Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Matrix Biol ; 133: 1-13, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39097038

RESUMEN

Skeletal defects are hallmark features of many extracellular matrix (ECM) and collagen-related disorders. However, a biological function in bone has never been defined for the highly evolutionarily conserved type IV collagen. Collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) form α1α1α2 (IV) heterotrimers that represent a fundamental basement membrane constituent present in every organ of the body, including the skeleton. COL4A1 and COL4A2 mutations cause Gould syndrome, a variable and clinically heterogenous multisystem disorder generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular manifestations. We have previously identified elevated TGFß signaling as a pathological insult resulting from Col4a1 mutations and demonstrated that reducing TGFß signaling ameliorate ocular and cerebrovascular phenotypes in Col4a1 mutant mouse models of Gould syndrome. In this study, we describe the first characterization of skeletal defects in Col4a1 mutant mice that include a developmental delay in osteogenesis and structural, biomechanical and vascular alterations of mature bones. Using distinct mouse models, we show that allelic heterogeneity influences the presentation of skeletal pathology resulting from Col4a1 mutations. Importantly, we found that TGFß target gene expression is elevated in developing bones from Col4a1 mutant mice and show that genetically reducing TGFß signaling partially ameliorates skeletal manifestations. Collectively, these findings identify a novel and unsuspected role for type IV collagen in bone biology, expand the spectrum of manifestations associated with Gould syndrome to include skeletal abnormalities, and implicate elevated TGFß signaling in skeletal pathogenesis in Col4a1 mutant mice.


Asunto(s)
Colágeno Tipo IV , Modelos Animales de Enfermedad , Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Ratones , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Huesos/metabolismo , Huesos/patología , Mutación , Osteogénesis/genética
2.
Cells Dev ; 179: 203926, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38729574

RESUMEN

The periocular mesenchyme (POM) is a transient migratory embryonic tissue derived from neural crest cells (NCCs) and paraxial mesoderm that gives rise to most of the structures in front of the eye. Morphogenetic defects of these structures can impair aqueous humor outflow, leading to elevated intraocular pressure and glaucoma. Mutations in collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause Gould syndrome - a multisystem disorder often characterized by variable cerebrovascular, ocular, renal, and neuromuscular manifestations. Approximately one-third of individuals with COL4A1 and COL4A2 mutations have ocular anterior segment dysgenesis (ASD), including congenital glaucoma resulting from abnormalities of POM-derived structures. POM differentiation has been a major focus of ASD research, but the underlying cellular mechanisms are still unclear. Moreover, earlier events including NCC migration and survival defects have been implicated in ASD; however, their roles are not as well understood. Vascular defects are among the most common consequences of COL4A1 and COL4A2 mutations and can influence NCC survival and migration. We therefore hypothesized that NCC migration might be impaired by COL4A1 and COL4A2 mutations. In this study, we used 3D confocal microscopy, gross morphology, and quantitative analyses to test NCC migration in Col4a1 mutant mice. We show that homozygous Col4a1 mutant embryos have severe embryonic growth retardation and lethality, and we identified a potential maternal effect on embryo development. Cerebrovascular defects in heterozygous Col4a1 mutant embryos were present as early as E9.0, showing abnormal cerebral vasculature plexus remodeling compared to controls. We detected abnormal NCC migration within the diencephalic stream and the POM in heterozygous Col4a1 mutants whereby mutant NCCs formed smaller diencephalic migratory streams and POMs. In these settings, migratory NCCs within the diencephalic stream and POM localize farther away from the developing vasculature. Our results show for the first time that Col4a1 mutations lead to cranial NCCs migratory defects in the context of early onset defective angiogenesis without affecting cell numbers, possibly impacting the relation between NCCs and the blood vessels during ASD development.


Asunto(s)
Movimiento Celular , Colágeno Tipo IV , Modelos Animales de Enfermedad , Anomalías del Ojo , Mutación , Cresta Neural , Cresta Neural/metabolismo , Cresta Neural/patología , Animales , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Movimiento Celular/genética , Ratones , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Mutación/genética , Segmento Anterior del Ojo/anomalías , Segmento Anterior del Ojo/patología
3.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38717426

RESUMEN

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.


Asunto(s)
Colágeno Tipo IV , Glaucoma , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Ratones , Segmento Anterior del Ojo/metabolismo , Segmento Anterior del Ojo/patología , Colágeno Tipo IV/metabolismo , Colágeno Tipo IV/genética , Modelos Animales de Enfermedad , Glaucoma/metabolismo , Glaucoma/genética , Glaucoma/patología , Presión Intraocular/fisiología , Ratones Endogámicos C57BL , Mutación , Nervio Óptico/patología , Nervio Óptico/metabolismo , Enfermedades del Nervio Óptico/metabolismo , Enfermedades del Nervio Óptico/genética , Fenotipo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Transducción de Señal/fisiología , Microscopía con Lámpara de Hendidura , Tomografía de Coherencia Óptica , Tonometría Ocular , Factor de Crecimiento Transformador beta/metabolismo
4.
Proc Natl Acad Sci U S A ; 121(5): e2311487121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261611

RESUMEN

Roughly one-half of mice with partial defects in two immune tolerance pathways (AireGW/+Lyn-/- mice) spontaneously develop severe damage to their retinas due to T cell reactivity to Aire-regulated interphotoreceptor retinoid-binding protein (IRBP). Single-cell T cell receptor (TCR) sequencing of CD4+ T cells specific for a predominate epitope of IRBP showed a remarkable diversity of autoantigen-specific TCRs with greater clonal expansions in mice with disease. TCR transgenic mice made with an expanded IRBP-specific TCR (P2.U2) of intermediate affinity exhibited strong but incomplete negative selection of thymocytes. This negative selection was absent in IRBP-/- mice and greatly defective in AireGW/+ mice. Most P2.U2+/- mice and all P2.U.2+/-AireGW/+ mice rapidly developed inflammation of the retina and adjacent uvea (uveitis). Aire-dependent IRBP expression in the thymus also promoted Treg differentiation, but the niche for this fate determination was small, suggesting differences in antigen presentation leading to negative selection vs. thymic Treg differentiation and a stronger role for negative selection in preventing autoimmune disease in the retina.


Asunto(s)
Presentación de Antígeno , Receptores de Antígenos de Linfocitos T , Animales , Ratones , Autoantígenos , Modelos Animales de Enfermedad , Ratones Endogámicos , Ratones Transgénicos
5.
Sci Signal ; 16(811): eadi3966, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37963192

RESUMEN

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.


Asunto(s)
Transducción de Señal , Canales Catiónicos TRPM , Ratones , Animales , Humanos , Transporte Iónico , Vasoconstricción/fisiología , Canales Catiónicos TRPM/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo
6.
Int J Mol Sci ; 24(20)2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37894834

RESUMEN

Mutations in the FKBP14 gene encoding the endoplasmic reticulum resident collagen-related proline isomerase FK506 binding protein 22 kDa (FKBP22) result in kyphoscoliotic Ehlers-Danlos Syndrome (EDS), which is characterized by a broad phenotypic outcome. A plausible explanation for this outcome is that FKBP22 participates in the biosynthesis of subsets of collagen types: FKBP22 selectively binds to collagens III, IV, VI, and X, but not to collagens I, II, V, and XI. However, these binding mechanisms have never been explored, and they may underpin EDS subtype heterogeneity. Here, we used collagen Toolkit peptide libraries to investigate binding specificity. We observed that FKBP22 binding was distributed along the collagen helix. Further, it (1) was higher on collagen III than collagen II peptides and it (2) was correlated with a positive peptide charge. These findings begin to elucidate the mechanism by which FKBP22 interacts with collagen.


Asunto(s)
Síndrome de Ehlers-Danlos , Proteínas de Unión a Tacrolimus , Humanos , Proteínas de Unión a Tacrolimus/metabolismo , Colágeno/genética , Isomerasa de Peptidilprolil/genética , Mutación , Síndrome de Ehlers-Danlos/genética
7.
Proc Natl Acad Sci U S A ; 120(35): e2306479120, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37607233

RESUMEN

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.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales , Hiperemia , Acoplamiento Neurovascular , Animales , Ratones , Células Endoteliales , Fosfatidilinositol 3-Quinasas/genética , Enfermedades de los Pequeños Vasos Cerebrales/genética , Fosfatidilinositol 3-Quinasa
8.
Invest Ophthalmol Vis Sci ; 64(4): 30, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37097227

RESUMEN

Purpose: The unfolded protein response (UPR) is triggered when the protein folding capacity of the endoplasmic reticulum (ER) is overwhelmed and misfolded proteins accumulate in the ER, a condition referred to as ER stress. IRE1α is an ER-resident protein that plays major roles in orchestrating the UPR. Several lines of evidence implicate the UPR and its transducers in neurodegenerative diseases, including retinitis pigmentosa (RP), a group of inherited diseases that cause progressive dysfunction and loss of rod and cone photoreceptors. This study evaluated the contribution of IRE1α to photoreceptor development, homeostasis, and degeneration. Methods: We used a conditional gene targeting strategy to selectively inactivate Ire1α in mouse rod photoreceptors. We used a combination of optical coherence tomography (OCT) imaging, histology, and electroretinography (ERG) to assess longitudinally the effect of IRE1α deficiency in retinal development and function. Furthermore, we evaluated the IRE1α-deficient retina responses to tunicamycin-induced ER stress and in the context of RP caused by the rhodopsin mutation RhoP23H. Results: OCT imaging, histology, and ERG analyses did not reveal abnormalities in IRE1α-deficient retinas up to 3 months old. However, by 6 months of age, the Ire1α mutant animals showed reduced outer nuclear layer thickness and deficits in retinal function. Furthermore, conditional inactivation of Ire1α in rod photoreceptors accelerated retinal degeneration caused by the RhoP23H mutation. Conclusions: These data suggest that IRE1α is dispensable for photoreceptor development but important for photoreceptor homeostasis in aging retinas and for protecting against ER stress-mediated photoreceptor degeneration.


Asunto(s)
Degeneración Retiniana , Retinitis Pigmentosa , Animales , Ratones , Envejecimiento , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Degeneración Retiniana/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Respuesta de Proteína Desplegada , Estrés del Retículo Endoplásmico
9.
bioRxiv ; 2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36945616

RESUMEN

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.

10.
Proc Natl Acad Sci U S A ; 120(5): e2217327120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36693102

RESUMEN

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.


Asunto(s)
Músculo Liso Vascular , Canales Catiónicos TRPM , Humanos , Ratones , Animales , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Arterias Cerebrales/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
11.
Matrix Biol ; 115: 48-70, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36435425

RESUMEN

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.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales , Colágeno Tipo IV , Animales , Ratones , Membrana Basal/metabolismo , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Enfermedades de los Pequeños Vasos Cerebrales/genética , Enfermedades de los Pequeños Vasos Cerebrales/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Mutación , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Modelos Animales de Enfermedad
12.
J Biol Chem ; 298(12): 102713, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36403858

RESUMEN

Collagens are the most abundant proteins in the body and among the most biosynthetically complex. A molecular ensemble of over 20 endoplasmic reticulum resident proteins participates in collagen biosynthesis and contributes to heterogeneous post-translational modifications. Pathogenic variants in genes encoding collagens cause connective tissue disorders, including osteogenesis imperfecta, Ehlers-Danlos syndrome, and Gould syndrome (caused by mutations in COL4A1 and COL4A2), and pathogenic variants in genes encoding proteins required for collagen biosynthesis can cause similar but overlapping clinical phenotypes. Notably, pathogenic variants in lysyl hydroxylase 3 (LH3) cause a multisystem connective tissue disorder that exhibits pathophysiological features of collagen-related disorders. LH3 is a multifunctional collagen-modifying enzyme; however, its precise role(s) and substrate specificity during collagen biosynthesis has not been defined. To address this critical gap in knowledge, we generated LH3 KO cells and performed detailed quantitative and molecular analyses of collagen substrates. We found that LH3 deficiency severely impaired secretion of collagen α1α1α2(IV) but not collagens α1α1α2(I) or α1α1α1(III). Amino acid analysis revealed that LH3 is a selective LH for collagen α1α1α2(IV) but a general glucosyltransferase for collagens α1α1α2(IV), α1α1α2(I), and α1α1α1(III). Importantly, we identified rare variants that are predicted to be pathogenic in the gene encoding LH3 in two of 113 fetuses with intracranial hemorrhage-a cardinal feature of Gould syndrome. Collectively, our findings highlight a critical role of LH3 in α1α1α2(IV) biosynthesis and suggest that LH3 pathogenic variants might contribute to Gould syndrome.


Asunto(s)
Colágeno , Enfermedades del Tejido Conjuntivo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa , Humanos , Colágeno/metabolismo , Glicosilación , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Procesamiento Proteico-Postraduccional
13.
Front Psychol ; 13: 647904, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35783760

RESUMEN

Globally, the COVID-19 pandemic has forced medical education toward more "online education" approaches, causing specific implications to arise for medical educators and learners. Considering an unprecedented and highly threatening, constrained, and confusing social and educational environment caused by the COVID-19 pandemic, we decided to shift the traditional focus of the Cognitive Load Theory (CLT) from students to instructors. In this process, we considered recent suggestions to acknowledge the psychological environment in which learning happens. According to this fundamental fact, "Learning and instructional procedures do not occur in a situational vacuum." Following this assertion, we adapted and implemented principles of CLT to reduce the extraneous load for our faculty to facilitate continued scholarly activity and support the overall wellbeing of our faculty during these trying times. The adoption of these principles enabled our team to cultivate attitudes and skills across multiple domains, such as online presentation technologies, implementing and maintaining a "classroom atmosphere" in a virtual environment, encouraging discussion among large online groups of students, facilitating group work, providing virtual office hours, and proactively planning for subsequent sessions.

14.
Matrix Biol ; 110: 151-173, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35525525

RESUMEN

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.


Asunto(s)
Colágeno Tipo IV/metabolismo , Anomalías del Ojo , Animales , Membrana Basal/metabolismo , Colágeno Tipo IV/genética , Ojo/metabolismo , Anomalías del Ojo/metabolismo , Ratones , Mutación , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
15.
Prenat Diagn ; 42(5): 601-610, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150448

RESUMEN

BACKGROUND: Variants of COL4A1/COL4A2 genes have been reported in fetal intracranial hemorrhage (ICH) cases but their prevalence and characteristics have not been established in a large series of fetuses. Fetal neonatal alloimmune thrombocytopenia is a major acquired ICH factor but the prevalence and characteristics of inherited platelet disorder (IPD) gene variants leading to thrombocytopenia are unknown. Herein, we screened COL4A1/COL4A2 and IPD genes in a large series of ICH fetuses. METHODS: A cohort of 194 consecutive ICH fetuses were first screened for COL4A1/COL4A2 variants. We manually curated a list of 64 genes involved in IPD and investigated them in COL4A1/COL4A2 negative fetuses, using exome sequencing data from 101 of these fetuses. RESULT: Pathogenic variants of COL4A1/COL4A2 genes were identified in 36 fetuses (19%). They occurred de novo in 70% of the 32 fetuses for whom parental DNA was available. Pathogenic variants in two megakaryopoiesis genes (MPL and MECOM genes) were identified in two families with recurrent and severe fetal ICH, with variable extraneurological pathological features. CONCLUSION: Our study emphasizes the genetic heterogeneity of fetal ICH and the need to screen both COL4A1/COL4A2 and IPD genes in the etiological investigation of fetal ICH to allow proper genetic counseling.


Asunto(s)
Feto , Hemorragias Intracraneales , Estudios de Cohortes , Colágeno Tipo IV/genética , Feto/patología , Humanos , Recién Nacido , Hemorragias Intracraneales/genética , Mutación
16.
Anat Sci Educ ; 15(4): 685-697, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34048140

RESUMEN

Gross anatomy is a source of anxiety for matriculating medical students due to the large volume of information presented in a truncated timeline, and because it may be their first exposure to human cadavers. This study aimed to assess if video-based resources would affect matriculating medical students' anatomy state anxiety levels. Videos were designed to be short, YouTube-based units that served to provide orientation information about the anatomy course, dissection facilities, and available study resources to dispel anxiety around beginning their anatomy studies. To evaluate the impact of the videos, students in two consecutive matriculating years (2018 and 2019) completed the validated State-Trait Anxiety Inventory and a demographic questionnaire. The 2019 cohort (n = 118) served as the experimental group with access to the videos; while the 2018 cohort (n = 120) without video access served as a historical control. Analyses revealed that the groups were equivalent in terms of trait anxiety (P = 0.854) and anatomy state anxiety even when student video exposure was controlled (P = 0.495). Anatomy state anxiety was only significantly lower in students with prior formal anatomy exposure (P = 0.006). Further inquiry into students' prior anatomy experience identified that individuals with post-secondary dissection experience were significantly less anxious than those without formal anatomical experience (P = 0.023). These results may serve as a cautionary tale to educators; while preference for video-based instructional materials is prevalent in the literature, videos delivered on public social media platforms fail to prepare students for the psychological impact of studying human anatomy.


Asunto(s)
Anatomía , Educación de Pregrado en Medicina , Medios de Comunicación Sociales , Estudiantes de Medicina , Anatomía/educación , Ansiedad , Educación de Pregrado en Medicina/métodos , Humanos , Estudiantes de Medicina/psicología
17.
Geroscience ; 44(1): 25-37, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34606040

RESUMEN

White matter pathologies are critically involved in the etiology of vascular cognitive impairment-dementia (VCID), Alzheimer's disease (AD), and Alzheimer's disease and related diseases (ADRD), and therefore need to be considered a treatable target ( Roseborough A, Hachinski V, Whitehead S. White matter degeneration - a treatable target? Roseborough et al. JAMA Neurol [Internet]. 2020 Apr 27;77(7):793-4, [1] . To help address this often-missed area of research, several workshops have been sponsored by the Leo and Anne Albert Charitable Trust since 2015, resulting in the incorporation of "The Albert Research Institute for White Matter and Cognition" in 2020. The first annual "Institute" meeting was held virtually on March 3-4, 2021. The Institute provides a forum and workspace for communication and support of the advancement of white matter science and research to better understand the evolution and prevention of dementia. It serves as a platform for young investigator development, to introduce new data and debate biology mechanisms and new ideas, and to encourage and support new research collaborations and directions to clarify how white matter changes, with other genetic and health risk factors, contribute to cognitive impairment. Similar to previous Albert Trust-sponsored workshops (Barone et al. in J Transl Med 14:1-14, [2]; Sorond et al. in GeroScience 42:81-96, [3]), established expert investigators were identified and invited to present. Opportunities to attend and present were also extended by invitation to talented research fellows and younger scientists. Also, updates on institute-funded research collaborations were provided and discussed. The summary that follows is a synopsis of topics and discussion covered in the workshop.


Asunto(s)
Demencia Vascular , Leucoencefalopatías , Sustancia Blanca , Academias e Institutos , Cognición , Humanos , Leucoencefalopatías/patología
18.
Epilepsy Behav ; 125: 108365, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34735964

RESUMEN

Recently, patient advocacy groups started using the name Gould syndrome to describe clinical features of COL4A1 and COL4A2 mutations. Gould syndrome is increasingly identified in genetic screening panels, and because it is a rare disease, there is a disproportionate burden on families to understand the disease and chart the course for clinical care. Among the chief concerns for caregivers of children with Gould syndrome are the challenges faced because of epilepsy, including severe manifestations such as infantile spasms. To document the concerns of the patient population, the Gould Syndrome Foundation established the Gould Syndrome Global Registry (GSGR). METHODS: The Gould Syndrome Foundation developed questions for the GSGR with iterative input from patients and caregivers. An institutional review board issued an exemption determination before data collection began. Participants were recruited through social media and clinician referrals. All participants consented electronically, and the data were collected and managed using REDCap electronic data capture tools. De-identified data representing responses received between October 2019 and February 2021 were exported and analyzed with IBM SPSS 27 using descriptive statistics (mean, standard deviation, frequency, range, and percent). RESULTS: Seventy families from twelve countries provided data for the registry, representing 100 affected people (40 adults and 60 children). This analysis represents a subanalysis of the 35 out of 60 children <=18 years of age who reported a history of seizures. Nearly half of these participants were diagnosed with infantile spasms. Participants with epilepsy frequently reported developmental delays (88.6%), stroke (60.0%), cerebral palsy (65.7%), and constipation (57.1%). Ten (28.6%) children use a feeding tube. Despite the fact that more than half of respondents reported stroke, only 34.3% reported ever receiving education on stroke recognition. CONCLUSION: Here we describe the development and deployment of the first global registry for individuals and family members with Gould syndrome, caused by mutations in COL4A1 and COL4A2. It is important for pediatric neurologists to have access to resources to provide families upon diagnosis. Specifically, all families with Gould Syndrome must have access to infantile spasms awareness and stroke education materials. The Gould Syndrome Foundation is planning several improvements to this patient registry which will encourage collaboration and innovation for the benefit of people living with Gould syndrome.


Asunto(s)
Anomalías Congénitas , Epilepsia , Accidente Cerebrovascular , Adulto , Niño , Colágeno Tipo IV , Anomalías Congénitas/genética , Epilepsia/genética , Humanos , Mutación/genética , Sistema de Registros , Síndrome
19.
Dis Model Mech ; 14(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34424299

RESUMEN

Collagen type IV alpha 1 and alpha 2 (COL4A1 and COL4A2) are major components of almost all basement membranes. COL4A1 and COL4A2 mutations cause a multisystem disorder that can affect any organ but typically involves the cerebral vasculature, eyes, kidneys and skeletal muscles. In recent years, patient advocacy and family support groups have united under the name of Gould syndrome. The manifestations of Gould syndrome are highly variable, and animal studies suggest that allelic heterogeneity and genetic context contribute to the clinical variability. We previously characterized a mouse model of Gould syndrome caused by a Col4a1 mutation in which the severities of ocular anterior segment dysgenesis (ASD), myopathy and intracerebral hemorrhage (ICH) were dependent on genetic background. Here, we performed a genetic modifier screen to provide insight into the mechanisms contributing to Gould syndrome pathogenesis and identified a single locus [modifier of Gould syndrome 1 (MoGS1)] on Chromosome 1 that suppressed ASD. A separate screen showed that the same locus ameliorated myopathy. Interestingly, MoGS1 had no effect on ICH, suggesting that this phenotype could be mechanistically distinct. We refined the MoGS1 locus to a 4.3 Mb interval containing 18 protein-coding genes, including Fn1, which encodes the extracellular matrix component fibronectin 1. Molecular analysis showed that the MoGS1 locus increased Fn1 expression, raising the possibility that suppression is achieved through a compensatory extracellular mechanism. Furthermore, we found evidence of increased integrin-linked kinase levels and focal adhesion kinase phosphorylation in Col4a1 mutant mice that is partially restored by the MoGS1 locus, implicating the involvement of integrin signaling. Taken together, our results suggest that tissue-specific mechanistic heterogeneity contributes to the variable expressivity of Gould syndrome and that perturbations in integrin signaling may play a role in ocular and muscular manifestations.


Asunto(s)
Anomalías Múltiples/genética , Colágeno Tipo IV/genética , Fibronectinas/genética , Genes Modificadores , Animales , Hemorragia Cerebral/complicaciones , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Anomalías del Ojo/complicaciones , Anomalías del Ojo/genética , Fibronectinas/metabolismo , Genes Supresores , Sitios Genéticos , Integrinas/metabolismo , Ratones Mutantes , Enfermedades Musculares/genética , Porencefalia/complicaciones , Transducción de Señal , Síndrome
20.
J Steroid Biochem Mol Biol ; 210: 105861, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675951

RESUMEN

BACKGROUND: Evidence is growing for a role of vitamin D in regulating skeletal muscle mass, strength and functional capacity. Given the role the kidneys play in activating total vitamin D, and the high prevalence of vitamin D deficiency in Chronic Kidney Disease (CKD), it is possible that deficiency contributes to the low levels of physical function and muscle mass in these patients. METHODS: This is a secondary cross-sectional analysis of previously published interventional study, with in vitro follow up work. 34 CKD patients at stages G3b-5 (eGFR 25.5 ± 8.3 mL/min/1.73m2; age 61 ± 12 years) were recruited, with a sub-group (n = 20) also donating a muscle biopsy. Vitamin D and associated metabolites were analysed in plasma by liquid chromatography tandem-mass spectroscopy and correlated to a range of physiological tests of muscle size, function, exercise capacity and body composition. The effects of 1α,25(OH)2D3 supplementation on myogenesis and myotube size was investigated in primary skeletal muscle cells from vitamin D deficient donors. RESULTS: In vivo, there was no association between total or active vitamin D and muscle size or strength, but a significant correlation with V̇O2Peak was seen with total vitamin D (25OHD). in vitro, 1α,25(OH)2D3 supplementation reduced IL-6 mRNA expression, but had no effect upon proliferation, differentiation or myotube diameter. CONCLUSIONS: Vitamin D deficiency is not a prominent factor driving the loss of muscle mass in CKD, but may play a role in reduced exercise capacity.


Asunto(s)
Tolerancia al Ejercicio/fisiología , Insuficiencia Renal Crónica/fisiopatología , Deficiencia de Vitamina D/fisiopatología , Anciano , Calcitonina/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estudios Transversales , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Insuficiencia Renal Crónica/complicaciones , Vitamina D/sangre , Vitamina D/metabolismo , Deficiencia de Vitamina D/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...