Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
1.
Nat Commun ; 15(1): 1227, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418480

RESUMEN

Exploring the molecular basis of disease severity in rare disease scenarios is a challenging task provided the limitations on data availability. Causative genes have been described for Congenital Myasthenic Syndromes (CMS), a group of diverse minority neuromuscular junction (NMJ) disorders; yet a molecular explanation for the phenotypic severity differences remains unclear. Here, we present a workflow to explore the functional relationships between CMS causal genes and altered genes from each patient, based on multilayer network community detection analysis of complementary biomedical information provided by relevant data sources, namely protein-protein interactions, pathways and metabolomics. Our results show that CMS severity can be ascribed to the personalized impairment of extracellular matrix components and postsynaptic modulators of acetylcholine receptor (AChR) clustering. This work showcases how coupling multilayer network analysis with personalized -omics information provides molecular explanations to the varying severity of rare diseases; paving the way for sorting out similar cases in other rare diseases.


Asunto(s)
Síndromes Miasténicos Congénitos , Humanos , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/diagnóstico , Unión Neuromuscular/metabolismo , Enfermedades Raras/metabolismo , Flujo de Trabajo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Mutación
2.
Traffic ; 25(1): e12927, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38272446

RESUMEN

Endoplasmic reticulum (ER) retention of misfolded glycoproteins is mediated by the ER-localized eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognizes a misfolded glycoprotein and flags it for ER retention by re-glucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease, even if the mutant glycoprotein retains activity ("responsive mutant"). Using confocal laser scanning microscopy, we investigated here the subcellular localization of the human Trop-2-Q118E, E227K and L186P mutants, which cause gelatinous drop-like corneal dystrophy (GDLD). Compared with the wild-type Trop-2, which is correctly localized at the plasma membrane, these Trop-2 mutants are retained in the ER. We studied fluorescent chimeras of the Trop-2 Q118E, E227K and L186P mutants in mammalian cells harboring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 genes. The membrane localization of the Trop-2 Q118E, E227K and L186P mutants was successfully rescued in UGGT1-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation would constitute a novel therapeutic strategy for the treatment of pathological conditions associated to misfolded membrane glycoproteins (whenever the mutation impairs but does not abrogate function), and it encourages the testing of modulators of ER glycoprotein folding quality control as broad-spectrum rescue-of-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.


Asunto(s)
Pliegue de Proteína , Enfermedades Raras , Animales , Humanos , Enfermedades Raras/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Retículo Endoplásmico/metabolismo , Mutación , Mamíferos/metabolismo , Glucosiltransferasas/metabolismo
3.
Ter Arkh ; 95(6): 505-510, 2023 Aug 17.
Artículo en Ruso | MEDLINE | ID: mdl-38158971

RESUMEN

Fabry-Andersen disease is a genetically determined, progressive disease related to lysosomal storage diseases, linked to the X chromosome, characterized by impaired glycosphingolipid metabolism, due to the deficiency or absence of the enzyme α-galactosidase A. Fabry disease is a multisystem disease and is characterized by damage to vital organs - kidneys, heart, brain, with the occurrence of complications that cause an unfavorable prognosis. Autoinflammation mechanisms with signs of chronic inflammation are involved in the pathogenesis of the disease. One of the features of Fabry disease are clinical manifestations in the form of arthralgia, fever, skin lesions, which are similar to rheumatological diseases. The article presents a clinical observation of the classical type of Fabry disease with multiple organ manifestation, which required differential diagnosis with rheumatological diseases. Rheumatologists are specialists who are involved in the early diagnosis of Fabry disease, so they should have a high awareness of this sphingolipidosis.


Asunto(s)
Enfermedad de Fabry , Enfermedades Reumáticas , Humanos , Enfermedad de Fabry/complicaciones , Enfermedad de Fabry/diagnóstico , Enfermedades Raras/diagnóstico , Enfermedades Raras/complicaciones , Enfermedades Raras/metabolismo , Riñón/patología , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Enfermedades Reumáticas/etiología , Enfermedades Reumáticas/complicaciones
4.
Int J Cancer ; 153(12): 1997-2010, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37548077

RESUMEN

Sarcomatoid carcinoma (SC), which can occur in any organ, is a rare disease. To elucidate common characteristics of SC beyond organs, we evaluated clinicopathological and immunological features of SC defined by the single histological criterion beyond organs compared to randomly matched conventional carcinoma (non-SC) adjusted for the disease stage. Immunological features were assessed by multiplex immunohistochemistry, comparing immune cell density in tumor tissues and tumor programmed death-ligand 1 (PD-L1) expression. A total of 101 patients with SC or non-SC (31 lung, 19 esophagus, 22 pancreas, 15 liver, 4 bile duct, 6 kidney, 2 uterus and 2 ovary) were identified among 7197 patients who underwent surgery at our institute (1997-2020). SC was significantly associated with worse survival (HR: 1.571; 95% CI: 1.084-2.277; P = .017). The frequency of postoperative progression within 6 months was significantly higher for SC patients (54% vs 28%; P = .002). The immune profiling revealed the densities of CD8+ T cells (130 vs 72 cells/mm2 ; P = .004) and tumor-associated macrophages (566 vs 413 cells/mm2 ; P < .0001) and the tumor PD-L1 expression score (40% vs 5%; P < .0001) were significantly higher in SCs than in non-SCs. Among 73 SC patients with postoperative progression, multivariate Cox regression analysis showed that immunotherapy tended to be associated with favorable survival (HR: 0.256; 95% CI: 0.062-1.057; P = .060). Collectively, SCs shared clinicopathological and immunological features across organs. Our study can initiate to standardize the pathological definition of SC and provide a rationale for the investigation and development for this rare disease in a cross-organ manner.


Asunto(s)
Carcinoma , Neoplasias Pulmonares , Femenino , Humanos , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/metabolismo , Pronóstico , Enfermedades Raras/metabolismo , Carcinoma/metabolismo , Neoplasias Pulmonares/patología
5.
J Inorg Biochem ; 247: 112334, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37499466

RESUMEN

The deregulation of copper homoeostasis can promote various diseases such as Menkes disease or hypertrophic cardioencephalomyopathy. We have recently synthesized solid copper(II) complexes ([Cu(His)2Cl2] and [Cu(Ser)2]), stable in physiological media and with potential as therapeutic agents. This report describes: i) the biocompatibility of these complexes at concentrations up to 100 µM using a differentiated Caco-2 cells model; ii) their transport across the intestinal epithelium using a transepithelial resistance assay and monitoring the amount of copper complexes at the apical and basolateral sides of the cells. The results suggest that the flow occurs through paracellular routes. The intracellular copper retention was <2.7% with no significant differences in intracellular copper content between 6 h and 48 h, suggesting an early copper retention process. Furthermore, this is the first evidence that demonstrates [Cu(His)2Cl2] and [Cu(Ser)2] induce transcriptional downregulation of the four major copper transporters (CTR1, DMT1, ATP7A, ATP7B), and the upregulation of the metallothionein gene expression. A remarkable finding was the increase in cytochrome c oxidase activity observed after the treatment of differentiated Caco-2 cells with copper(II) complexes at concentrations of 50-100 µM. The understanding of the transport mechanisms of these copper(II) complexes across the intestinal epithelium and of their subsequent biological activities could contribute to the development of optimal pharmaceutical formulations for the therapy of copper deficiency-related diseases.


Asunto(s)
Proteínas de Transporte de Catión , Cobre , Humanos , Cobre/farmacología , Células CACO-2 , Enfermedades Raras/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Mucosa Intestinal/metabolismo , ATPasas Transportadoras de Cobre/genética , ATPasas Transportadoras de Cobre/metabolismo
6.
Cells ; 11(22)2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36429129

RESUMEN

Lysosome-related organelles (LROs) are a group of functionally diverse, cell type-specific compartments. LROs include melanosomes, alpha and dense granules, lytic granules, lamellar bodies and other compartments with distinct morphologies and functions allowing specialised and unique functions of their host cells. The formation, maturation and secretion of specific LROs are compromised in a number of hereditary rare multisystem disorders, including Hermansky-Pudlak syndromes, Griscelli syndrome and the Arthrogryposis, Renal dysfunction and Cholestasis syndrome. Each of these disorders impacts the function of several LROs, resulting in a variety of clinical features affecting systems such as immunity, neurophysiology and pigmentation. This has demonstrated the close relationship between LROs and led to the identification of conserved components required for LRO biogenesis and function. Here, we discuss aspects of this conserved machinery among LROs in relation to the heritable multisystem disorders they associate with, and present our current understanding of how dysfunctions in the proteins affected in the disease impact the formation, motility and ultimate secretion of LROs. Moreover, we have analysed the expression of the members of the CHEVI complex affected in Arthrogryposis, Renal dysfunction and Cholestasis syndrome, in different cell types, by collecting single cell RNA expression data from the human protein atlas. We propose a hypothesis describing how transcriptional regulation could constitute a mechanism that regulates the pleiotropic functions of proteins and their interacting partners in different LROs.


Asunto(s)
Artrogriposis , Colestasis , Enfermedades Renales , Humanos , Lisosomas/metabolismo , Melanosomas/metabolismo , Enfermedades Raras/metabolismo , Colestasis/metabolismo , Enfermedades Renales/metabolismo
7.
Biomolecules ; 12(7)2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35883565

RESUMEN

The importance of mitochondria in mammalian cells is widely known. Several biochemical reactions and pathways take place within mitochondria: among them, there are those involving the biogenesis of the iron-sulfur (Fe-S) clusters. The latter are evolutionarily conserved, ubiquitous inorganic cofactors, performing a variety of functions, such as electron transport, enzymatic catalysis, DNA maintenance, and gene expression regulation. The synthesis and distribution of Fe-S clusters are strictly controlled cellular processes that involve several mitochondrial proteins that specifically interact each other to form a complex machinery (Iron Sulfur Cluster assembly machinery, ISC machinery hereafter). This machinery ensures the correct assembly of both [2Fe-2S] and [4Fe-4S] clusters and their insertion in the mitochondrial target proteins. The present review provides a structural and molecular overview of the rare diseases associated with the genes encoding for the accessory proteins of the ISC machinery (i.e., GLRX5, ISCA1, ISCA2, IBA57, FDX2, BOLA3, IND1 and NFU1) involved in the assembly and insertion of [4Fe-4S] clusters in mitochondrial proteins. The disease-related missense mutations were mapped on the 3D structures of these accessory proteins or of their protein complexes, and the possible impact that these mutations have on their specific activity/function in the frame of the mitochondrial [4Fe-4S] protein biogenesis is described.


Asunto(s)
Proteínas Hierro-Azufre , Humanos , Hierro/metabolismo , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Enfermedades Raras/metabolismo , Azufre/metabolismo
8.
Int J Mol Sci ; 22(24)2021 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-34948153

RESUMEN

Rare genetic diseases are a group of pathologies with often unmet clinical needs. Even if rare by a single genetic disease (from 1/2000 to 1/more than 1,000,000), the total number of patients concerned account for approximatively 400 million peoples worldwide. Finding treatments remains challenging due to the complexity of these diseases, the small number of patients and the challenge in conducting clinical trials. Therefore, innovative preclinical research strategies are required. The zebrafish has emerged as a powerful animal model for investigating rare diseases. Zebrafish combines conserved vertebrate characteristics with high rate of breeding, limited housing requirements and low costs. More than 84% of human genes responsible for diseases present an orthologue, suggesting that the majority of genetic diseases could be modelized in zebrafish. In this review, we emphasize the unique advantages of zebrafish models over other in vivo models, particularly underlining the high throughput phenotypic capacity for therapeutic screening. We briefly introduce how the generation of zebrafish transgenic lines by gene-modulating technologies can be used to model rare genetic diseases. Then, we describe how zebrafish could be phenotyped using state-of-the-art technologies. Two prototypic examples of rare diseases illustrate how zebrafish models could play a critical role in deciphering the underlying mechanisms of rare genetic diseases and their use to identify innovative therapeutic solutions.


Asunto(s)
Enfermedades Genéticas Congénitas , Modelos Genéticos , Enfermedades Raras , Pez Cebra , Animales , Investigación Biomédica , Modelos Animales de Enfermedad , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/terapia , Humanos , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Enfermedades Raras/terapia , Pez Cebra/genética , Pez Cebra/metabolismo
9.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884638

RESUMEN

Diagnosis and cure for rare diseases represent a great challenge for the scientific community who often comes up against the complexity and heterogeneity of clinical picture associated to a high cost and time-consuming drug development processes. Here we show a drug repurposing strategy applied to nephropathic cystinosis, a rare inherited disorder belonging to the lysosomal storage diseases. This approach consists in combining mechanism-based and cell-based screenings, coupled with an affordable computational analysis, which could result very useful to predict therapeutic responses at both molecular and system levels. Then, we identified potential drugs and metabolic pathways relevant for the pathophysiology of nephropathic cystinosis by comparing gene-expression signature of drugs that share common mechanisms of action or that involve similar pathways with the disease gene-expression signature achieved with RNA-seq.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinosis/tratamiento farmacológico , Cistinosis/genética , Reposicionamiento de Medicamentos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Enfermedades Raras/tratamiento farmacológico , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/efectos de la radiación , Células Cultivadas , Biología Computacional/métodos , Cistinosis/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Redes y Vías Metabólicas , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Transcriptoma
10.
Nat Commun ; 12(1): 6306, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753928

RESUMEN

Rare genetic diseases are typically caused by a single gene defect. Despite this clear causal relationship between genotype and phenotype, identifying the pathobiological mechanisms at various levels of biological organization remains a practical and conceptual challenge. Here, we introduce a network approach for evaluating the impact of rare gene defects across biological scales. We construct a multiplex network consisting of over 20 million gene relationships that are organized into 46 network layers spanning six major biological scales between genotype and phenotype. A comprehensive analysis of 3,771 rare diseases reveals distinct phenotypic modules within individual layers. These modules can be exploited to mechanistically dissect the impact of gene defects and accurately predict rare disease gene candidates. Our results show that the disease module formalism can be applied to rare diseases and generalized beyond physical interaction networks. These findings open up new venues to apply network-based tools for cross-scale data integration.


Asunto(s)
Redes Reguladoras de Genes , Enfermedades Raras/genética , Enfermedades Raras/patología , Algoritmos , Biología Computacional/métodos , Genotipo , Humanos , Modelos Biológicos , Fenotipo , Mapeo de Interacción de Proteínas/métodos , Enfermedades Raras/metabolismo
11.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34360713

RESUMEN

Rare central nervous system (CNS) tumours represent a unique challenge. Given the difficulty of conducting dedicated clinical trials, there is a lack of therapies for these tumours supported by high quality evidence, and knowledge regarding the impact of standard treatments (i.e., surgery, radiotherapy or chemotherapy) is commonly based on retrospective studies. Recently, new molecular techniques have led to the discovery of actionable molecular alterations. The aim of this article is to review recent progress in the molecular understanding of and therapeutic options for rare brain tumours, both in children and adults. We will discuss options such as targeting the mechanistic target of rapamycin (mTOR) pathway in subependymal giant cells astrocytomas (SEGAs) of tuberous sclerosis and BRAF V600E mutation in rare glial (pleomorphic xanthoastrocytomas) or glioneuronal (gangliogliomas) tumours, which are a model of how specific molecular treatments can also favourably impact neurological symptoms (such as seizures) and quality of life. Moreover, we will discuss initial experiences in targeting new molecular alterations in gliomas, such as isocitrate dehydrogenase (IDH) mutations and neurotrophic tyrosine receptor kinase (NTRK) fusions, and in medulloblastomas such as the sonic hedgehog (SHH) pathway.


Asunto(s)
Neoplasias Encefálicas , Terapia Molecular Dirigida , Mutación , Proteínas de Neoplasias , Enfermedades Raras , Transducción de Señal , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Enfermedades Raras/terapia
12.
Sci Rep ; 11(1): 15062, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301987

RESUMEN

High-throughput gene expression analysis is widely used. However, analysis is not straightforward. Multiple approaches should be applied and methods to combine their results implemented and investigated. We present methodology for the comprehensive analysis of expression data, including co-expression module detection and result integration via data-fusion, threshold based methods, and a Naïve Bayes classifier trained on simulated data. Application to rare-disease model datasets confirms existing knowledge related to immune cell infiltration and suggest novel hypotheses including the role of calcium channels. Application to simulated and spike-in experiments shows that combining multiple methods using consensus and classifiers leads to optimal results. ExpHunter Suite is implemented as an R/Bioconductor package available from https://bioconductor.org/packages/ExpHunterSuite . It can be applied to model and non-model organisms and can be run modularly in R; it can also be run from the command line, allowing scalability with large datasets. Code and reports for the studies are available from https://github.com/fmjabato/ExpHunterSuiteExamples .


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , RNA-Seq/métodos , Programas Informáticos , Algoritmos , Arabidopsis/genética , Teorema de Bayes , Canales de Calcio/genética , Humanos , Enfermedades Raras/genética , Enfermedades Raras/metabolismo
13.
Trends Cancer ; 7(8): 671-681, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34127435

RESUMEN

Melanoma is derived from melanocytes located in multiple regions of the body. Cutaneous melanoma (CM) represents the major subgroup, but less-common subtypes including uveal melanoma (UM), mucosal melanoma (MM), and acral melanoma (AM) arise that have distinct genetic profiles. Treatments effective for CM are ineffective in UM, AM, and MM, and patient survival remains poor. As reprogrammed cancer metabolism is associated with tumorigenesis, the underlying mechanisms are well studied and provide therapeutic opportunities in many cancers; however, metabolism is less well studied in rarer melanoma subtypes. We summarize current knowledge of the metabolic alterations in rare melanoma and potential applications of targeting cancer metabolism to improve the therapeutic options available to UM, AM, and MM patients.


Asunto(s)
Melanoma/metabolismo , Redes y Vías Metabólicas/genética , Membrana Mucosa/patología , Enfermedades Raras/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias de la Úvea/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Progresión de la Enfermedad , Humanos , Melanoma/genética , Melanoma/mortalidad , Melanoma/patología , Mutación , Enfermedades Raras/genética , Enfermedades Raras/mortalidad , Enfermedades Raras/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Tasa de Supervivencia , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/mortalidad , Neoplasias de la Úvea/patología
14.
Nat Rev Neurol ; 17(7): 433-447, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34050331

RESUMEN

The pathophysiology of complex neuroimmunological diseases, such as multiple sclerosis and autoimmune encephalitis, remains puzzling - various mechanisms that are difficult to dissect seem to contribute, hampering the understanding of the processes involved. Some rare neuroimmunological diseases are easier to study because their presentation and pathogenesis are more homogeneous. The investigation of these diseases can provide fundamental insights into neuroimmunological pathomechanisms that can in turn be applied to more complex diseases. In this Review, we summarize key mechanistic insights into three such rare but paradigmatic neuroimmunological diseases - Susac syndrome, Rasmussen encephalitis and narcolepsy type 1 - and consider the implications of these insights for the study of other neuroimmunological diseases. In these diseases, the combination of findings in humans, different modalities of investigation and animal models has enabled the triangulation of evidence to validate and consolidate the pathomechanistic features and to develop diagnostic and therapeutic strategies; this approach has provided insights that are directly relevant to other neuroimmunological diseases and applicable in other contexts. We also outline how next-generation technologies and refined animal models can further improve our understanding of pathomechanisms, including cell-specific and antigen-specific CNS immune responses, thereby paving the way for the development of targeted therapeutic approaches.


Asunto(s)
Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Nervioso/inmunología , Neuroinmunomodulación/fisiología , Enfermedades Raras/inmunología , Animales , Humanos , Enfermedades del Sistema Inmune/diagnóstico por imagen , Enfermedades del Sistema Inmune/metabolismo , Enfermedades del Sistema Nervioso/diagnóstico por imagen , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades Raras/diagnóstico por imagen , Enfermedades Raras/metabolismo , Síndrome de Susac/diagnóstico por imagen , Síndrome de Susac/inmunología , Síndrome de Susac/metabolismo
15.
Cells ; 10(4)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33917450

RESUMEN

Tripartite motif (TRIM) proteins are RING E3 ubiquitin ligases defined by a shared domain structure. Several of them are implicated in rare genetic diseases, and mutations in TRIM32 and TRIM-like malin are associated with Limb-Girdle Muscular Dystrophy R8 and Lafora disease, respectively. These two proteins are evolutionary related, share a common ancestor, and both display NHL repeats at their C-terminus. Here, we revmniew the function of these two related E3 ubiquitin ligases discussing their intrinsic and possible common pathophysiological pathways.


Asunto(s)
Enfermedades del Sistema Nervioso/metabolismo , Enfermedades Neuromusculares/metabolismo , Enfermedades Raras/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Humanos , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades Neuromusculares/fisiopatología , Enfermedades Raras/fisiopatología , Transducción de Señal , Proteínas de Motivos Tripartitos/química , Ubiquitina-Proteína Ligasas/química
16.
Int J Mol Sci ; 22(7)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916664

RESUMEN

DNA methylation (DNAme) profiling is used to establish specific biomarkers to improve the diagnosis of patients with inherited neurodevelopmental disorders and to guide mutation screening. In the specific case of mendelian disorders of the epigenetic machinery, it also provides the basis to infer mechanistic aspects with regard to DNAme determinants and interplay between histone and DNAme that apply to humans. Here, we present comparative methylomes from patients with mutations in the de novo DNA methyltransferases DNMT3A and DNMT3B, in their catalytic domain or their N-terminal parts involved in reading histone methylation, or in histone H3 lysine (K) methylases NSD1 or SETD2 (H3 K36) or KMT2D/MLL2 (H3 K4). We provide disease-specific DNAme signatures and document the distinct consequences of mutations in enzymes with very similar or intertwined functions, including at repeated sequences and imprinted loci. We found that KMT2D and SETD2 germline mutations have little impact on DNAme profiles. In contrast, the overlapping DNAme alterations downstream of NSD1 or DNMT3 mutations underlines functional links, more specifically between NSD1 and DNMT3B at heterochromatin regions or DNMT3A at regulatory elements. Together, these data indicate certain discrepancy with the mechanisms described in animal models or the existence of redundant or complementary functions unforeseen in humans.


Asunto(s)
Metilación de ADN/genética , Enfermedades Genéticas Congénitas/genética , Histonas/genética , Mutación , Enfermedades Raras/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Enfermedades Genéticas Congénitas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Proteínas de Neoplasias/genética , Enfermedades Raras/metabolismo , ADN Metiltransferasa 3B
17.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669056

RESUMEN

Warsaw breakage syndrome (WABS) is a genetic disorder characterized by sister chromatid cohesion defects, growth retardation, microcephaly, hearing loss and other variable clinical manifestations. WABS is due to biallelic mutations of the gene coding for the super-family 2 DNA helicase DDX11/ChlR1, orthologous to the yeast chromosome loss protein 1 (Chl1). WABS is classified in the group of "cohesinopathies", rare hereditary diseases that are caused by mutations in genes coding for subunits of the cohesin complex or protein factors having regulatory roles in the sister chromatid cohesion process. In fact, among the cohesion regulators, an important player is DDX11, which is believed to be important for the functional coupling of DNA synthesis and cohesion establishment at the replication forks. Here, we will review what is known about the molecular and cellular functions of human DDX11 and its role in WABS etiopathogenesis, even in light of recent findings on the role of cohesin and its regulator network in promoting chromatin loop formation and regulating chromatin spatial organization.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Enfermedades Raras/metabolismo , Anomalías Múltiples/genética , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Cromátides/patología , Cromatina/patología , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica , ARN Helicasas DEAD-box/genética , Replicación del ADN/genética , Regulación de la Expresión Génica/genética , Humanos , Mutación , Filogenia , Enfermedades Raras/congénito , Enfermedades Raras/enzimología , Enfermedades Raras/fisiopatología , Cohesinas
18.
Oncology ; 99(6): 373-379, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33774637

RESUMEN

INTRODUCTION: Extrapulmonary small-cell cancer (EPSCC) is a relatively rare malignancy. The management of EPSCC is usually extrapolated from small-cell lung cancer (SCLC). In spite of the morphological similarity of the 2 malignancies, there are many differences in clinical features, prognosis, and recommendations of treatment of these disorders. The data on the correlation of clinical-pathological characteristics of EPSCC and treatment results is scarce. MATERIALS AND METHODS: This retrospective analysis of 41 consecutively treated patients diagnosed with EPSCC in 2015-2018 was performed in a tertiary medical center. The correlation between the clinical and pathological characteristics and the treatment outcome (response rate, disease-free interval, and overall medial survival) was done using the standard statistics, Kaplan-Meier method, and multivariate analyses. The stratification was done on the stage of the disease, Ki-67 proliferative index, the location of the tumor, and smoking. RESULTS: Forty-one patients were included with a median age of 66.3 years. The most common primary site was the gastrointestinal tract (28, 68.3%) including the pancreas. The most common distant metastasis site was the liver (23, 56.1%). Only 2 patients (4.9%) had brain metastases. Unlike in SCLC, most patients did not have any history of smoking (23, 56.1%). Nineteen patients with metastatic disease received systemic treatment, mostly cisplatin-based chemotherapy, with a response rate of 57.9%. The results of treatment were significantly better in patients with disseminated EPSCC with Ki-67 <55%, while its role in limited disease was nonsignificant. DISCUSSION: The results of our study show the unique entity of EPSCC. The rarity of brain metastases proves that prophylactic brain irradiation should not be recommended in practice. The provocative idea of prophylactic liver irradiation in limited-stage EPSCC of gastrointestinal origin can be evaluated in future studies. The predictive role of Ki-67 is important in metastatic EPSCC. There is probably no role of smoking in developing EPSCC.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/terapia , Carcinoma de Células Pequeñas/terapia , Cisplatino/uso terapéutico , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Tumores Neuroendocrinos/terapia , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/patología , Carcinoma de Células Pequeñas/metabolismo , Carcinoma de Células Pequeñas/patología , Quimioradioterapia , Femenino , Humanos , Estimación de Kaplan-Meier , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Enfermedades Raras/metabolismo , Enfermedades Raras/patología , Enfermedades Raras/terapia , Estudios Retrospectivos , Centros de Atención Terciaria , Resultado del Tratamiento , Adulto Joven
19.
Expert Rev Proteomics ; 18(1): 65-73, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33583303

RESUMEN

OBJECTIVES: Renal amyloidosis (RA) is a rare disease, typically manifested with proteinuria, nephrotic syndrome, and ultimately leads to renal failure. The present study aims to profile the proteomes of renal amyloidosis patient's serum and healthy controls, along with relative quantification to find out robust markers for RA. METHODS: In this study, 12 RA patients and their corresponding age and gender-matched healthy controls were recruited from the Nephrology department of Max Super Specialty Hospital, New Delhi. We employed gel-based proteomic approach coupled with MALDI-TOF MS to compare protein expression patterns in RA patients and controls. Furthermore, validation of differential proteins (selected) was done using bio-layer interferometry. RESULTS: Eleven proteins showed remarkably altered expression levels. Moreover, expression modulation of three proteins (LLPH, SLC25A51, and CHMP2B) was validated which corroborated with two-dimensional gel electrophoresis (2-DE) results showing significant upregulation (p < 0.05) in RA patients followed by ROC analysis which demonstrated the diagnostic potential of these proteins. A protein-protein master network was generated implicating the above identified proteins along with their interactors, fishing out the routes leading to amyloidosis. CONCLUSION: This study indicates that the identified serum proteomic signatures could improve early diagnosis and lead to possible therapeutic targets in RA.


Asunto(s)
Amiloidosis/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Enfermedades Renales/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Nucleares/metabolismo , Proteómica , Proteínas de Unión al ARN/metabolismo , Electroforesis en Gel Bidimensional , Femenino , Humanos , Masculino , Proteoma/análisis , Proteoma/metabolismo , Enfermedades Raras/metabolismo
20.
FEBS J ; 288(5): 1457-1461, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32805742

RESUMEN

In neurodegenerative diseases, a select set of neuron population displays early vulnerability and undergoes progressive degeneration. The heterogeneity of the cerebral cortex and the heterogeneity of patient populations diagnosed with the same disease offer many challenges for developing effective and long-term treatment options. Currently, patients who are considered to have a 'rare' disease are left with no hopes for cure, and many of the neurodegenerative diseases progress fast without any effective solutions. However, as our understanding of disease mechanisms evolve, we begin to realize that the boundaries between diseases are not as sharp as once believed. There are many patients who develop disease due to common underlying causes and mechanisms. As we move forward with drug discovery effort, it becomes obvious that we will have to shift our focus from finding a cure for a disease, to finding solutions to the disease-causing cellular mechanisms so that patients can be treated by mechanism-based strategies. This paradigm shift will lay the foundation for personalized medicine approaches for neurodegenerative disease patients and patients diagnosed with a rare disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Drogas en Investigación/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Medicina de Precisión/tendencias , Enfermedades Raras/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Autofagia/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Reposicionamiento de Medicamentos/métodos , Accesibilidad a los Servicios de Salud/organización & administración , Humanos , Inflamación , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedades Raras/metabolismo , Enfermedades Raras/patología , Respuesta de Proteína Desplegada/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...