Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
1.
Cell Death Dis ; 15(9): 681, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39289348

RESUMEN

Liver regeneration is an intricate pathophysiological process that has been a subject of great interest to the scientific community for many years. The capacity of liver regeneration is very critical for patients with liver diseases. Therefore, exploring the mechanisms of liver regeneration and finding good ways to improve it are very meaningful. Mesencephalic astrocyte-derived neurotrophic factor (MANF), a member of newly identified neurotrophic factors (NTFs) family, extensively expresses in the liver and has demonstrated cytoprotective effects during ER stress and inflammation. However, the role of MANF in liver regeneration remains unclear. Here, we used hepatocyte-specific MANF knockout (MANFHep-/-) mice to investigate the role of MANF in liver regeneration after 2/3 partial hepatectomy (PH). Our results showed that MANF expression was up-regulated in a time-dependent manner, and the peak level of mRNA and protein appeared at 24 h and 36 h after 2/3 PH, respectively. Notably, MANF knockout delayed hepatocyte proliferation, and the peak proliferation period was delayed by 24 h. Mechanistically, our in vitro results showed that MANF physically interacts with LRP5 and ß-catenin, two essential components of Wnt/ß-catenin pathway. Specifically, as a cofactor, MANF binds to the extracellular segment of LRP5 to activate Wnt/ß-catenin signaling. On the other hand, MANF interacts with ß-catenin to stabilize cytosolic ß-catenin level and promote its nuclear translocation, which further enhance the Wnt/ß-catenin signaling. We also found that MANF knockout does not affect the c-Met/ß-catenin complex after 2/3 PH. In summary, our study confirms that MANF may serve as a novel hepatocyte factor that is closely linked to the activation of the Wnt/ß-catenin pathway via intracellular and extracellular targets.


Asunto(s)
Proliferación Celular , Hepatectomía , Hepatocitos , Regeneración Hepática , Ratones Noqueados , Factores de Crecimiento Nervioso , Vía de Señalización Wnt , beta Catenina , Regeneración Hepática/fisiología , Animales , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/genética , Hepatocitos/metabolismo , beta Catenina/metabolismo , Ratones , Humanos , Ratones Endogámicos C57BL , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Hígado/metabolismo
2.
Hum Genomics ; 18(1): 87, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39148098

RESUMEN

BACKGROUND: Recent studies suggested that genetic variants associated with monogenic bone disorders were involved in the pathogenesis of atypical femoral fractures (AFF). Here, we aim to identify rare genetic variants by whole exome sequencing in genes involved in monogenic rare skeletal diseases in 12 women with AFF and 4 controls without any fracture. RESULTS: Out of 33 genetic variants identified in women with AFF, eleven (33.3%) were found in genes belonging to the Wnt pathway (LRP5, LRP6, DAAM2, WNT1, and WNT3A). One of them was rated as pathogenic (p.Pro582His in DAAM2), while all others were rated as variants of uncertain significance according to ClinVar and ACMG criteria. CONCLUSIONS: Osteoporosis, rare bone diseases, and AFFs may share the same genes, thus making it even more difficult to identify unique risk factors.


Asunto(s)
Secuenciación del Exoma , Fracturas del Fémur , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Humanos , Femenino , Fracturas del Fémur/genética , Fracturas del Fémur/patología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Persona de Mediana Edad , Anciano , Predisposición Genética a la Enfermedad , Proteína Wnt1/genética , Proteína Wnt3A/genética , Vía de Señalización Wnt/genética , Osteoporosis/genética , Osteoporosis/patología , Enfermedades Óseas/genética , Estudios de Casos y Controles
3.
Arch Toxicol ; 98(10): 3365-3380, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38971901

RESUMEN

Steroid-induced osteonecrosis of the femoral head (SONFH) is a prevalent form of osteonecrosis in young individuals. More efficacious clinical strategies must be used to prevent and treat this condition. One of the mechanisms through which SONFH operates is the disruption of normal differentiation in bone marrow adipocytes and osteoblasts due to prolonged and extensive use of glucocorticoids (GCs). In vitro, it was observed that atorvastatin (ATO) effectively suppressed the impact of dexamethasone (DEX) on bone marrow mesenchymal stem cells (BMSCs), specifically by augmenting their lipogenic differentiation while impeding their osteogenic differentiation. To investigate the underlying mechanisms further, we conducted transcriptome sequencing of BMSCs subjected to different treatments, leading to the identification of Wnt5a as a crucial gene regulated by ATO. The analyses showed that ATO exhibited the ability to enhance the expression of Wnt5a and modulate the MAPK pathway while regulating the Wnt canonical signaling pathway via the WNT5A/LRP5 pathway. Our experimental findings provide further evidence that the combined treatment of ATO and DEX effectively mitigates the effects of DEX, resulting in the upregulation of osteogenic genes (Runx2, Alpl, Tnfrsf11b, Ctnnb1, Col1a) and the downregulation of adipogenic genes (Pparg, Cebpb, Lpl), meanwhile leading to the upregulation of Wnt5a expression. So, this study offers valuable insights into the potential mechanism by which ATO can be utilized in the prevention of SONFH, thereby holding significant implications for the prevention and treatment of SONFH in clinical settings.


Asunto(s)
Atorvastatina , Dexametasona , Necrosis de la Cabeza Femoral , Glucocorticoides , Células Madre Mesenquimatosas , Osteogénesis , Proteína Wnt-5a , Proteína Wnt-5a/metabolismo , Proteína Wnt-5a/genética , Atorvastatina/farmacología , Animales , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/prevención & control , Dexametasona/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Glucocorticoides/farmacología , Osteogénesis/efectos de los fármacos , Masculino , Diferenciación Celular/efectos de los fármacos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Ratas Sprague-Dawley , Células Cultivadas , Adipogénesis/efectos de los fármacos , Ratas
4.
Int J Mol Sci ; 25(12)2024 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-38928468

RESUMEN

Low-density lipoprotein receptor-related protein 5 (LRP5) is a constitutively expressed receptor with observed roles in bone homeostasis, retinal development, and cardiac metabolism. However, the function of LRP5 in the brain remains unexplored. This study investigates LRP5's role in the central nervous system by conducting an extensive analysis using RNA-seq tools and in silico assessments. Two protein-coding Lrp5 transcripts are expressed in mice: full-length Lrp5-201 and a truncated form encoded by Lrp5-202. Wt mice express Lrp5-201 in the liver and brain and do not express the truncated form. Lrp5-/- mice express Lrp5-202 in the liver and brain and do not express Lrp5-201 in the liver. Interestingly, Lrp5-/- mouse brains show full-length Lrp5-201 expression, suggesting that LRP5 has a role in preserving brain function during development. Functional gene enrichment analysis on RNA-seq unveils dysregulated expression of genes associated with neuronal differentiation and synapse formation in the brains of Lrp5-/- mice compared to Wt mice. Furthermore, Gene Set Enrichment Analysis highlights downregulated expression of genes involved in retinol and linoleic acid metabolism in Lrp5-/- mouse brains. Tissue-specific alternative splicing of Lrp5 in Lrp5-/- mice supports that the expression of LRP5 in the brain is needed for the correct synthesis of vitamins and fatty acids, and it is indispensable for correct brain development.


Asunto(s)
Empalme Alternativo , Encéfalo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Animales , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Ratones , Ratones Noqueados , Hígado/metabolismo , Hígado/crecimiento & desarrollo , Ratones Endogámicos C57BL
5.
Bone ; 187: 117172, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38909879

RESUMEN

Gain-of-function mutations in the low-density lipoprotein receptor-related protein 5 (LRP5) can cause high-bone-mass (HBM) phenotype, with 19 identified mutations so far. The A242T mutation in LRP5 has been found in 9 families, making it one of the most prevalent mutations. However, the correlation between the A242T mutation and HBM phenotype remains unverified in animal models. This study aimed to investigate the bone properties in a new transgenic mouse model carrying the LRP5 A241T missense mutation, equivalent to A242T in humans. Heterozygous Lrp5A241T mice were generated using CRISPR/Cas9 genome editing. Body weight increased with age from 4 to 16 weeks, higher in males than females, with no difference between Lrp5A241T mice and wild-type control. Micro-CT showed slightly longer femur and notably elevated trabecular bone mass of the femur and fifth lumbar spine with higher bone mineral density, bone volume fraction, and trabecular thickness in Lrp5A241T mice compared to wild-type mice. Additionally, increased cortical bone thickness and volume of the femur shaft and skull were observed in Lrp5A241T mice. Three-point bending tests of the tibia demonstrated enhanced bone strength properties in Lrp5A241T mice. Histomorphometry confirmed that the A241T mutation increased bone formation without affecting osteoblast number and reduced resorption activities in vivo. In vitro experiments indicated that the LRP5 A241T mutation enhanced osteogenic capacity of osteoblasts with upregulation of the Wnt signaling pathway, with no significant impact on the resorptive activity of osteoclasts. In summary, mice carrying the LRP5 A241T mutation displayed high bone mass and quality due to enhanced bone formation and reduced bone resorption in vivo, potentially mediated by the augmented osteogenic potential of osteoblasts. Continued investigation into the regulatory mechanisms of its bone metabolism and homeostasis may contribute to the advancement of novel therapeutic strategies for bone disorders.


Asunto(s)
Densidad Ósea , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones Transgénicos , Fenotipo , Animales , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Femenino , Masculino , Densidad Ósea/genética , Osteoblastos/metabolismo , Mutación/genética , Ratones , Huesos/metabolismo , Huesos/diagnóstico por imagen , Huesos/patología , Microtomografía por Rayos X , Tamaño de los Órganos , Osteogénesis/genética , Peso Corporal/genética , Fémur/diagnóstico por imagen , Fémur/patología , Fémur/metabolismo , Osteoclastos/metabolismo
6.
Br J Pharmacol ; 181(19): 3556-3575, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38804080

RESUMEN

BACKGROUND AND PURPOSE: Insulin and exenatide are two hypoglycaemic agents that exhibit different osteogenic effects. This study compared the differences between exenatide and insulin in osseointegration in a rat model of Type 2 diabetes (T2D) and explored the mechanisms promoting osteogenesis in this model of T2D. EXPERIMENTAL APPROACH: In vivo, micro-CT was used to detect differences in the peri-implant bone microstructure in vivo. Histology, dual-fluorescent labelling, immunofluorescence and immunohistochemistry were used to detect differences in tissue, cell and protein expression around the implants. In vitro, RT-PCR and western blotting were used to measure the expression of osteogenesis- and Wnt signalling-related genes and proteins in bone marrow mesenchymal stromal cells (BMSCs) from rats with T2D (TBMSCs) after PBS, insulin and exenatide treatment. RT-PCR was used to detect the expression of Wnt bypass cascade reactions under Wnt inactivation. KEY RESULTS: Micro-CT and section staining showed exenatide extensively promoted peri-implant osseointegration. Both in vivo and in vitro experiments showed exenatide substantially increased the expression of osteogenesis-related and activated the LRP5/6/GSK-3ß/ß-catenin-related Wnt pathway. Furthermore, exenatide suppressed expression of Bmpr1a to inhibit lipogenesis and promoted expression of Btrc to suppress inflammation. CONCLUSION AND IMPLICATIONS: Compared to insulin, exenatide significantly improved osteogenesis in T2D rats and TBMSCs. In addition to its dependence on LRP5/6/GSK-3ß/ß-catenin signalling for osteogenic differentiation, exenatide-mediated osteomodulation also involves inhibition of inflammation and adipogenesis by BMPR1A and ß-TrCP, respectively.


Asunto(s)
Diabetes Mellitus Tipo 2 , Exenatida , Insulina , Osteogénesis , beta Catenina , Animales , Masculino , Ratas , beta Catenina/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Exenatida/farmacología , Exenatida/administración & dosificación , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipoglucemiantes/farmacología , Insulina/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Oseointegración/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ratas Sprague-Dawley
7.
Hum Genomics ; 18(1): 53, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802968

RESUMEN

BACKGROUND: The human lineage has undergone a postcranial skeleton gracilization (i.e. lower bone mass and strength relative to body size) compared to other primates and archaic populations such as the Neanderthals. This gracilization has been traditionally explained by differences in the mechanical load that our ancestors exercised. However, there is growing evidence that gracilization could also be genetically influenced. RESULTS: We have analyzed the LRP5 gene, which is known to be associated with high bone mineral density conditions, from an evolutionary and functional point of view. Taking advantage of the published genomes of archaic Homo populations, our results suggest that this gene has a complex evolutionary history both between archaic and living humans and within living human populations. In particular, we identified the presence of different selective pressures in archaics and extant modern humans, as well as evidence of positive selection in the African and South East Asian populations from the 1000 Genomes Project. Furthermore, we observed a very limited evidence of archaic introgression in this gene (only at three haplotypes of East Asian ancestry out of the 1000 Genomes), compatible with a general erasing of the fingerprint of archaic introgression due to functional differences in archaics compared to extant modern humans. In agreement with this hypothesis, we observed private mutations in the archaic genomes that we experimentally validated as putatively increasing bone mineral density. In particular, four of five archaic missense mutations affecting the first ß-propeller of LRP5 displayed enhanced Wnt pathway activation, of which two also displayed reduced negative regulation. CONCLUSIONS: In summary, these data suggest a genetic component contributing to the understanding of skeletal differences between extant modern humans and archaic Homo populations.


Asunto(s)
Evolución Molecular , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Hombre de Neandertal , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Animales , Hombre de Neandertal/genética , Selección Genética/genética , Hominidae/genética , Haplotipos/genética , Densidad Ósea/genética , Genoma Humano/genética
8.
Ophthalmic Surg Lasers Imaging Retina ; 55(8): 462-466, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38752919

RESUMEN

This report describes a unique case of a Coats-like presentation of familial exudative vitreoretinopathy in an 11-year-old girl. The patient was originally referred for evaluation of presumed Coats disease and presented with telangiectatic vessels, perivascular exudates, diffuse peripheral exudation, and intraretinal hemorrhages. Clinical and angiographical findings were consistent with familial exudative vitreoretinopathy, while genetic testing identified variants of uncertain significance in two associated genes, LRP5 and ZNF408. In silico analysis predicts the LRP5 variant to be pathogenic. Retinal vasculopathies often have phenotypic overlap, warranting angiographic examination of both eyes and genetic testing to uncover the correct diagnosis and guide proper treatment. [Ophthalmic Surg Lasers Imaging Retina 2024;55:462-466.].


Asunto(s)
Vitreorretinopatías Exudativas Familiares , Angiografía con Fluoresceína , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Humanos , Femenino , Vitreorretinopatías Exudativas Familiares/diagnóstico , Niño , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Angiografía con Fluoresceína/métodos , Telangiectasia Retiniana/diagnóstico , Telangiectasia Retiniana/genética , Tomografía de Coherencia Óptica/métodos , Fondo de Ojo , Mutación , Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/genética , Linaje , Proteínas de Unión al ADN , Factores de Transcripción
9.
Eur J Ophthalmol ; 34(5): NP8-NP12, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38720524

RESUMEN

PURPOSE: This report aims to delineate distinct phenotypes of Familial Exudative Vitreoretinopathy (FEVR) observed in a mother and her daughter, both harboring a novel LRP5 pathogenic variation. METHODS: The investigation involves a retrospective review of medical records accompanied by multimodal imaging. Molecular characterization was performed using whole exon sequencing, and the pathogenic variant was subsequently confirmed through Sanger sequencing. RESULT: A 6-year-old girl diagnosed with anisometropic amblyopia exhibited macular dragging and peripheral avascular retina in her right eye. Whole exon sequencing identified a previously unreported heterozygous missense LRP5 pathogenic variation, Glu528Lys. Simultaneously, her 43-year-old mother also carried the same mutation, manifesting peripheral exudations, avascular areas, and multiple microaneurysms. Notably, both cases presented distinctive phenotypes of FEVR. CONCLUSION: Our findings underscore the diversity in clinical presentations associated with FEVR, emphasizing the pivotal role of genetic evaluation. Despite variations in severity between the eyes of the same patient, it is crucial to remain vigilant for potential progression to a pathological status in the seemingly normal eye. Additionally, this study contributes to expanding the genetic spectrum of FEVR.


Asunto(s)
Vitreorretinopatías Exudativas Familiares , Angiografía con Fluoresceína , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Linaje , Fenotipo , Tomografía de Coherencia Óptica , Humanos , Femenino , Vitreorretinopatías Exudativas Familiares/diagnóstico , Vitreorretinopatías Exudativas Familiares/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Adulto , Niño , Estudios Retrospectivos , Análisis Mutacional de ADN , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/diagnóstico , Agudeza Visual/fisiología , Mutación Missense , Madres , Imagen Multimodal , ADN/genética , Exones/genética
10.
Osteoporos Int ; 35(8): 1395-1406, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38625381

RESUMEN

Osteoporosis-pseudoglioma syndrome (OPPG) and LRP5 high bone mass (LRP5-HBM) are two rare bone diseases with opposite clinical symptoms caused by loss-of-function and gain-of-function mutations in LRP5. Bisphosphonates are an effective treatment for OPPG patients. LRP5-HBM has a benign course, and age-related bone loss is found in one LRP5-HBM patient. PURPOSE: Low-density lipoprotein receptor-related protein 5 (LRP5) is involved in the canonical Wnt signaling pathway. The gain-of-function mutation leads to high bone mass (LRP5-HBM), while the loss-of-function mutation leads to osteoporosis-pseudoglioma syndrome (OPPG). In this study, the clinical manifestations, disease-causing mutations, treatment, and follow-up were summarized to improve the understanding of these two diseases. METHODS: Two OPPG patients and four LRP5-HBM patients were included in this study. The clinical characteristics, biochemical and radiological examinations, pathogenic mutations, and structural analysis were summarized. Furthermore, several patients were followed up to observe the treatment effect and disease progress. RESULTS: Congenital blindness, persistent bone pain, low bone mineral density (BMD), and multiple brittle fractures were the main clinical manifestations of OPPG. Complex heterozygous mutations were detected in two OPPG patients. The c.1455G > T mutation in exon 7 was first reported. During the follow-up, BMD of two patients was significantly improved after bisphosphonate treatment. On the contrary, typical clinical features of LRP5-HBM included extremely high BMD without fractures, torus palatinus and normal vision. X-ray showed diffuse osteosclerosis. Two heterozygous missense mutations were detected in four patients. In addition, age-related bone loss was found in one LRP5-HBM patient after 12-year of follow-up. CONCLUSION: This study deepened the understanding of the clinical characteristics, treatment, and follow-up of OPPG and LRP5-HBM; expanded the pathogenic gene spectrum of OPPG; and confirmed that bisphosphonates were effective for OPPG. Additionally, it was found that Ala242Thr mutation could not protect LRP5-HBM patients from age-related bone loss. This phenomenon deserves further study.


Asunto(s)
Conservadores de la Densidad Ósea , Densidad Ósea , Difosfonatos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Mutación , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Densidad Ósea/fisiología , Densidad Ósea/genética , Femenino , Conservadores de la Densidad Ósea/uso terapéutico , Conservadores de la Densidad Ósea/farmacología , Masculino , Difosfonatos/uso terapéutico , Difosfonatos/farmacología , Adulto , Estudios de Seguimiento , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/fisiopatología , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/complicaciones , Adolescente , Adulto Joven , Persona de Mediana Edad , Niño
11.
J Bone Miner Res ; 39(3): 315-325, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38477773

RESUMEN

Environmental factors and genetic variation individually impact bone. However, it is not clear how these factors interact to influence peak bone mass accrual. Here we tested whether genetically programmed high bone formation driven by missense mutations in the Lrp5 gene (Lrp5A214V) altered the sensitivity of mice to an environment of inadequate dietary calcium (Ca) intake. Weanling male Lrp5A214V mice and wildtype littermates (control) were fed AIN-93G diets with 0.125%, 0.25%, 0.5% (reference, basal), or 1% Ca from weaning until 12 weeks of age (ie, during bone growth). Urinary Ca, serum Ca, Ca regulatory hormones (PTH, 1,25 dihydroxyvitamin D3 (1,25(OH)2D3)), bone parameters (µCT, ash), and renal/intestinal gene expression were analyzed. As expected, low dietary Ca intake negatively impacted bones and Lrp5A214V mice had higher bone mass and ash content. Although bones of Lrp5A214V mice have more matrix to mineralize, their bones were not more susceptible to low dietary Ca intake. In control mice, low dietary Ca intake exerted expected effects on serum Ca (decreased), PTH (increased), and 1,25(OH)2D3 (increased) as well as their downstream actions (ie, reducing urinary Ca, increasing markers of intestinal Ca absorption). In contrast, Lrp5A214V mice had elevated serum Ca with a normal PTH response but a blunted 1,25(OH)2D3 response to low dietary Ca that was reflected in the renal 1,25(OH)2D3 producing/degrading enzymes, Cyp27b1 and Cyp24a1. Despite elevated serum Ca in Lrp5A214V mice, urinary Ca was not elevated. Despite an abnormal serum 1,25(OH)2D3 response to low dietary Ca, intestinal markers of Ca absorption (Trpv6, S100g mRNA) were elevated in Lrp5A214V mice and responded to low Ca intake. Collectively, our data indicate that the Lrp5A214V mutation induces changes in Ca homeostasis that permit mice to retain more Ca and support their high bone mass phenotype.


Optimizing peak bone mass (PBM) is critical for strong bones and osteoporosis prevention. Both genetics and dietary factors like calcium (Ca) contribute to PBM. The goal of this research study was to determine how dietary Ca intake and genetics interact with each other to impact bone mass. Lowering dietary Ca in control mice causes hormonal changes that increase intestinal Ca absorption and reduce urinary Ca loss to protect bone; but this process fails when dietary Ca becomes too low. However, mice with genetically programmed high bone mass could maintain high bone mass even when challenged with Ca deficient diets. This protection is because the high bone mass mice maintain higher serum Ca, have altered production and utilization of Ca-regulating hormones, and have increased molecular indicators of intestinal Ca absorption and kidney Ca retention. Our findings are important because they demonstrate how a genetic program that increases bone formation can drive improved efficiency of Ca utilization to accommodate the increased need for Ca deposition into bone. We believe that our preclinical study provides important proof-of-principle support for the concept of personalized recommendations for bone health management.


Asunto(s)
Calcio de la Dieta , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Animales , Masculino , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Calcio de la Dieta/farmacología , Calcio de la Dieta/metabolismo , Ratones , Vitamina D/metabolismo , Vitamina D/farmacología , Vitamina D/administración & dosificación , Hormona Paratiroidea/sangre , Hormona Paratiroidea/metabolismo , Huesos/metabolismo , Huesos/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Calcio/metabolismo , Calcio/orina , Calcitriol/sangre , Calcitriol/farmacología , Calcitriol/metabolismo , Tamaño de los Órganos/efectos de los fármacos
12.
J Biochem Mol Toxicol ; 38(4): e23677, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528715

RESUMEN

The study investigated the potential association of the low-density lipoprotein (LDL) genome with endometrial cancer progression based on the Gene Expression Omnibus data set and The Cancer Genome Atlas data set. Differential and weighted gene coexpression network analysis was performed on endometrial cancer transcriptome datasets GSE9750 and GSE106191. The protein-protein interaction network was built using LDL-receptor proteins and the top 50 tumor-associated genes. Low-density lipoprotein-related receptors 5/6 (LRP5/6) in endometrial cancer tissues were correlated with oncogenes, cell cycle-related genes, and immunological checkpoints using Spearman correlation. MethPrimer predicted the LRP5/6 promoter CpG island. LRP2, LRP6, LRP8, LRP12, low-density lipoprotein receptor-related protein-associated protein, and LRP5 were major LDL-receptor-related genes associated with endometrial cancer. LRP5/6 was enriched in various cancer-related pathways and may be a key LDL-receptor-related gene in cancer progression. LRP5/6 may be involved in the proliferation process of endometrial cancer cells by promoting the expression of cell cycle-related genes. LRP5/6 may be involved in the proliferation of endometrial cancer cells by promoting the expression of cell cycle-related genes. LRP5/6 may promote the immune escape of cancer cells by promoting the expression of immune checkpoints, promoting endometrial cancer progression. The MethPrimer database predicted that the LRP5/6 promoter region contained many CpG islands, suggesting that DNA methylation can occur in the LRP5/6 promoter region. LRP5/6 may aggravate endometrial cancer by activating the phosphoinositide 3-kinase/protein kinase B pathway.


Asunto(s)
Neoplasias Endometriales , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Humanos , Femenino , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Fosfatidilinositol 3-Quinasas , Receptores de LDL , Neoplasias Endometriales/genética , Lipoproteínas LDL
13.
J Orthop Surg Res ; 19(1): 104, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302983

RESUMEN

OBJECTIVE: To analyze the relationship between the polymorphism and mutation of rs7125942 and rs3736228 locus in the low-density lipoprotein receptor-related protein 5 (LRP5) genotype and bone mineral density (BMD) in postmenopausal women in Xinjiang, China, to provide a basis for prevention and treatment of the disease. METHODS: According to the results of dual-energy X-ray (DEXA) determination of BMD, the 136 subjects were divided into three groups: Group A: normal bone mass, Group B: osteopenia, Group C: osteoporosis. 1. Age, body, mass index (BMI), and menopause of all subjects were recorded. 2. Fasting blood glucose (FBG), glycosylated hemoglobin (HbA1c), calcium (Ca), phosphorus (P), alkaline phosphatase (ALP), and clinical biochemical data were determined. 3. LRP5 locus polymorphisms were determined by time-of-flight mass spectrometry. RESULTS: 1. Compared with group A, the age, ALP, Cr, and BUN levels in group B and group C were increased, but UA levels were lower (P < 0.05), and Serum P was higher in the group C (P < 0.05). 2. There was no statistically significant difference in the prevalence of diabetes between the three groups (P > 0.05). 3. The ROC curves for different BMD sites such as L1, L2, L3, L4, L total, and femoral neck were 0.929, 0.955, 0.901, 0.914, 0.885, and 0.873 (P < 0.01). 4. At rs7125942 locus, there was statistically significant difference in the distribution of wild-type (CC) and mutant (CG) with the normal bone mass (NBM) group and the abnormal bone mass (ABM) group (P < 0.05). 5. At rs7125942 locus, compared with wild-type (CC), mutant (CG) had lower LDL and FPG in NBM group (P < 0.05), and lower serum ALP in the ABM group (P < 0.05). At rs3736228 locus, the BMD (Femoral neck) of mutant (CT/TT) was lower than that of wild-type (CC) in the NBM group (P < 0.05). 6. Age and menopausal years were negatively correlated with BMD of the femoral neck and L1-4 (P < 0.05), and BMI and TG were positively (P < 0.05), and the results of multiple linear regression analysis showed that age, BMI, and TG were both independent factors affecting BMD (P < 0.05).


Asunto(s)
Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Osteoporosis Posmenopáusica , Humanos , Femenino , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Posmenopausia/genética , Densidad Ósea/genética , Polimorfismo Genético , Mutación , Osteoporosis Posmenopáusica/genética
14.
J Cell Physiol ; 239(4): e31183, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38348695

RESUMEN

Osteogenic differentiation is important for fracture healing. Microfibrial-associated glycoprotein 2 (MAGP2) is found to function as a proangiogenic regulator in bone formation; however, its role in osteogenic differentiation during bone repair is not clear. Here, a mouse model of critical-sized femur fracture was constructed, and the adenovirus expressing MAGP2 was delivered into the fracture site. Mice with MAGP2 overexpression exhibited increased bone mineral density and bone volume fraction (BV/TV) at Day 14 postfracture. Within 7 days postfracture, overexpression of MAGP2 increased collagen I and II expression at the fracture callus, with increasing chondrogenesis. MAGP2 inhibited collagen II level but elevated collagen I by 14 days following fracture, accompanied by increased endochondral bone formation. In mouse osteoblast precursor MC3T3-E1 cells, MAGP2 treatment elevated the expression of osteoblastic factors (osterix, BGLAP and collagen I) and enhanced ALP activity and mineralization through activating ß-catenin signaling after osteogenic induction. Besides, MAGP2 could interact with lipoprotein receptor-related protein 5 (LRP5) and upregulated its expression. Promotion of osteogenic differentiation and ß-catenin activation mediated by MAGP2 was partially reversed by LRP5 knockdown. Interestingly, ß-catenin/transcription factor 4 (TCF4) increased MAGP2 expression probably by binding to MAGP2 promoter. These findings suggest that MAGP2 may interact with ß-catenin/TCF4 to enhance ß-catenin/TCF4's function and activate LRP5-activated ß-catenin signaling pathway, thus promoting osteogenic differentiation for fracture repair. mRNA sequencing identified the potential targets of MAGP2, providing novel insights into MAGP2 function and the directions for future research.


Asunto(s)
Fracturas Óseas , Osteogénesis , Animales , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Diferenciación Celular/genética , Colágeno/metabolismo , Curación de Fractura , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Osteoblastos/metabolismo , Vía de Señalización Wnt , Masculino , Ratones Endogámicos C57BL , Línea Celular
15.
Am J Ophthalmol ; 262: 73-85, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38280677

RESUMEN

PURPOSE: This study aimed to ascertain the occurrence of foveal hypoplasia (FH) in individuals diagnosed with familial exudative vitreoretinopathy (FEVR). DESIGN: Retrospective cohort study. METHODS: In this study, FEVR families and sporadic cases were diagnosed at the Eye and ENT Hospital, Fudan University, between 2017 and 2023. All patients attended routine ophthalmologic examinations and genetic screenings. The classification of FH was determined using optical coherence tomography (OCT) scans. The FH condition was classified into 2 subgroups: group A (FH being limited to the inner layers) and group B (FH affecting the outer layers). A total of 102 eyes from 58 patients were suitable for analysis. RESULTS: Forty-nine mutations in LRP5, FZD4, NDP, TSPAN12, KIF11, CTNNB1, and ZNF408 were examined and detected, with 26 of them being novel. Forty-seven eyes (46.1%) revealed FH. The majority (53.2%) were due to the typical grade 1 FH. Patients with mutations in LRP5 and KIF11 were found to exhibit a higher prevalence of FH (P = .0088). Group B displayed the lowest visual acuity compared with group A (P = .048) and the group without FH (P < .001). The retinal arteriolar angle in group B was significantly smaller than in group A (P = .001) and those without FH (P < .001). CONCLUSIONS: This study offers a new diagnostic approach and expands the spectrum of FEVR mutations. LRP5 and KIF11 were found to be more susceptible to causing FH in patients with FEVR. FEVR eyes with FH exhibited both greater visual impairment and reduced retinal arteriolar angles. The assessment of foveal status in patients with FEVR should be valued.


Asunto(s)
Enfermedades Hereditarias del Ojo , Proteínas del Ojo , Vitreorretinopatías Exudativas Familiares , Fóvea Central , Receptores Frizzled , Cinesinas , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Mutación , Tetraspaninas , Tomografía de Coherencia Óptica , Agudeza Visual , Humanos , Masculino , Vitreorretinopatías Exudativas Familiares/diagnóstico , Femenino , Estudios Retrospectivos , Fóvea Central/anomalías , Cinesinas/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Adulto , Proteínas del Ojo/genética , Agudeza Visual/fisiología , Niño , Receptores Frizzled/genética , Adolescente , Tetraspaninas/genética , Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/fisiopatología , Adulto Joven , Enfermedades de la Retina/genética , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/fisiopatología , Análisis Mutacional de ADN , Linaje , Angiografía con Fluoresceína/métodos , Preescolar , Persona de Mediana Edad , Anomalías del Ojo/genética , Anomalías del Ojo/diagnóstico , Proteínas de Unión al ADN , Proteínas del Tejido Nervioso , Factores de Transcripción
16.
Exp Cell Res ; 434(1): 113857, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38008278

RESUMEN

Genetic factors coordinate with environmental factors to drive the pathogenesis of prostate adenocarcinoma (PRAD). SPOP is one of the most mutated genes and LRP5 mediates lipid metabolism that is abnormally altered in PRAD. Here, we investigated the potential cross-talk between SPOP and LRP5 in PRAD. We find a negative correlation between SPOP and LRP5 proteins in PRAD. SPOP knockdown increased LRP5 protein while SPOP overexpression resulted in LRP5 reduction that was fully rescued by proteasome inhibitors. LRP5 intracellular tail has SPOP binding site and the direct interaction between LRP5 and SPOP was confirmed by Co-IP and GST-pulldown. Moreover, LRP5 competed with Daxx for SPOP-mediated degradation, establishing a dynamic balance among SPOP, LRP5 and Daxx. Overexpression of LRP5 tail could shift this balance to enhance Daxx-mediated transcriptional inhibition, and inhibit T cell activity in a co-culture system. Further, we generated human and mouse prostate cancer cell lines expressing SPOP variants (F133V, A227V, R368H). SPOP-F133V and SPOP-A227V have specific effects in up-regulating the protein levels of PD-1 and PD-L1. Consistently, SPOP-F133V and SPOP-A227V show robust inhibitory effects on T cells compared to WT SPOP in co-culture. This is further supported by the mouse syngeneic model showing that SPOP-F133V and SPOP-A227V enhance tumorigenesis of prostate cancer in in-vivo condition. Taken together, our study provides evidence that SPOP-LRP5 crosstalk plays an essential role, and the genetic variants of SPOP differentially modulate the expression and activity of immune checkpoints in prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Proteínas Represoras , Masculino , Animales , Ratones , Humanos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Antígeno B7-H1/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/patología , Carcinogénesis/genética , Transformación Celular Neoplásica , Mutación , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Chaperonas Moleculares/genética , Proteínas Co-Represoras/genética
17.
BMC Ophthalmol ; 23(1): 489, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38030997

RESUMEN

BACKGROUND: Familial exudative vitreoretinopathy (FEVR) is a genetic eye disorder that leads to abnormal development of retinal blood vessels, resulting in vision impairment. This study aims to identify pathogenic variants by targeted exome sequencing in 9 independent pedigrees with FEVR and characterize the novel pathogenic variants by molecular dynamics simulation. METHODS: Clinical data were collected from 9 families with FEVR. The causative genes were screened by targeted next-generation sequencing (TGS) and verified by Sanger sequencing. In silico analyses (SIFT, Polyphen2, Revel, MutationTaster, and GERP + +) were carried out to evaluate the pathogenicity of the variants. Molecular dynamics was simulated to predict protein conformation and flexibility transformation alterations on pathogenesis. Furthermore, molecular docking techniques were employed to explore the interactions and binding properties between LRP5 and DKK1 proteins relevant to the disease. RESULTS: A 44% overall detection rate was achieved with four variants including c.4289delC: p.Pro1431Argfs*8, c.2073G > T: p.Trp691Cys, c.1801G > A: p.Gly601Arg in LRP5 and c.633 T > A: p.Tyr211* in TSPAN12 in 4 unrelated probands. Based on in silico analysis and ACMG standard, two of them, c.4289delC: p.Pro1431Argfs*8 and c.2073G > T: p.Trp691Cys of LRP5 were identified as novel pathogenic variants. Based on computational predictions using molecular dynamics simulations and molecular docking, there are indications that these two variants might lead to alterations in the secondary structure and spatial conformation of the protein, potentially impacting its rigidity and flexibility. Furthermore, these pathogenic variants are speculated to potentially influence hydrogen bonding interactions and could result in an increased binding affinity with the DKK1 protein. CONCLUSIONS: Two novel genetic variants of the LRP5 gene were identified, expanding the range of mutations associated with FEVR. Through molecular dynamics simulations and molecular docking, the potential impact of these variants on protein structure and their interactions with the DKK1 protein has been explored. These findings provide further support for the involvement of these variants in the pathogenesis of the disease.


Asunto(s)
Enfermedades Hereditarias del Ojo , Enfermedades de la Retina , Humanos , Vitreorretinopatías Exudativas Familiares , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Simulación del Acoplamiento Molecular , Enfermedades Hereditarias del Ojo/genética , Tetraspaninas/genética , Análisis Mutacional de ADN , Mutación , Linaje , Fenotipo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo
18.
Genes (Basel) ; 14(10)2023 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-37895195

RESUMEN

The formation and maintenance of the gross structure and microarchitecture of the human skeleton require the concerted functioning of a plethora of morphogenic signaling processes. Through recent discoveries in the field of genetics, numerous genotypic variants have been implicated in pathologic skeletal phenotypes and disorders arising from the disturbance of one or more of these processes. For example, total loss-of-function variants of LRP5 were found to be the cause of osteoporosis-pseudoglioma syndrome (OPPG). LRP5 encodes for the low-density lipoprotein receptor-related protein 5, a co-receptor in the canonical WNT-ß-catenin signaling pathway and a crucial protein involved in the formation and maintenance of homeostasis of the human skeleton. Beyond OPPG, other partial loss-of-function variants of LRP5 have been found to be associated with other low bone mass phenotypes and disorders, while LRP5 gain-of-function variants have been implicated in high bone mass phenotypes. This review introduces the roles that LRP5 plays in skeletal morphogenesis and discusses some of the structural consequences that result from abnormalities in LRP5. A greater understanding of how the LRP5 receptor functions in bone and other body tissues could provide insights into a variety of pathologies and their potential treatments, from osteoporosis and a variety of skeletal abnormalities to congenital disorders that can lead to lifelong disabilities.


Asunto(s)
Osteogénesis Imperfecta , Osteoporosis , Humanos , Densidad Ósea/genética , Osteoporosis/genética , Osteoporosis/complicaciones , Osteogénesis Imperfecta/genética , Huesos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética
19.
Arch Osteoporos ; 18(1): 112, 2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37659026

RESUMEN

PURPOSE: LRP5 high bone mass (HBM) is an autosomal dominant endosteal hyperostosis caused by mutations of the low-density lipoprotein receptor-related protein 5 (LRP5) gene. Alternative names included "autosomal dominant osteosclerosis" and "Worth disease." The aim of the paper is to provide an historical overview of a disorder whose literature is complicated and confusing due to the past use of several denominations and lack of reviews. METHODS: We collected case reports of HBM with evidence of autosomal dominant transmission preceding the identification of the LRP5 mutations in 2002 (Worth-type endosteal hyperostosis) and cases of LRP5 HBM confirmed by genetic analysis since 2002. The prevalence of relevant clinical and laboratory findings was estimated. We described an affected woman with neurological manifestations. RESULTS: A 44-year-old Caucasian woman with torus palatinus complained of headache, hypo-/anosmia, and complete mixed deafness. Dual-energy X-ray absorptiometry (DEXA) scan revealed elevated bone mass. The A242T mutation of the LRP5 gene was detected. Including the present case, 155 patients have been reported to date. Neurological involvement and increased serum alkaline phosphatase (ALP) were present in 19.4% and 3.7% of cases, respectively. Facial changes and torus palatinus were observed in 61% and 41% of cases, respectively. CONCLUSIONS: We present the only historical review on Worth-type endosteal hyperostosis, now known as LRP5 HBM. Neurological manifestations, previously considered absent in the disease, affect 19.4% of the patients. Genetic analysis and appropriate denomination of LRP5 HBM are fundamental for diagnosis and to mitigate the confusion that has long characterized this disease.


Asunto(s)
Artrogriposis , Hiperostosis Cortical Congénita , Femenino , Humanos , Adulto , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética
20.
Bone Res ; 11(1): 47, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37612291

RESUMEN

Proper regulation of Wnt signaling is critical for normal bone development and homeostasis. Mutations in several Wnt signaling components, which increase the activity of the pathway in the skeleton, cause high bone mass in human subjects and mouse models. Increased bone mass is often accompanied by severe headaches from increased intracranial pressure, which can lead to fatality and loss of vision or hearing due to the entrapment of cranial nerves. In addition, progressive forehead bossing and mandibular overgrowth occur in almost all subjects. Treatments that would provide symptomatic relief in these subjects are limited. Porcupine-mediated palmitoylation is necessary for Wnt secretion and binding to the frizzled receptor. Chemical inhibition of porcupine is a highly selective method of Wnt signaling inhibition. We treated three different mouse models of high bone mass caused by aberrant Wnt signaling, including homozygosity for loss-of-function in Sost, which models sclerosteosis, and two strains of mice carrying different point mutations in Lrp5 (equivalent to human G171V and A214V), at 3 months of age with porcupine inhibitors for 5-6 weeks. Treatment significantly reduced both trabecular and cortical bone mass in all three models. This demonstrates that porcupine inhibition is potentially therapeutic for symptomatic relief in subjects who suffer from these disorders and further establishes that the continued production of Wnts is necessary for sustaining high bone mass in these models.


Asunto(s)
Mutación con Ganancia de Función , Hiperostosis , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales , Secreciones Corporales , Modelos Animales de Enfermedad , Hiperostosis/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...