Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(5): e2217327120, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36693102

RESUMO

Gould syndrome is a rare multisystem disorder resulting from autosomal dominant mutations in the collagen-encoding genes COL4A1 and COL4A2. Human patients and Col4a1 mutant mice display brain pathology that typifies cerebral small vessel diseases (cSVDs), including white matter hyperintensities, dilated perivascular spaces, lacunar infarcts, microbleeds, and spontaneous intracerebral hemorrhage. The underlying pathogenic mechanisms are unknown. Using the Col4a1+/G394V mouse model, we found that vasoconstriction in response to internal pressure-the vascular myogenic response-is blunted in cerebral arteries from middle-aged (12 mo old) but not young adult (3 mo old) animals, revealing age-dependent cerebral vascular dysfunction. The defect in the myogenic response was associated with a significant decrease in depolarizing cation currents conducted by TRPM4 (transient receptor potential melastatin 4) channels in native cerebral artery smooth muscle cells (SMCs) isolated from mutant mice. The minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is necessary for TRPM4 activity. Dialyzing SMCs with PIP2 and selective blockade of phosphoinositide 3-kinase (PI3K), an enzyme that converts PIP2 to phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3), restored TRPM4 currents. Acute inhibition of PI3K activity and blockade of transforming growth factor-beta (TGF-ß) receptors also rescued the myogenic response, suggesting that hyperactivity of TGF-ß signaling pathways stimulates PI3K to deplete PIP2 and impair TRPM4 channels. We conclude that age-related cerebral vascular dysfunction in Col4a1+/G394V mice is caused by the loss of depolarizing TRPM4 currents due to PIP2 depletion, revealing an age-dependent mechanism of cSVD.


Assuntos
Músculo Liso Vascular , Canais de Cátion TRPM , Humanos , Camundongos , Animais , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Artérias Cerebrais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(35): e2306479120, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37607233

RESUMO

Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) in brain capillary endothelial cells, leading to the loss of inwardly rectifying K+ (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP2 by converting it to phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD.


Assuntos
Doenças de Pequenos Vasos Cerebrais , Hiperemia , Acoplamento Neurovascular , Animais , Camundongos , Células Endoteliais , Fosfatidilinositol 3-Quinases/genética , Doenças de Pequenos Vasos Cerebrais/genética , Fosfatidilinositol 3-Quinase
3.
PLoS Genet ; 17(3): e1009458, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33755662

RESUMO

Precise regulation of ocular size is a critical determinant of normal visual acuity. Although it is generally accepted that ocular growth relies on a cascade of signaling events transmitted from the retina to the sclera, the factors and mechanism(s) involved are poorly understood. Recent studies have highlighted the importance of the retinal secreted serine protease PRSS56 and transmembrane glycoprotein MFRP, a factor predominantly expressed in the retinal pigment epithelium (RPE), in ocular size determination. Mutations in PRSS56 and MFRP constitute a major cause of nanophthalmos, a condition characterized by severe reduction in ocular axial length/extreme hyperopia. Interestingly, common variants of these genes have been implicated in myopia, a condition associated with ocular elongation. Consistent with these findings, mice with loss of function mutation in PRSS56 or MFRP exhibit a reduction in ocular axial length. However, the molecular network and cellular processes involved in PRSS56- and MFRP-mediated ocular axial growth remain elusive. Here, we show that Adamts19 expression is significantly upregulated in the retina of mice lacking either Prss56 or Mfrp. Importantly, using genetic mouse models, we demonstrate that while ADAMTS19 is not required for ocular growth during normal development, its inactivation exacerbates ocular axial length reduction in Prss56 and Mfrp mutant mice. These results suggest that the upregulation of retinal Adamts19 is part of an adaptive molecular response to counteract impaired ocular growth. Using a complementary genetic approach, we show that loss of PRSS56 or MFRP function prevents excessive ocular axial growth in a mouse model of early-onset myopia caused by a null mutation in Irbp, thus, demonstrating that PRSS56 and MFRP are also required for pathological ocular elongation. Collectively, our findings provide new insights into the molecular network involved in ocular axial growth and support a role for molecular crosstalk between the retina and RPE involved in refractive development.


Assuntos
Proteínas ADAMTS/genética , Proteínas do Olho/genética , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Proteínas de Membrana/genética , Organogênese/genética , Serina Proteases/genética , Proteínas ADAMTS/metabolismo , Animais , Biomarcadores , Olho/embriologia , Olho/crescimento & desenvolvimento , Proteínas do Olho/metabolismo , Imuno-Histoquímica , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteínas de Ligação ao Retinol/genética , Serina Proteases/metabolismo , Transdução de Sinais
4.
Am J Hum Genet ; 104(5): 847-860, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31051113

RESUMO

Collagen type IV alpha 1 and alpha 2 chains form heterotrimers ([α1(IV)]2α2(IV)) that represent a fundamental basement membrane constituent. Dominant COL4A1 and COL4A2 mutations cause a multisystem disorder that is marked by clinical heterogeneity and variable expressivity and that is generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular involvement. Despite the fact that muscle pathology is reported in up to one-third of individuals with COL4A1 and COL4A2 mutations and in animal models with mutations in COL4A1 and COL4A2 orthologs, the pathophysiological mechanisms underlying COL4A1-related myopathy are unknown. In general, mutations are thought to impair [α1(IV)]2α2(IV) secretion. Whether pathogenesis results from intracellular retention, extracellular deficiency, or the presence of mutant proteins in basement membranes represents an important gap in knowledge and a major obstacle for developing targeted interventions. We report that Col4a1 mutant mice develop progressive neuromuscular pathology that models human disease. We demonstrate that independent muscular, neural, and vascular insults contribute to neuromyopathy and that there is mechanistic heterogeneity among tissues. Importantly, we provide evidence of a COL4A1 functional subdomain with disproportionate significance for tissue-specific pathology and demonstrate that a potential therapeutic strategy aimed at promoting [α1(IV)]2α2(IV) secretion can ameliorate or exacerbate myopathy in a mutation-dependent manner. These data have important translational implications for prediction of clinical outcomes based on genotype, development of mechanism-based interventions, and genetic stratification for clinical trials. Collectively, our data underscore the importance of the [α1(IV)]2α2(IV) network as a multifunctional signaling platform and show that allelic and tissue-specific mechanistic heterogeneities contribute to the variable expressivity of COL4A1 and COL4A2 mutations.


Assuntos
Colágeno Tipo IV/genética , Doenças Musculares/etiologia , Mutação , Doenças Neuromusculares/etiologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Musculares/patologia , Doenças Neuromusculares/patologia , Especificidade de Órgãos , Fenótipo
5.
PLoS Genet ; 14(3): e1007244, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29529029

RESUMO

A mismatch between optical power and ocular axial length results in refractive errors. Uncorrected refractive errors constitute the most common cause of vision loss and second leading cause of blindness worldwide. Although the retina is known to play a critical role in regulating ocular growth and refractive development, the precise factors and mechanisms involved are poorly defined. We have previously identified a role for the secreted serine protease PRSS56 in ocular size determination and PRSS56 variants have been implicated in the etiology of both hyperopia and myopia, highlighting its importance in refractive development. Here, we use a combination of genetic mouse models to demonstrate that Prss56 mutations leading to reduced ocular size and hyperopia act via a loss of function mechanism. Using a conditional gene targeting strategy, we show that PRSS56 derived from Müller glia contributes to ocular growth, implicating a new retinal cell type in ocular size determination. Importantly, we demonstrate that persistent activity of PRSS56 is required during distinct developmental stages spanning the pre- and post-eye opening periods to ensure optimal ocular growth. Thus, our mouse data provide evidence for the existence of a molecule contributing to both the prenatal and postnatal stages of human ocular growth. Finally, we demonstrate that genetic inactivation of Prss56 rescues axial elongation in a mouse model of myopia caused by a null mutation in Egr1. Overall, our findings identify PRSS56 as a potential therapeutic target for modulating ocular growth aimed at preventing or slowing down myopia, which is reaching epidemic proportions.


Assuntos
Olho/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Erros de Refração/genética , Serina Proteases/metabolismo , Animais , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Olho/citologia , Olho/embriologia , Feminino , Humanos , Hiperopia/genética , Masculino , Camundongos Mutantes , Camundongos Transgênicos , Miopia/genética , Miopia/patologia , Neuroglia/metabolismo , Refração Ocular/genética , Refração Ocular/fisiologia , Erros de Refração/prevenção & controle , Serina Proteases/genética
6.
Hum Mol Genet ; 23(7): 1709-22, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24203695

RESUMO

Collagen type IV alpha 1 and 2 (COL4A1 and COL4A2) are present in nearly all basement membranes. COL4A1 and COL4A2 mutations are pleiotropic, affecting multiple organ systems to differing degrees, and both genetic-context and environmental factors influence this variable expressivity. Here, we report important phenotypic and molecular differences in an allelic series of Col4a1 and Col4a2 mutant mice that are on a uniform genetic background. We evaluated three organs commonly affected by COL4A1 and COL4A2 mutations and discovered allelic heterogeneity in the penetrance and severity of ocular dysgenesis, myopathy and brain malformations. Similarly, we show allelic heterogeneity in COL4A1 and COL4A2 biosynthesis. While most mutations that we examined caused increased intracellular and decreased extracellular COL4A1 and COL4A2, we identified three mutations with distinct biosynthetic signatures. Reduced temperature or presence of 4-phenylbutyrate ameliorated biosynthetic defects in primary cell lines derived from mutant mice. Together, our data demonstrate the effects and clinical implications of allelic heterogeneity in Col4a1- and Col4a2-related diseases. Understanding allelic differences will be valuable for increasing prognostic accuracy and for the development of therapeutic interventions that consider the nature of the molecular cause in patients with COL4A1 and COL4A2 mutations.


Assuntos
Encéfalo/anormalidades , Colágeno Tipo IV/genética , Anormalidades do Olho/genética , Doenças Musculares/genética , Doenças do Nervo Óptico/congênito , Alelos , Animais , Antineoplásicos/farmacologia , Membrana Basal/metabolismo , Células Cultivadas , Estresse do Retículo Endoplasmático/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Doenças do Nervo Óptico/genética , Penetrância , Fenilbutiratos/farmacologia , Dobramento de Proteína , Temperatura
7.
Am J Hum Genet ; 90(1): 91-101, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22209247

RESUMO

Collagen, type IV, alpha 1 (COL4A1) and alpha 2 (COL4A2) form heterotrimers and are abundant components of basement membranes, including those of the cerebral vasculature. COL4A1 mutations are an increasingly recognized cause of multisystem disorders, including highly penetrant cerebrovascular disease and intracerebral hemorrhage (ICH). Because COL4A1 and COL4A2 are structurally and functionally associated, we hypothesized that variants in COL4A2 would also cause ICH. We sequence COL4A2 in 96 patients with ICH and identify three rare, nonsynonymous coding variants in four patients that are not present in a cohort of 144 ICH-free individuals. All three variants change evolutionarily conserved amino acids. Using a cellular assay, we show that these putative mutations cause intracellular accumulation of COL4A1 and COL4A2 at the expense of their secretion, which supports their pathogenecity. Furthermore, we show that Col4a2 mutant mice also have completely penetrant ICH and that mutations in mouse and human lead to retention of COL4A1 and COL4A2 within the endoplasmic reticulum (ER). Importantly, two of the three putative mutations found in patients trigger ER stress and activate the unfolded protein response. The identification of putative COL4A2 mutations that might contribute to ICH in human patients provides insight into the pathogenic mechanisms of this disease. Our data suggest that COL4A2 mutations impair COL4A1 and COL4A2 secretion and can also result in cytotoxicity. Finally, our findings suggest that, collectively, mutations in COL4A1 and COL4A2 contribute to sporadic cases of ICH.


Assuntos
Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Hemorragias Intracranianas/genética , Mutação , Acidente Vascular Cerebral/genética , Adulto , Idoso , Sequência de Aminoácidos , Animais , Sequência de Bases , Estresse do Retículo Endoplasmático/genética , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Resposta a Proteínas não Dobradas
8.
Curr Top Membr ; 76: 61-116, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26610912

RESUMO

Basement membranes are highly specialized extracellular matrices. Once considered inert scaffolds, basement membranes are now viewed as dynamic and versatile environments that modulate cellular behaviors to regulate tissue development, function, and repair. Increasing evidence suggests that, in addition to providing structural support to neighboring cells, basement membranes serve as reservoirs of growth factors that direct and fine-tune cellular functions. Type IV collagens are a major component of all basement membranes. They evolved along with the earliest multicellular organisms and have been integrated into diverse fundamental biological processes as time and evolution shaped the animal kingdom. The roles of basement membranes in humans are as complex and diverse as their distributions and molecular composition. As a result, basement membrane defects result in multisystem disorders with ambiguous and overlapping boundaries that likely reflect the simultaneous interplay and integration of multiple cellular pathways and processes. Consequently, there will be no single treatment for basement membrane disorders, and therapies are likely to be as varied as the phenotypes. Understanding tissue-specific pathology and the underlying molecular mechanism is the present challenge; personalized medicine will rely upon understanding how a given mutation impacts diverse cellular functions.


Assuntos
Membrana Basal/patologia , Biologia Celular , Colágeno Tipo IV , Doença , Animais , Membrana Basal/metabolismo , Colágeno Tipo IV/química , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Genômica , Humanos
9.
Hum Mol Genet ; 21(R1): R97-110, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22914737

RESUMO

Heterotrimers composed of collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) constitute one of the most abundant components of nearly all basement membranes. Accordingly, mutations in COL4A1 or COL4A2 are pleiotropic and contribute to a broad spectrum of disorders, including myopathy, glaucoma and hemorrhagic stroke. Here, we summarize the contributions of COL4A1 and COL4A2 mutations in human disease, integrate knowledge gained from model organisms and evaluate the implications for pathogenic mechanisms and therapeutic approaches.


Assuntos
Colágeno Tipo IV/genética , Glaucoma/genética , Doenças Musculares/genética , Acidente Vascular Cerebral/genética , Animais , Membrana Basal/metabolismo , Transporte Biológico , Colágeno Tipo IV/química , Colágeno Tipo IV/metabolismo , Predisposição Genética para Doença , Humanos , Camundongos , Terapia de Alvo Molecular , Mutação , Fenótipo , Multimerização Proteica
10.
PLoS Genet ; 7(5): e1002062, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21625620

RESUMO

Muscle-eye-brain disease (MEB) and Walker Warburg Syndrome (WWS) belong to a spectrum of autosomal recessive diseases characterized by ocular dysgenesis, neuronal migration defects, and congenital muscular dystrophy. Until now, the pathophysiology of MEB/WWS has been attributed to alteration in dystroglycan post-translational modification. Here, we provide evidence that mutations in a gene coding for a major basement membrane protein, collagen IV alpha 1 (COL4A1), are a novel cause of MEB/WWS. Using a combination of histological, molecular, and biochemical approaches, we show that heterozygous Col4a1 mutant mice have ocular dysgenesis, neuronal localization defects, and myopathy characteristic of MEB/WWS. Importantly, we identified putative heterozygous mutations in COL4A1 in two MEB/WWS patients. Both mutations occur within conserved amino acids of the triple-helix-forming domain of the protein, and at least one mutation interferes with secretion of the mutant proteins, resulting instead in intracellular accumulation. Expression and posttranslational modification of dystroglycan is unaltered in Col4a1 mutant mice indicating that COL4A1 mutations represent a distinct pathogenic mechanism underlying MEB/WWS. These findings implicate a novel gene and a novel mechanism in the etiology of MEB/WWS and expand the clinical spectrum of COL4A1-associated disorders.


Assuntos
Colágeno Tipo IV/genética , Olho/patologia , Doenças Musculares/genética , Mutação , Neurônios/patologia , Síndrome de Walker-Warburg/genética , Animais , Apoptose , Sequência de Bases , Colágeno Tipo IV/metabolismo , Humanos , Camundongos , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Alinhamento de Sequência , Síndrome de Walker-Warburg/metabolismo , Síndrome de Walker-Warburg/patologia
11.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38717426

RESUMO

Purpose: Mutations in the genes encoding type IV collagen alpha 1 (COL4A1) and alpha 2 (COL4A2) cause a multisystem disorder that includes ocular anterior segment dysgenesis (ASD) and glaucoma. We previously showed that transforming growth factor beta (TGFß) signaling was elevated in developing anterior segments from Col4a1 mutant mice and that reducing TGFß signaling ameliorated ASD, supporting a role for the TGFß pathway in disease pathogenesis. Here, we tested whether altered TGFß signaling also contributes to glaucoma-related phenotypes in Col4a1 mutant mice. Methods: To test the role of TGFß signaling in glaucoma-relevant phenotypes, we genetically reduced TGFß signaling using mice with mutated Tgfbr2, which encodes the common receptor for all TGFß ligands in Col4a1+/G1344D mice. We performed slit-lamp biomicroscopy and optical coherence tomography for qualitative and quantitative analyses of anterior and posterior ocular segments, histological analyses of ocular tissues and optic nerves, and intraocular pressure assessments using rebound tonometry. Results: Col4a1+/G1344D mice showed defects of the ocular drainage structures, including iridocorneal adhesions, and phenotypes consistent with glaucomatous neurodegeneration, including thinning of the nerve fiber layer, retinal ganglion cell loss, optic nerve head excavation, and optic nerve degeneration. We found that reducing TGFß receptor 2 (TGFBR2) was protective for ASD, ameliorated ocular drainage structure defects, and protected against glaucomatous neurodegeneration in Col4a1+/G1344D mice. Conclusions: Our results suggest that elevated TGFß signaling contributes to glaucomatous neurodegeneration in Col4a1 mutant mice.


Assuntos
Colágeno Tipo IV , Glaucoma , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Camundongos , Segmento Anterior do Olho/metabolismo , Segmento Anterior do Olho/patologia , Colágeno Tipo IV/metabolismo , Colágeno Tipo IV/genética , Modelos Animais de Doenças , Glaucoma/metabolismo , Glaucoma/genética , Glaucoma/patologia , Pressão Intraocular/fisiologia , Camundongos Endogâmicos C57BL , Mutação , Nervo Óptico/patologia , Nervo Óptico/metabolismo , Doenças do Nervo Óptico/metabolismo , Doenças do Nervo Óptico/genética , Fenótipo , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/fisiologia , Microscopia com Lâmpada de Fenda , Tomografia de Coerência Óptica , Tonometria Ocular , Fator de Crescimento Transformador beta/metabolismo
12.
Ann Neurol ; 71(4): 470-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22522439

RESUMO

OBJECTIVE: Mutations in the type IV collagen alpha 1 gene (COL4A1) cause dominantly inherited cerebrovascular disease. We seek to determine the extent to which COL4A1 mutations contribute to sporadic, nonfamilial, intracerebral hemorrhages (ICHs). METHODS: We sequenced COL4A1 in 96 patients with sporadic ICH. The presence of putative mutations was tested in 145 ICH-free controls. The effects of rare coding variants on COL4A1 biosynthesis were compared to previously validated mutations that cause porencephaly, small vessel disease, and hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC) syndrome. RESULTS: We identified 2 rare nonsynonymous variants in ICH patients that were not detected in controls, 2 rare nonsynonymous variants in controls that were not detected in patients, and 2 common nonsynonymous variants that were detected in patients and controls. No variant found in controls affected COL4A1 biosynthesis. Both variants (COL4A1(P352L) and COL4A1(R538G)) found only in patients changed conserved amino acids and impaired COL4A1 secretion much like mutations that cause familial cerebrovascular disease. INTERPRETATION: This is the first assessment of the broader role for COL4A1 mutations in the etiology of ICH beyond a contribution to rare and severe familial cases and the first functional evaluation of the biosynthetic consequences of an allelic series of COL4A1 mutations that cause cerebrovascular disease. We identified 2 putative mutations in 96 patients with sporadic ICH and showed that these and other previously validated mutations inhibit secretion of COL4A1. Our data support the hypothesis that increased intracellular accumulation of COL4A1, decreased extracellular COL4A1, or both, contribute to sporadic cerebrovascular disease and ICH.


Assuntos
Hemorragia Cerebral/genética , Colágeno Tipo IV/genética , Predisposição Genética para Doença/genética , Mutação , Idoso , Sequência de Aminoácidos , Western Blotting , Transtornos Cerebrovasculares/genética , Análise Mutacional de DNA , Feminino , Humanos , Masculino , Dados de Sequência Molecular
13.
Matrix Biol ; 115: 48-70, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36435425

RESUMO

Cerebral small vessel disease (CSVD) is a leading cause of stroke and vascular cognitive impairment and dementia. Studying monogenic CSVD can reveal pathways that are dysregulated in common sporadic forms of the disease and may represent therapeutic targets. Mutations in collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause highly penetrant CSVD as part of a multisystem disorder referred to as Gould syndrome. COL4A1 and COL4A2 form heterotrimers [a1α1α2(IV)] that are fundamental constituents of basement membranes. However, their functions are poorly understood and the mechanism(s) by which COL4A1 and COL4A2 mutations cause CSVD are unknown. We used histological, molecular, genetic, pharmacological, and in vivo imaging approaches to characterize central nervous system (CNS) vascular pathologies in Col4a1 mutant mouse models of monogenic CSVD to provide insight into underlying pathogenic mechanisms. We describe developmental CNS angiogenesis abnormalities characterized by impaired retinal vascular outgrowth and patterning, increased numbers of mural cells with abnormal morphologies, altered contractile protein expression in vascular smooth muscle cells (VSMCs) and age-related loss of arteriolar VSMCs in Col4a1 mutant mice. Importantly, we identified elevated TGFß signaling as a pathogenic consequence of Col4a1 mutations and show that genetically suppressing TGFß signaling ameliorated CNS vascular pathologies, including partial rescue of retinal vascular patterning defects, prevention of VSMC loss, and significant reduction of intracerebral hemorrhages in Col4a1 mutant mice aged up to 8 months. This study identifies a novel biological role for collagen α1α1α2(IV) as a regulator of TGFß signaling and demonstrates that elevated TGFß signaling contributes to CNS vascular pathologies caused by Col4a1 mutations. Our findings suggest that pharmacologically suppressing TGFß signaling could reduce the severity of CSVD, and potentially other manifestations associated with Gould syndrome and have important translational implications that could extend to idiopathic forms of CSVD.


Assuntos
Doenças de Pequenos Vasos Cerebrais , Colágeno Tipo IV , Animais , Camundongos , Membrana Basal/metabolismo , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Doenças de Pequenos Vasos Cerebrais/genética , Doenças de Pequenos Vasos Cerebrais/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Mutação , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Modelos Animais de Doenças
14.
bioRxiv ; 2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36945616

RESUMO

Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP 2 ) in brain capillary endothelial cells, leading to the loss of inwardly rectifier K + (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP 2 by converting it to phosphatidylinositol (3,4,5)-trisphosphate (PIP 3 ), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD. One-sentence summary: PI3K inhibition rescues neurovascular coupling defects in cerebral small vessel disease.

15.
Sci Signal ; 16(811): eadi3966, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37963192

RESUMO

Humans and mice with mutations in COL4A1 and COL4A2 manifest hallmarks of cerebral small vessel disease (cSVD). Mice with a missense mutation in Col4a1 at amino acid 1344 (Col4a1+/G1344D) exhibit age-dependent intracerebral hemorrhages (ICHs) and brain lesions. Here, we report that this pathology was associated with the loss of myogenic vasoconstriction, an intrinsic vascular response essential for the autoregulation of cerebral blood flow. Electrophysiological analyses showed that the loss of myogenic constriction resulted from blunted pressure-induced smooth muscle cell (SMC) membrane depolarization. Furthermore, we found that dysregulation of membrane potential was associated with impaired Ca2+-dependent activation of large-conductance Ca2+-activated K+ (BK) and transient receptor potential melastatin 4 (TRPM4) cation channels linked to disruptions in sarcoplasmic reticulum (SR) Ca2+ signaling. Col4a1 mutations impair protein folding, which can cause SR stress. Treating Col4a1+/G1344D mice with 4-phenylbutyrate, a compound that promotes the trafficking of misfolded proteins and alleviates SR stress, restored SR Ca2+ signaling, maintained BK and TRPM4 channel activity, prevented loss of myogenic tone, and reduced ICHs. We conclude that alterations in SR Ca2+ handling that impair ion channel activity result in dysregulation of SMC membrane potential and loss of myogenic tone and contribute to age-related cSVD in Col4a1+/G1344D mice.


Assuntos
Transdução de Sinais , Canais de Cátion TRPM , Camundongos , Animais , Humanos , Transporte de Íons , Vasoconstrição/fisiologia , Canais de Cátion TRPM/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo
16.
J Med Genet ; 48(6): 375-82, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21507892

RESUMO

BACKGROUND: Manitoba-oculo-tricho-anal (MOTA) syndrome is a rare condition defined by eyelid colobomas, cryptophthalmos and anophthalmia/microphthalmia, an aberrant hairline, a bifid or broad nasal tip, and gastrointestinal anomalies such as omphalocele and anal stenosis. Autosomal recessive inheritance had been assumed because of consanguinity in the Oji-Cre population of Manitoba and reports of affected siblings, but no locus or cytogenetic aberration had previously been described. METHODS AND RESULTS: This study shows that MOTA syndrome is caused by mutations in FREM1, a gene previously mutated in bifid nose, renal agenesis, and anorectal malformations (BNAR) syndrome. MOTA syndrome and BNAR syndrome can therefore be considered as part of a phenotypic spectrum that is similar to, but distinct from and less severe than, Fraser syndrome. Re-examination of Frem1(bat/bat) mutant mice found new evidence that Frem1 is involved in anal and craniofacial development, with anal prolapse, eyelid colobomas, telecanthus, a shortened snout and reduced philtral height present in the mutant mice, similar to the human phenotype in MOTA syndrome. CONCLUSIONS: The milder phenotypes associated with FREM1 deficiency in humans (MOTA syndrome and BNAR syndrome) compared to that resulting from FRAS1 and FREM2 loss of function (Fraser syndrome) are also consistent with the less severe phenotypes resulting from Frem1 loss of function in mice. Together, Fraser, BNAR and MOTA syndromes constitute a clinically overlapping group of FRAS-FREM complex diseases.


Assuntos
Anormalidades Múltiplas/genética , Anus Imperfurado/genética , Coloboma/genética , Proteínas da Matriz Extracelular/genética , Síndrome de Fraser/genética , Hipertelorismo/genética , Receptores de Interleucina/genética , Anormalidades Múltiplas/patologia , Adolescente , Adulto , Canal Anal/anormalidades , Canal Anal/patologia , Animais , Malformações Anorretais , Anus Imperfurado/patologia , Sequência de Bases , Criança , Pré-Escolar , Coloboma/patologia , Pálpebras/anormalidades , Feminino , Síndrome de Fraser/patologia , Dosagem de Genes , Hérnia Umbilical/genética , Hérnia Umbilical/patologia , Humanos , Hipertelorismo/patologia , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Nariz/anormalidades , Doenças Nasais/genética , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome
17.
Matrix Biol ; 110: 151-173, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35525525

RESUMO

Ocular anterior segment dysgenesis (ASD) refers to a collection of developmental disorders affecting the anterior structures of the eye. Although a number of genes have been implicated in the etiology of ASD, the underlying pathogenetic mechanisms remain unclear. Mutations in genes encoding collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause Gould syndrome, a multi-system disorder that often includes ocular manifestations such as ASD and glaucoma. COL4A1 and COL4A2 are abundant basement membrane proteins that provide structural support to tissues and modulate signaling through interactions with other extracellular matrix proteins, growth factors, and cell surface receptors. In this study, we used a combination of histological, molecular, genetic and pharmacological approaches to demonstrate that altered TGFß signaling contributes to ASD in mouse models of Gould syndrome. We show that TGFß signaling was elevated in anterior segments from Col4a1 mutant mice and that genetically reducing TGFß signaling partially prevented ASD. Notably, we identified distinct roles for TGFß1 and TGFß2 in ocular defects observed in Col4a1 mutant mice. Importantly, we show that pharmacologically promoting type IV collagen secretion or reducing TGFß signaling ameliorated ocular pathology in Col4a1 mutant mice. Overall, our findings demonstrate that altered TGFß signaling contributes to COL4A1-related ocular dysgenesis and implicate this pathway as a potential therapeutic target for the treatment of Gould syndrome.


Assuntos
Colágeno Tipo IV/metabolismo , Anormalidades do Olho , Animais , Membrana Basal/metabolismo , Colágeno Tipo IV/genética , Olho/metabolismo , Anormalidades do Olho/metabolismo , Camundongos , Mutação , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
18.
Dis Model Mech ; 13(5)2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32152063

RESUMO

Glaucoma is a leading cause of blindness, affecting up to 70 million people worldwide. High intraocular pressure (IOP) is a major risk factor for glaucoma. It is well established that inefficient aqueous humor (AqH) outflow resulting from structural or functional alterations in ocular drainage tissues causes high IOP, but the genes and pathways involved are poorly understood. We previously demonstrated that mutations in the gene encoding the serine protease PRSS56 induces ocular angle closure and high IOP in mice and identified reduced ocular axial length as a potential contributing factor. Here, we show that Prss56-/- mice also exhibit an abnormal iridocorneal angle configuration characterized by a posterior shift of ocular drainage structures relative to the ciliary body and iris. Notably, we show that retina-derived PRSS56 is required between postnatal days 13 and 18 for proper iridocorneal configuration and that abnormal positioning of the ocular drainage tissues is not dependent on ocular size reduction in Prss56-/- mice. Furthermore, we demonstrate that the genetic context modulates the severity of IOP elevation in Prss56 mutant mice and describe a progressive degeneration of ocular drainage tissues that likely contributes to the exacerbation of the high IOP phenotype observed on the C3H/HeJ genetic background. Finally, we identify five rare PRSS56 variants associated with human primary congenital glaucoma, a condition characterized by abnormal development of the ocular drainage structures. Collectively, our findings point to a role for PRSS56 in the development and maintenance of ocular drainage tissues and IOP homeostasis, and provide new insights into glaucoma pathogenesis.


Assuntos
Suscetibilidade a Doenças , Olho/patologia , Olho/fisiopatologia , Pressão Intraocular , Serina Proteases/deficiência , Sequência de Aminoácidos , Animais , Córnea/patologia , Feminino , Glaucoma/genética , Glaucoma/patologia , Iris/patologia , Masculino , Camundongos Knockout , Camundongos Mutantes , Tamanho do Órgão , Serina Proteases/química , Serina Proteases/genética , Serina Proteases/metabolismo
19.
Eur J Neurosci ; 30(7): 1318-28, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19788579

RESUMO

During the pubertal period the mesocortical dopamine (DA) system undergoes substantial reorganization of neuronal connectivity and functional refinement. Netrins are guidance cues involved in the organization of neuronal circuitry. We have previously shown that adult mice that develop with reduced levels of the netrin-1 receptor [deleted in colorectal cancer (DCC)] display selective reorganization of mesocortical DA circuitry, show enhanced mesocortical DA function and exhibit a behavioural phenotype opposite to that observed in animal models of schizophrenia. Here we assess whether the dcc behavioural and DA phenotypes are present prior to the maturation of the mesocortical DA system by comparing dcc-heterozygous and wild-type mice at the post-weaning and peri-pubertal periods on various indices of DA function. At both the post-weaning and peri-pubertal ages, but unlike in adulthood, dcc-heterozygous and wild-type mice show no differences in the number of midbrain DA neurones or in tyrosine hydroxylase protein levels in the medial prefrontal cortex. Furthermore, the elevated baseline concentration of mesocortical DA and DA metabolites observed in adult dcc-heterozygous mice is not present in either post-weanling or peri-pubertal mice. Interestingly, post-weanling, but not peri-pubertal, dcc-heterozygous mice show greater baseline concentrations of DA metabolites in the nucleus accumbens, opposite to what was observed in adulthood. Finally, neither post-weanling nor peri-pubertal dcc-heterozygous mice demonstrate the blunted amphetamine-induced locomotor response observed in adulthood. Thus, these findings show that the 'protective' dcc phenotype has a post-pubertal emergence and indicate that DCC may play a role in the normal maturation of the mesocorticolimbic DA system.


Assuntos
Envelhecimento/fisiologia , Comportamento Animal/fisiologia , Encéfalo/fisiologia , Dopamina/metabolismo , Receptores de Superfície Celular/metabolismo , Maturidade Sexual/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Anfetamina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Estimulantes do Sistema Nervoso Central/farmacologia , Receptor DCC , Feminino , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/crescimento & desenvolvimento , Mesencéfalo/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/crescimento & desenvolvimento , Núcleo Accumbens/fisiologia , Fenótipo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/fisiologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Tirosina 3-Mono-Oxigenase/metabolismo
20.
Synapse ; 63(1): 54-60, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18932228

RESUMO

Neonatal ventral hippocampal (nVH) lesions in rats, which model certain features of schizophrenia, alter dopamine (DA)-mediated behaviors in adulthood. The precise mechanisms underlying these effects remain elusive; however, neuronal reorganization within the medial prefrontal cortex (mPFC) has been suggested. Netrins are developmental cues that organize brain wiring, including the mesocorticolimbic DA circuitry. We showed recently that the netrin-1 receptors DCC and UNC5H are highly expressed by DA neurons and that variation in DCC levels during development lead to profound changes in mesocorticolimbic DA function and behavior in adulthood. We hypothesized that changes in netrin-1 receptor function could be one of the mechanisms producing enduring changes in DA function in nVH-lesioned animals. To begin to explore this idea, we examined the effects of nVH lesions on DCC and UNC5H expression in brain regions receiving robust DA innervation; the mPFC, striatum, and nucleus accumbens (NAcc) at three developmental time points; 3 days after lesion, before puberty and during early adulthood. Expression was also examined in the cerebellar simple lobule; a brain region deprived of DA innervation. Neonatal VH lesions produced dynamic changes in DCC expression in the mPFC and NAcc. The direction and magnitude of these changes depended on the developmental age and brain region examined and were specific to regions receiving DA innervation. Although further studies are required to understand the functional significance of these changes, these results raise the interesting possibility that nVH lesions, and perinatal insults in general, may exert their neuronal reorganizational effects by modulating netrin-1 function.


Assuntos
Dopamina/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipocampo/patologia , Terminações Pré-Sinápticas/fisiologia , Receptores de Superfície Celular/biossíntese , Animais , Animais Recém-Nascidos , Feminino , Hipocampo/fisiologia , Masculino , Receptores de Netrina , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA