & inhibitors , Amino Acid Oxidoreductases/genetics , Animals , Autoantigens/genetics , Collagen Type IV/genetics , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/pharmacology , Fibrosis , Glomerular Basement Membrane/metabolism , Glomerular Mesangium/metabolism , Humans , Laminin/metabolism , Mice , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , RNA, Messenger/metabolism , Up-Regulation
6.
Hear Res ; 341: 100-108, 2016 11.
Article in English | MEDLINE | ID: mdl-27553900

ABSTRACT

Alport syndrome, a type IV collagen disorder, manifests as glomerular disease associated with hearing loss with thickening of the glomerular and strial capillary basement membranes (SCBMs). We have identified a role for endothelin-1 (ET-1) activation of endothelin A receptors (ETARs) in glomerular pathogenesis. Here we explore whether ET-1 plays a role in strial pathology. Wild type (WT) and Alport mice were treated with the ETAR antagonist, sitaxentan. The stria vascularis was analyzed for SCBM thickness and for extracellular matrix (ECM) proteins. Additional WT and Alport mice were exposed to noise or hypoxia and the stria analyzed for hypoxia-related and ECM genes. A strial marginal cell line cultured under hypoxic conditions, or stimulated with ET-1 was analyzed for expression of hypoxia-related and ECM transcripts. Noise exposure resulted in significantly elevated ABR thresholds in Alport mice relative to wild type littermates. Alport stria showed elevated expression of collagen α1(IV), laminin α2, and laminin α5 proteins relative to WT. SCBM thickening and elevated ECM protein expression was ameliorated by ETAR blockade. Stria from normoxic Alport mice and hypoxic WT mice showed upregulation of hypoxia-related, ECM, and ET-1 transcripts. Both ET-1 stimulation and hypoxia up-regulated ECM transcripts in cultured marginal cells. We conclude that ET-1 mediated activation of ETARs on strial marginal cells results in elevated expression of ECM genes and thickening of the SCBMs in Alport mice. SCBM thickening results in hypoxic stress further elevating ECM and ET-1 gene expression, exacerbating strial pathology.


Subject(s)
Endothelin-1/metabolism , Extracellular Matrix/genetics , Nephritis, Hereditary/physiopathology , Stria Vascularis/cytology , Animals , Basement Membrane/pathology , Body Temperature , Capillaries/pathology , Cell Line , Collagen Type IV/metabolism , Disease Models, Animal , Gene Expression Regulation , Hypoxia/pathology , Isoxazoles/chemistry , Laminin/metabolism , Mice , Oxidative Stress , Phenotype , Stria Vascularis/metabolism , Thiophenes/chemistry
7.
Kidney Int ; 90(2): 300-310, 2016 08.
Article in English | MEDLINE | ID: mdl-27165837

ABSTRACT

Recent work demonstrates that Alport glomerular disease is mediated through a biomechanical strain-sensitive activation of mesangial actin dynamics. This occurs through a Rac1/CDC42 cross-talk mechanism that results in the invasion of the subcapillary spaces by mesangial filopodia. The filopodia deposit mesangial matrix proteins in the glomerular basement membrane, including laminin 211, which activates focal adhesion kinase in podocytes culminating in the up-regulation of proinflammatory cytokines and metalloproteinases. These events drive the progression of glomerulonephritis. Here we test whether endothelial cell-derived endothelin-1 is up-regulated in Alport glomeruli and further elevated by hypertension. Treatment of cultured mesangial cells with endothelin-1 activates the formation of drebrin-positive actin microspikes. These microspikes do not form when cells are treated with the endothelin A receptor antagonist sitaxentan or under conditions of small, interfering RNA knockdown of endothelin A receptor mRNA. Treatment of Alport mice with sitaxentan results in delayed onset of proteinuria, normalized glomerular basement membrane morphology, inhibition of mesangial filopodial invasion of the glomerular capillaries, normalization of glomerular expression of metalloproteinases and proinflammatory cytokines, increased life span, and prevention of glomerulosclerosis and interstitial fibrosis. Thus endothelin A receptor activation on mesangial cells is a key event in initiation of Alport glomerular disease in this model.


Subject(s)
Endothelin-1/metabolism , Mesangial Cells/metabolism , Nephritis, Hereditary/metabolism , Podocytes/metabolism , Receptor, Endothelin A/metabolism , Animals , Biomechanical Phenomena , Disease Models, Animal , Endothelial Cells/metabolism , Endothelin Receptor Antagonists/pharmacology , Endothelin Receptor Antagonists/therapeutic use , Fluorescent Antibody Technique , Gene Knockdown Techniques , Glomerular Basement Membrane/metabolism , Hypertension/metabolism , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Laminin/metabolism , Mesangial Cells/drug effects , Mice , Mice, Inbred C57BL , Nephritis, Hereditary/genetics , Proteinuria/drug therapy , Pseudopodia/physiology , RNA Interference , RNA, Small Interfering/genetics , Receptor, Endothelin A/genetics , Signal Transduction , Thiophenes/pharmacology , Thiophenes/therapeutic use , Up-Regulation
8.
Biol Direct ; 9(1): 20, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25319552

ABSTRACT

BACKGROUND: The rhesus macaque (Macaca mulatta) is a key species for advancing biomedical research. Like all draft mammalian genomes, the draft rhesus assembly (rheMac2) has gaps, sequencing errors and misassemblies that have prevented automated annotation pipelines from functioning correctly. Another rhesus macaque assembly, CR_1.0, is also available but is substantially more fragmented than rheMac2 with smaller contigs and scaffolds. Annotations for these two assemblies are limited in completeness and accuracy. High quality assembly and annotation files are required for a wide range of studies including expression, genetic and evolutionary analyses. RESULTS: We report a new de novo assembly of the rhesus macaque genome (MacaM) that incorporates both the original Sanger sequences used to assemble rheMac2 and new Illumina sequences from the same animal. MacaM has a weighted average (N50) contig size of 64 kilobases, more than twice the size of the rheMac2 assembly and almost five times the size of the CR_1.0 assembly. The MacaM chromosome assembly incorporates information from previously unutilized mapping data and preliminary annotation of scaffolds. Independent assessment of the assemblies using Ion Torrent read alignments indicates that MacaM is more complete and accurate than rheMac2 and CR_1.0. We assembled messenger RNA sequences from several rhesus tissues into transcripts which allowed us to identify a total of 11,712 complete proteins representing 9,524 distinct genes. Using a combination of our assembled rhesus macaque transcripts and human transcripts, we annotated 18,757 transcripts and 16,050 genes with complete coding sequences in the MacaM assembly. Further, we demonstrate that the new annotations provide greatly improved accuracy as compared to the current annotations of rheMac2. Finally, we show that the MacaM genome provides an accurate resource for alignment of reads produced by RNA sequence expression studies. CONCLUSIONS: The MacaM assembly and annotation files provide a substantially more complete and accurate representation of the rhesus macaque genome than rheMac2 or CR_1.0 and will serve as an important resource for investigators conducting next-generation sequencing studies with nonhuman primates. REVIEWERS: This article was reviewed by Dr. Lutz Walter, Dr. Soojin Yi and Dr. Kateryna Makova.


Subject(s)
Genome , Macaca mulatta/genetics , Amino Acid Sequence , Animals , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Molecular Sequence Annotation , Molecular Sequence Data , RNA, Messenger/metabolism , Sequence Alignment
9.
PLoS One ; 9(6): e99083, 2014.
Article in English | MEDLINE | ID: mdl-24915008

ABSTRACT

It has been known for some time that laminins containing α1 and α2 chains, which are normally restricted to the mesangial matrix, accumulate in the glomerular basement membranes (GBM) of Alport mice, dogs, and humans. We show that laminins containing the α2 chain, but not those containing the α1 chain activates focal adhesion kinase (FAK) on glomerular podocytes in vitro and in vivo. CD151-null mice, which have weakened podocyte adhesion to the GBM rendering these mice more susceptible to biomechanical strain in the glomerulus, also show progressive accumulation of α2 laminins in the GBM, and podocyte FAK activation. Analysis of glomerular mRNA from both models demonstrates significant induction of MMP-9, MMP-10, MMP-12, MMPs linked to GBM destruction in Alport disease models, as well as the pro-inflammatory cytokine IL-6. SiRNA knockdown of FAK in cultured podocytes significantly reduced expression of MMP-9, MMP-10 and IL-6, but not MMP-12. Treatment of Alport mice with TAE226, a small molecule inhibitor of FAK activation, ameliorated fibrosis and glomerulosclerosis, significantly reduced proteinuria and blood urea nitrogen levels, and partially restored GBM ultrastructure. Glomerular expression of MMP-9, MMP-10 and MMP-12 mRNAs was significantly reduced in TAE226 treated animals. Collectively, this work identifies laminin α2-mediated FAK activation in podocytes as an important early event in Alport glomerular pathogenesis and suggests that FAK inhibitors, if safe formulations can be developed, might be employed as a novel therapeutic approach for treating Alport renal disease in its early stages.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Laminin/metabolism , Nephritis, Hereditary/enzymology , Nephritis, Hereditary/etiology , Animals , Biomechanical Phenomena/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Gene Knockdown Techniques , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/enzymology , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/ultrastructure , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Kinetics , Matrix Metalloproteinases/metabolism , Mice, Knockout , Morpholines/pharmacology , Morpholines/therapeutic use , NF-KappaB Inhibitor alpha , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/pathology , Podocytes/enzymology , Podocytes/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Tetraspanin 24/metabolism
10.
Am J Pathol ; 183(4): 1269-1280, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23911822

ABSTRACT

Alport syndrome, hereditary glomerulonephritis with hearing loss, results from mutations in type IV collagen COL4A3, COL4A4, or COL4A5 genes. The mechanism for delayed glomerular disease onset is unknown. Comparative analysis of Alport mice and CD151 knockout mice revealed progressive accumulation of laminin 211 in the glomerular basement membrane. We show mesangial processes invading the capillary loops of both models as well as in human Alport glomeruli, as the likely source of this laminin. L-NAME salt-induced hypertension accelerated mesangial cell process invasion. Cultured mesangial cells showed reduced migratory potential when treated with either integrin-linked kinase inhibitor or Rac1 inhibitor, or by deletion of integrin α1. Treatment of Alport mice with Rac1 inhibitor or deletion of integrin α1 reduced mesangial cell process invasion of the glomerular capillary tuft. Laminin α2-deficient Alport mice show reduced mesangial process invasion, and cultured laminin α2-null cells showed reduced migratory potential, indicating a functional role for mesangial laminins in progression of Alport glomerular pathogenesis. Collectively, these findings predict a role for biomechanical insult in the induction of integrin α1ß1-dependent Rac1-mediated mesangial cell process invasion of the glomerular capillary tuft as an initiation mechanism of Alport glomerular pathology.


Subject(s)
Capillaries/pathology , Glomerular Mesangium/blood supply , Glomerular Mesangium/pathology , Integrin alpha1beta1/metabolism , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Biomechanical Phenomena/drug effects , Capillaries/drug effects , Capillaries/metabolism , Capillaries/physiopathology , Cell Movement/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Deletion , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/physiopathology , Glomerular Basement Membrane/ultrastructure , Glomerular Mesangium/physiopathology , Glomerular Mesangium/ultrastructure , Humans , Hypertension/complications , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Laminin/metabolism , Mice , Mice, Knockout , Nephritis, Hereditary/complications , Nephritis, Hereditary/physiopathology , Protein Transport/drug effects , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
11.
J Neurosci ; 32(40): 13841-59, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035094

ABSTRACT

Usher syndrome is a genetically heterogeneous disorder characterized by hearing and balance dysfunction and progressive retinitis pigmentosa. Mouse models carrying mutations for the nine Usher-associated genes have splayed stereocilia, and some show delayed maturation of ribbon synapses suggesting these proteins may play different roles in terminal differentiation of auditory hair cells. The presence of the Usher proteins at the basal and apical aspects of the neurosensory epithelia suggests the existence of regulated trafficking through specific transport proteins and routes. Immature mouse cochleae and UB/OC-1 cells were used in this work to address whether specific variants of PCDH15 and VLGR1 are being selectively transported to opposite poles of the hair cells. Confocal colocalization studies between apical and basal vesicular markers and the different PCDH15 and VLGR1 variants along with sucrose density gradients and the use of vesicle trafficking inhibitors show the existence of Usher protein complexes in at least two vesicular subpools. The apically trafficked pool colocalized with the early endosomal vesicle marker, rab5, while the basally trafficked pool associated with membrane microdomains and SNAP25. Moreover, coimmunoprecipitation experiments between SNAP25 and VLGR1 show a physical interaction of these two proteins in organ of Corti and brain. Collectively, these findings establish the existence of a differential vesicular trafficking mechanism for specific Usher protein variants in mouse cochlear hair cells, with the apical variants playing a potential role in endosomal recycling and stereocilia development/maintenance, and the basolateral variants involved in vesicle docking and/or fusion through SNAP25-mediated interactions.


Subject(s)
Cadherins/metabolism , Cell Polarity/physiology , Hair Cells, Auditory/ultrastructure , Protein Precursors/metabolism , Receptors, G-Protein-Coupled/metabolism , Transport Vesicles/physiology , ADP-Ribosylation Factor 1/analysis , Animals , Brain Chemistry , Cadherin Related Proteins , Cadherins/biosynthesis , Cadherins/genetics , Cell Compartmentation , Cell Differentiation , Disease Models, Animal , Gene Knockdown Techniques , Hair Cells, Auditory/metabolism , Immunoprecipitation , Mice , Mice, Neurologic Mutants , Mutation , Organ of Corti/chemistry , Organ of Corti/ultrastructure , Protein Interaction Mapping , Protein Precursors/biosynthesis , Protein Precursors/genetics , Protein Transport/drug effects , RNA Interference , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Structure-Activity Relationship , Synaptosomal-Associated Protein 25/analysis , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/metabolism , Transport Vesicles/chemistry , Usher Syndromes/metabolism , rab5 GTP-Binding Proteins/analysis
12.
PLoS One ; 7(2): e30573, 2012.
Article in English | MEDLINE | ID: mdl-22363448

ABSTRACT

The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1-/- mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzer(av3J) mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.


Subject(s)
Cadherins/metabolism , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Protein Precursors/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism , Adsorption , Animals , Antibodies/immunology , Cadherin Related Proteins , Cell Line , Cell Polarity , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Multiprotein Complexes/metabolism , Protein Isoforms/metabolism , Synapses/ultrastructure , Time Factors
13.
Am J Pathol ; 177(5): 2527-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20864678

ABSTRACT

Alport syndrome is a common hereditary basement membrane disorder caused by mutations in the collagen IV α3, α4, or α5 genes that results in progressive glomerular and interstitial renal disease. Interstitial monocytes that accumulate in the renal cortex from Alport mice are immunopositive for integrin α1ß1, while only a small fraction of circulating monocytes are immunopositive for this integrin. We surmised that such a disparity might be due to the selective recruitment of α1ß1-positive monocytes. In this study, we report the identification of collagen XIII as a ligand that facilitates this selective recruitment of α1ß1 integrin-positive monocytes. Collagen XIII is absent in the vascular endothelium from normal renal cortex and abundant in Alport renal cortex. Neutralizing antibodies against the binding site in collagen XIII for α1ß1 integrin selectively block VLA1-positive monocyte migration in transwell assays. Injection of these antibodies into Alport mice slows monocyte recruitment and protects against renal fibrosis. Thus, the induction of collagen XIII in endothelial cells of Alport kidneys mediates the selective recruitment of α1ß1 integrin-positive monocytes and may potentially serve as a therapeutic target for inflammatory diseases in which lymphocyte/monocyte recruitment involves the interaction with α1ß1 integrin.


Subject(s)
Collagen Type XIII/metabolism , Endothelium, Vascular/metabolism , Integrin alpha1beta1/metabolism , Monocytes/physiology , Nephritis, Hereditary/pathology , Nephritis, Hereditary/physiopathology , Transendothelial and Transepithelial Migration/physiology , Animals , Antibodies/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , CHO Cells , Cells, Cultured , Collagen Type XIII/genetics , Cricetinae , Cricetulus , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Fibrosis , Integrin alpha1beta1/genetics , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Monocytes/cytology
14.
Kidney Int ; 76(9): 968-76, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710627

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.


Subject(s)
Glomerular Basement Membrane/metabolism , Interleukin-6/genetics , Kidney Glomerulus/metabolism , Matrix Metalloproteinases/genetics , Nephritis, Hereditary/genetics , Podocytes/metabolism , Adaptation, Physiological/genetics , Animals , Blood Pressure , Cells, Cultured , Cytoskeleton/metabolism , Disease Models, Animal , Gene Expression Regulation , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Interleukin-6/metabolism , Kidney Glomerulus/physiopathology , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/physiopathology , Proteinuria/chemically induced , Proteinuria/genetics , Proteinuria/physiopathology , RNA, Messenger/metabolism , Sodium Chloride, Dietary , Stress, Mechanical , Time Factors
15.
Hear Res ; 255(1-2): 109-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19539019

ABSTRACT

The Usher syndrome 3A (CLRN1) gene encodes clarin-1, which is a member of the tetraspanin family of transmembrane proteins. Although identified more than 6 years ago, little is known about its localization or function in the eye and ear. We developed a polyclonal antibody that react with all clarin-1 isoforms and used it to characterize protein expression in cochlea and retina. In the cochlea, we observe clarin-1expression in the stereocilia of P0 mice, and in synaptic terminals present at the base of the auditory hair cells from E18 to P6. In the retina, clarin-1 localizes to the connecting cilia, inner segment of photoreceptors and to the ribbon synapses. RT-PCR from P0 cochlea and P28 retina show mRNAs encoding only isoforms 2 and 3. Western blots show that only isoform 2 is present in protein extracts from these same tissues. We examined clarin-1 expression in the immortomouse-derived hair cell line UB/OC-1. Only isoform 2 is expressed in UB/OC-1 at both mRNA and protein levels, suggesting this isoform is biologically relevant to hair cell function. The protein co-localizes with microtubules and post-transgolgi vesicles. The subcellular localization of clarin-1 in hair cells and photoreceptors suggests it functions at both the basal and apical poles of neurosensoriepithelia.


Subject(s)
Hair Cells, Auditory/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Animals , Animals, Newborn , Base Sequence , Cell Line , Cochlea/embryology , Cochlea/growth & development , Cochlea/metabolism , DNA Primers/genetics , Female , Gene Expression , Gestational Age , Membrane Proteins/deficiency , Mice , Mice, Knockout , Microscopy, Fluorescence , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics
16.
Invest Ophthalmol Vis Sci ; 49(10): 4647-54, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18614805

ABSTRACT

PURPOSE: The role of integrin/cell matrix interactions between the RPE and the basement membrane in retinal maintenance and function is not well characterized. In this study the functional importance of alpha1beta1 integrin for retinal pigment epithelial cell homeostasis and retinal health was assessed by comparing alpha1 integrin knockout mice with strain- and age-matched wild-type mice. METHODS: Immunolocalization and Western blot analysis of retinas and ARPE19 cells were performed to examine the expression of alpha1beta1 integrin in the RPE. Retinal abnormality was assessed by funduscopy, histology, and transmission electron microscopy. Progressive retinal damage was quantified by direct counting of rod photoreceptors. Light-induced translocation of arrestin and alpha-transducin was documented by immunohistochemical analysis of retinal cryosections. RESULTS: Integrin alpha1beta1 localizes to the basal aspect of retinal pigment epithelial cells colocalizing with the basal lamina of the RPE. Integrin alpha1-null mice have delayed-onset progressive retinal degeneration associated with thickening of the basement membrane, dysmorphology of basal processes, synaptic malformations, and funduscopic abnormalities. Integrin alpha1-null mice display marked delays in transducin translocation compared with dark-adapted wild-type mice after exposure to light. CONCLUSIONS: Collectively, these data suggest an essential role for alpha1beta1 integrin/basement membrane interactions in the RPE in basement membrane metabolism and translocation of transducin in photoreceptors. This is the first report describing evidence supporting an essential role for integrin/basement membrane interaction in the RPE. Further, this report demonstrates a direct link between integrin alpha1beta1 function in retinal pigment epithelial and molecular defects in photoreceptor cell function before retinal abnormality is apparent.


Subject(s)
Integrin alpha1beta1/physiology , Photoreceptor Cells, Vertebrate/ultrastructure , Pigment Epithelium of Eye/metabolism , Presynaptic Terminals/ultrastructure , Retinal Degeneration/metabolism , Animals , Arrestin/metabolism , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blotting, Western , Cell Count , Cell Line , Dark Adaptation , Fluorescent Antibody Technique, Indirect , Homeostasis/physiology , Humans , Mice , Mice, Knockout , Microscopy, Fluorescence , Pigment Epithelium of Eye/ultrastructure , Retinal Degeneration/pathology , Signal Transduction/physiology , Transducin/metabolism
17.
BMC Biotechnol ; 8: 31, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18366794

ABSTRACT

BACKGROUND: Gene targeting in nonhuman primates has the potential to produce critical animal models for translational studies related to human diseases. Successful gene targeting in fibroblasts followed by somatic cell nuclear transfer (SCNT) has been achieved in several species of large mammals but not yet in primates. Our goal was to establish the protocols necessary to achieve gene targeting in primary culture of adult rhesus macaque fibroblasts as a first step in creating nonhuman primate models of genetic disease using nuclear transfer technology. RESULTS: A primary culture of adult male fibroblasts was transfected with hTERT to overcome senescence and allow long term in vitro manipulations. Successful gene targeting of the HPRT locus in rhesus macaques was achieved by electroporating S-phase synchronized cells with a construct containing a SV40 enhancer. CONCLUSION: The cell lines reported here could be used for the production of null mutant rhesus macaque models of human genetic disease using SCNT technology. In addition, given the close evolutionary relationship and biological similarity between rhesus macaques and humans, the protocols described here may prove useful in the genetic engineering of human somatic cells.


Subject(s)
Electroporation/methods , Fibroblasts/cytology , Fibroblasts/physiology , Gene Targeting/methods , Macaca mulatta/genetics , Transfection/methods , Animals , Cells, Cultured
18.
Am J Pathol ; 172(3): 761-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18258846

ABSTRACT

Previous work has shown that integrin alpha1-null Alport mice exhibit attenuated glomerular disease with decreased matrix accumulation and live much longer than strain-matched Alport mice. However, the mechanism underlying this observation is unknown. Here we show that glomerular gelatinase expression, specifically matrix metalloproteinase-2 (MMP-2), MMP-9, and MMP-14, was significantly elevated in both integrin alpha1-null mice and integrin alpha1-null Alport mice relative to wild-type mice; however, only MMP-9 was elevated in glomeruli of Alport mice that express integrin alpha1. Similarly, cultured mesangial cells from alpha1-null mice showed elevated expression levels of all three MMPs, whereas mesangial cells from Alport mice show elevated expression levels of only MMP-9. In both glomeruli and cultured mesangial cells isolated from integrin alpha1-null mice, activation of the p38 and ERK branches of the mitogen-activated protein kinase pathway was also observed. The use of small molecule inhibitors demonstrated that the activation of the p38, but not ERK, pathway was linked to elevated MMP-2, -9, and -14 expression levels in mesangial cells from integrin alpha1-null mice. In contrast, elevated MMP-9 levels in mesangial cells from Alport mice were linked to ERK pathway activation. Blockade of gelatinase activity using a small molecule inhibitor (BAY-12-9566) ameliorated progression of proteinuria and restored the architecture of the glomerular basement membrane in alpha1 integrin-null Alport mice, suggesting that elevated gelatinase activity exacerbates glomerular disease progression in these mice.


Subject(s)
Gene Expression Regulation, Enzymologic , Integrin alpha1beta1/physiology , Matrix Metalloproteinases/genetics , Mesangial Cells/enzymology , Nephritis, Hereditary/genetics , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Autoantigens/genetics , Biphenyl Compounds , Cells, Cultured , Collagen Type IV/genetics , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Integrin alpha1beta1/genetics , Matrix Metalloproteinases/metabolism , Mesangial Cells/metabolism , Mice , Mice, Knockout , Nephritis, Hereditary/enzymology , Nephritis, Hereditary/pathology , Organic Chemicals/pharmacology , Phenylbutyrates , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
19.
Am J Pathol ; 169(1): 32-46, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816359

ABSTRACT

Alport syndrome is a glomerular basement membrane (GBM) disease caused by mutations in type IV collagen genes. A unique irregular thickening and thinning of the GBM characterizes the progressive glomerular pathology. The metabolic imbalances responsible for these GBM irregularities are not known. Here we show that macrophage metalloelastase (MMP-12) expression is >40-fold induced in glomeruli from Alport mice and is markedly induced in glomeruli of both humans and dogs with Alport syndrome. Treatment of Alport mice with MMI270 (CGS27023A), a broad spectrum MMP inhibitor that blocks MMP-12 activity, results in largely restored GBM ultrastructure and function. Treatment with BAY-129566, a broad spectrum MMP inhibitor that does not inhibit MMP-12, had no effect. We show that inhibition of CC chemokine receptor 2 (CCR2) receptor signaling with propagermanium blocks induction of MMP-12 mRNA and prevents GBM damage. CCR2 receptor is expressed in glomerular podocytes of Alport mice, suggesting MCP-1 activation of CCR2 on podocytes may underlie induction of MMP-12. These data indicate that the irregular GBM that characterizes Alport syndrome may be mediated, in part, by focal degradation of the GBM due to MMP dysregulation, in particular, MMP-12. Thus, MMP-12/CCR2 inhibitors may provide a novel and effective therapeutic stra-tegy for Alport glomerular disease.


Subject(s)
Glomerular Basement Membrane/pathology , Metalloendopeptidases/metabolism , Nephritis, Hereditary/enzymology , Nephritis, Hereditary/pathology , Animals , Blotting, Northern , Blotting, Western , Enzyme Inhibitors/pharmacology , Glomerular Basement Membrane/drug effects , Humans , Immunohistochemistry , In Situ Hybridization , Matrix Metalloproteinase 12 , Metalloendopeptidases/drug effects , Mice , Microscopy, Electron, Transmission , Receptors, CCR2 , Receptors, Chemokine/drug effects , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
Am J Pathol ; 166(5): 1465-74, 2005 May.
Article in English | MEDLINE | ID: mdl-15855646

ABSTRACT

Alport syndrome results from mutations in genes encoding collagen alpha3(IV), alpha4(IV), or alpha5(IV) and is characterized by progressive glomerular disease associated with a high-frequency sensorineural hearing loss. Earlier studies of a gene knockout mouse model for Alport syndrome noted thickening of strial capillary basement membranes in the cochlea, suggesting that the stria vascularis is the primary site of cochlear pathogenesis. Here we combine a novel cochlear microdissection technique with molecular analyses to illustrate significant quantitative alterations in strial expression of mRNAs encoding matrix metalloproteinases-2, -9, -12, and -14. Gelatin zymography of extracts from the stria vascularis confirmed these findings. Treatment of Alport mice with a small molecule inhibitor of these matrix metalloproteinases exacerbated strial capillary basement membrane thickening, demonstrating that alterations in basement membrane metabolism result in matrix accumulation in the strial capillary basement membranes. This is the first demonstration of true quantitative analysis of specific mRNAs for matrix metalloproteinases in a cochlear microcompartment. Further, these data suggest that the altered basement membrane composition in Alport stria influences the expression of genes involved in basement membrane metabolism.


Subject(s)
Matrix Metalloproteinases/metabolism , Nephritis, Hereditary/enzymology , Stria Vascularis/enzymology , Animals , Basement Membrane/pathology , Capillaries/drug effects , Capillaries/pathology , Computer Systems , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Mice , Mice, Knockout , Nephritis, Hereditary/pathology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stria Vascularis/drug effects , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...