Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 387
Filtrar
1.
Adv Exp Med Biol ; 1460: 575-594, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287865

RESUMEN

Lipotoxicity can mediate endothelial dysfunction in obesity. Altered endothelial cell phenotype during the pathobiological course of the lipotoxicity may lead to hemostatic abnormalities, which is a hallmark of several hematological disorders. Impaired hemostasis could also be directly related to numerous metabolic diseases such as hypertension, diabetes, and atherosclerosis. On the other hand, the local hematopoietic bone marrow (BM) renin-angiotensin system (RAS) contributes to the development of atherosclerosis via acting on the lipotoxicity processes. Local BM RAS, principally an autocrine/paracrine/intracrine hematological system, is located at the crossroads of cellular regulation, molecular interactions, and lipotoxicity-mediated vascular endothelial dysfunction. The positive regulatory role of plasma LDL on AT1 receptor-mediated hematopoietic stem cell (HSC) differentiation and the production of pro-atherogenic monocytes have been described. LDL-regulated HSC function may explain in part hypercholesterolemia-induced inflammation as well as the anti-inflammatory and anti-atherosclerotic effects of AT1 receptor blockers. The role of local adipose tissue RAS is directly related to the pathogenesis of metabolic derangements in obesity. There may be a crosstalk between local BM RAS and local adipose tissue RAS at the genomics and transcriptomics levels. This chapter aims to review hematological alterations propagating the pathological influences of lipotoxicity on the vascular endothelium.


Asunto(s)
Enfermedades Hematológicas , Obesidad , Sistema Renina-Angiotensina , Humanos , Obesidad/metabolismo , Obesidad/complicaciones , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/patología , Enfermedades Hematológicas/etiología , Sistema Renina-Angiotensina/fisiología , Animales , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/etiología
2.
Int J Mol Sci ; 25(16)2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39201517

RESUMEN

Hematological and oncological diseases are still among the leading causes of childhood mortality. Expression of growth hormone-releasing hormone (GHRH) and its receptors (GHRH-R) has been previously demonstrated in various human tumors, but very limited findings are available about the presence and potential function of GHRH-Rs in oncological and hematological disorders of children. In this study, we aimed to investigate the expression of mRNA for GHRH and splice variant 1 (SV) of GHRH-R in 15 pediatric hematological/oncological specimens by RT-PCR. The presence and binding characteristics of GHRH-R protein were also studied by Western blot and ligand competition assays. Of the fifteen specimens studied, eleven pediatric samples (73%) showed the expression of mRNA for GHRH. These eleven samples also expressed mRNA for GHRH receptor SV1. GHRH-R protein was found to be expressed in two benign tumor samples and five malignant tumors examined by Western blot. The presence of specific, high affinity binding sites on GHRH-R was demonstrated in all of the seven human pediatric solid tumor samples investigated. Our results show that the expression of GHRH and SV1 of GHRH-R in hemato-oncological diseases in children can pave the way for further investigation of GHRH-Rs as potential molecular targets for diagnosis and therapy.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento , Neoplasias , Receptores de Neuropéptido , Receptores de Hormona Reguladora de Hormona Hipofisaria , Humanos , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Niño , Masculino , Femenino , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Proyectos Piloto , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Preescolar , Adolescente , Neoplasias/genética , Neoplasias/metabolismo , Hungría , Lactante , ARN Mensajero/genética , ARN Mensajero/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Estudios de Cohortes
3.
Int J Mol Sci ; 25(13)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-39000374

RESUMEN

Cell adhesion is a dynamic process that plays a fundamental role in cell proliferation, maintenance, differentiation, and migration. Basal cell adhesion molecule (BCAM), also known as Lutheran (Lu), belongs to the immunoglobulin superfamily of cell adhesion molecules. Lu/BCAM, which is widely expressed in red blood cells, endothelial cells, smooth muscle cells and epithelial cells across various tissues, playing a crucial role in many cellular processes, including cell adhesion, cell motility and cell migration. Moreover, Lu/BCAM, dysregulated in many diseases, such as blood diseases and various types of cancer, may act as a biomarker and target for the treatment of these diseases. This review explores the significance of Lu/BCAM in cell adhesion and its potential as a novel target for treating hematological diseases and tumors.


Asunto(s)
Enfermedades Hematológicas , Neoplasias , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades Hematológicas/metabolismo , Sistema del Grupo Sanguíneo Lutheran/metabolismo , Adhesión Celular , Animales , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular
4.
EBioMedicine ; 104: 105156, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768529

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is a genetic disorder caused by DNA mutations in KMT2D, a lysine methyltransferase that methylates histones and other proteins, and therefore modifies chromatin structure and subsequent gene expression. Ketones, derived from the ketogenic diet, are histone deacetylase inhibitors that can 'open' chromatin and encourage gene expression. Preclinical studies have shown that the ketogenic diet rescues hippocampal memory neurogenesis in mice with KS via the epigenetic effects of ketones. METHODS: Single-cell RNA sequencing and mass spectrometry-based proteomics were used to explore molecular mechanisms of disease in individuals with KS (n = 4) versus controls (n = 4). FINDINGS: Pathway enrichment analysis indicated that loss of function mutations in KMT2D are associated with ribosomal protein dysregulation at an RNA and protein level in individuals with KS (FDR <0.05). Cellular proteomics also identified immune dysregulation and increased abundance of other lysine modification and histone binding proteins, representing a potential compensatory mechanism. A 12-year-old boy with KS, suffering from recurrent episodes of cognitive decline, exhibited improved cognitive function and neuropsychological assessment performance after 12 months on the ketogenic diet, with concomitant improvement in transcriptomic ribosomal protein dysregulation. INTERPRETATION: Our data reveals that lysine methyltransferase deficiency is associated with ribosomal protein dysfunction, with secondary immune dysregulation. Diet and the production of bioactive molecules such as ketone bodies serve as a significant environmental factor that can induce epigenetic changes and improve clinical outcomes. Integrating transcriptomic, proteomic, and clinical data can define mechanisms of disease and treatment effects in individuals with neurodevelopmental disorders. FUNDING: This study was supported by the Dale NHMRC Investigator Grant (APP1193648) (R.D), Petre Foundation (R.D), and The Sydney Children's Hospital Foundation/Kids Research Early and Mid-Career Researcher Grant (E.T).


Asunto(s)
Proteínas de Unión al ADN , Dieta Cetogénica , Cara , Enfermedades Hematológicas , Proteómica , Proteínas Ribosómicas , Enfermedades Vestibulares , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/metabolismo , Enfermedades Vestibulares/dietoterapia , Humanos , Cara/anomalías , Masculino , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/etiología , Enfermedades Hematológicas/dietoterapia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Niño , Proteómica/métodos , Femenino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regulación de la Expresión Génica , Mutación , Transcriptoma , Anomalías Múltiples
5.
Genome Res ; 34(5): 696-710, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38702196

RESUMEN

Many Mendelian developmental disorders caused by coding variants in epigenetic regulators have now been discovered. Epigenetic regulators are broadly expressed, and each of these disorders typically shows phenotypic manifestations from many different organ systems. An open question is whether the chromatin disruption-the root of the pathogenesis-is similar in the different disease-relevant cell types. This is possible in principle, because all these cell types are subject to effects from the same causative gene, which has the same kind of function (e.g., methylates histones) and is disrupted by the same germline variant. We focus on mouse models for Kabuki syndrome types 1 and 2 and find that the chromatin accessibility changes in neurons are mostly distinct from changes in B or T cells. This is not because the neuronal accessibility changes occur at regulatory elements that are only active in neurons. Neurons, but not B or T cells, show preferential chromatin disruption at CpG islands and at regulatory elements linked to aging. A sensitive analysis reveals that regulatory elements disrupted in B/T cells do show chromatin accessibility changes in neurons, but these are very subtle and of uncertain functional significance. Finally, we are able to identify a small set of regulatory elements disrupted in all three cell types. Our findings reveal the cellular-context-specific effect of variants in epigenetic regulators and suggest that blood-derived episignatures, although useful diagnostically, may not be well suited for understanding the mechanistic basis of neurodevelopment in Mendelian disorders of the epigenetic machinery.


Asunto(s)
Anomalías Múltiples , Envejecimiento , Cromatina , Islas de CpG , Cara , Enfermedades Hematológicas , Neuronas , Enfermedades Vestibulares , Animales , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Ratones , Cara/anomalías , Cromatina/metabolismo , Cromatina/genética , Enfermedades Vestibulares/genética , Neuronas/metabolismo , Envejecimiento/genética , Anomalías Múltiples/genética , Modelos Animales de Enfermedad , Epigénesis Genética , Linfocitos T/metabolismo , Linfocitos B/metabolismo
6.
Biomed Pharmacother ; 173: 116334, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38422658

RESUMEN

Neutrophil extracellular traps (NETs) have emerged as a critical factor in malignant hematologic disease pathogenesis. These structures, comprising DNA, histones, and cytoplasmic proteins, were initially recognized for their role in immune defense against microbial threats. Growing evidence suggests that NETs contribute to malignant cell progression and dissemination, representing a double-edged sword. However, there is a paucity of reports on its involvement in hematological disorders. A comprehensive understanding of the intricate relationship between malignant cells and NETs is necessary to explore effective therapeutic strategies. This review highlights NET formation and mechanisms underlying disease pathogenesis. Moreover, we discuss recent advancements in targeted inhibitor development for selective NET disruption, empowering precise design and efficacious therapeutic interventions for malignant hematologic diseases.


Asunto(s)
Trampas Extracelulares , Enfermedades Hematológicas , Neoplasias Hematológicas , Neoplasias , Humanos , Neutrófilos/metabolismo , Histonas/metabolismo , ADN/metabolismo , Neoplasias Hematológicas/metabolismo , Neoplasias/patología , Enfermedades Hematológicas/metabolismo
7.
Exp Hematol ; 128: 10-18, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37816445

RESUMEN

Our dietary choices significantly impact all the cells in our body. Increasing evidence suggests that diet-derived metabolites influence hematopoietic stem cell (HSC) metabolism and function, thereby actively modulating blood homeostasis. This is of particular relevance because regulating the metabolic activity of HSCs is crucial for maintaining stem cell fitness and mitigating the risk of hematologic disorders. In this review, we examine the current scientific knowledge of the impact of diet on stemness features, and we specifically highlight the established mechanisms by which dietary components modulate metabolic and transcriptional programs in adult HSCs. Gaining a deeper understanding of how nutrition influences our HSC compartment may pave the way for targeted dietary interventions with the potential to decelerate aging and improve the effectiveness of transplantation and cancer therapies.


Asunto(s)
Enfermedades Hematológicas , Células Madre Hematopoyéticas , Humanos , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/fisiología , Enfermedades Hematológicas/metabolismo
8.
Exp Hematol ; 127: 8-13, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37647982

RESUMEN

Chronic inflammation, although subtle, puts the body in a constant state of alertness and is associated with many diseases, including cancer and cardiovascular diseases. It leads hematopoietic cells to produce and release proinflammatory cytokines, which trigger specific signaling pathways in hematopoietic stem cells (HSCs) that cause changes in proliferation, differentiation, and migration. This response is essential when HSCs are needed to produce specific blood cells to eliminate an intruder, such as a pathogenic virus, but mutant HSCs can use these proinflammatory signals to their advantage and accelerate the development of hematologic disease or malignancy. Understanding this complex process is vital for monitoring and controlling disease progression in patients. In the 2023 International Society for Experimental Hematology winter webinar, Dr. Eric Pietras (University of Colorado Anschutz Medical Campus, United States) and Dr. Katherine Y. King (Baylor College of Medicine, United States) gave a presentation on this topic, which is summarized in this review article.


Asunto(s)
Enfermedades Hematológicas , Células Madre Hematopoyéticas , Humanos , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Transducción de Señal , Enfermedades Hematológicas/metabolismo , Inflamación/patología
9.
Exp Hematol ; 125-126: 1-5, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37433369

RESUMEN

Aging of hematopoietic stem cells (HSCs) is characterized by lineage bias, increased clonal expansion, and functional decrease. At the molecular level, aged HSCs typically display metabolic dysregulation, upregulation of inflammatory pathways, and downregulation of DNA repair pathways. Cellular aging of HSCs, driven by cell-intrinsic and cell-extrinsic factors, causes a predisposition to anemia, adaptive immune compromise, myelodys, plasia, and malignancy. Most hematologic diseases are strongly associated with age. But what is the biological foundation for decreased fitness with age? And are there therapeutic windows to resolve age-related hematopoietic decline? These questions were the focus of the International Society for Experimental Hematology (ISEH) New Investigator Committee Fall 2022 Webinar. This review touches on the latest insights from two leading laboratories into inflammatory- and niche-driven stem cell aging and includes speculation on strategies to prevent or correct age-related decline in HSC function.


Asunto(s)
Envejecimiento , Enfermedades Hematológicas , Humanos , Anciano , Envejecimiento/patología , Células Madre Hematopoyéticas/metabolismo , Senescencia Celular/genética , Enfermedades Hematológicas/metabolismo
10.
Front Immunol ; 13: 1041010, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36561751

RESUMEN

With the major advances in cancer immunology and immunotherapy, it is critical to consider that most immune cells are short-lived and need to be continuously replenished from hematopoietic stem and progenitor cells. Hematologic abnormalities are prevalent in cancer patients, and many ground-breaking studies over the past decade provide insights into their underlying cellular and molecular mechanisms. Such studies demonstrate that the dysfunction of hematopoiesis is more than a side-effect of cancer pathology, but an important systemic feature of cancer disease. Here we review these many advances, covering the cancer-associated phenotypes of hematopoietic stem and progenitor cells, the dysfunction of myelopoiesis and erythropoiesis, the importance of extramedullary hematopoiesis in cancer disease, and the developmental origins of tumor associated macrophages. We address the roles of many secreted mediators, signaling pathways, and transcriptional and epigenetic mechanisms that mediate such hematopoietic dysfunction. Furthermore, we discuss the important contribution of the hematopoietic dysfunction to cancer immunosuppression, the possible avenues for therapeutic intervention, and highlight the unanswered questions and directions for future work. Overall, hematopoietic dysfunction is established as an active component of the cancer disease mechanisms and an important target for therapeutic intervention.


Asunto(s)
Enfermedades Hematológicas , Neoplasias , Humanos , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis/genética , Neoplasias/metabolismo , Mielopoyesis , Enfermedades Hematológicas/metabolismo , Progresión de la Enfermedad
11.
Biomed Pharmacother ; 153: 113519, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076604

RESUMEN

Secreted protein acidic and rich in cysteine (SPARC), also known as osteonectin or BM-40, is a matricellular protein involved in several biological processes including cell adhesion, growth factor availability, extracellular matrix remodeling and immune-regulation. SPARC has also been associated with a variety of diseases including diabetes, colon cancer, and leukemia. The expression of SPARC in different diseases exhibits some degree of ambiguity, especially in hemopathies. Herein, we review the current expression and effects of SPARC in various hematologic disorders with respect to nanoparticle albumin bound innovative therapies and related diagnostic research, providing a clinical perspective on the use of NAB technology in the frontier treatment of hematologic diseases.


Asunto(s)
Neoplasias Hematológicas , Osteonectina , Albúminas , Adhesión Celular , Matriz Extracelular/metabolismo , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Humanos , Osteonectina/genética , Osteonectina/metabolismo
12.
Biomolecules ; 12(6)2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35740926

RESUMEN

Microvesicles or ectosomes represent a major type of extracellular vesicles that are formed by outward budding of the plasma membrane. Typically, they are bigger than exosomes but smaller than apoptotic vesicles, although they may overlap with both in size and content. Their release by cells is a means to dispose redundant, damaged, or dangerous material; to repair membrane lesions; and, primarily, to mediate intercellular communication. By participating in these vital activities, microvesicles may impact a wide array of cell processes and, consequently, changes in their concentration or components have been associated with several pathologies. Of note, microvesicles released by leukocytes, red blood cells, and platelets, which constitute the vast majority of plasma microvesicles, change under a plethora of diseases affecting not only the hematological, but also the nervous, cardiovascular, and urinary systems, among others. In fact, there is evidence that microvesicles released by blood cells are significant contributors towards pathophysiological states, having inflammatory and/or coagulation and/or immunomodulatory arms, by either promoting or inhibiting the relative disease phenotypes. Consequently, even though microvesicles are typically considered to have adverse links with disease prognosis, progression, or outcomes, not infrequently, they exert protective roles in the affected cells. Based on these functional relations, microvesicles might represent promising disease biomarkers with diagnostic, monitoring, and therapeutic applications, equally to the more thoroughly studied exosomes. In the current review, we provide a summary of the features of microvesicles released by blood cells and their potential implication in hematological and non-hematological diseases.


Asunto(s)
Micropartículas Derivadas de Células , Exosomas , Vesículas Extracelulares , Enfermedades Hematológicas , Plaquetas , Micropartículas Derivadas de Células/metabolismo , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Enfermedades Hematológicas/metabolismo , Humanos
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(4): 374-377, 2022 Apr.
Artículo en Chino | MEDLINE | ID: mdl-35583068

RESUMEN

Neutrophil extracellular traps (NETs) are characterized by a extracellular fibrous network structure produced by neutrophils with DNA and proteins. NETs can be formed by NADPH-dependent NETosis and NADPH-independent NETosis. After formation, it is not only hydrolyzed by DNase in plasma but can also be degraded intracellularly and extracellularly by macrophages and dendritic cells. Recent researches on NETs have reported the increased expression of NETs in a variety of hematological diseases and its immunomodulatory effect on blood system diseases. For example, NETs are closely linked to infections related to leukemia treatment, which affects the treatment and prognosis of leukemia; NETs are also involved in the progression of multiple myeloma, promote the progression of diffuse large B-cell lymphoma, and mediate the thrombotic events of chronic myeloproliferative tumors.


Asunto(s)
Trampas Extracelulares , Enfermedades Hematológicas , Leucemia , Trampas Extracelulares/metabolismo , Enfermedades Hematológicas/metabolismo , Humanos , NADP/metabolismo , Neutrófilos/metabolismo
14.
Front Immunol ; 13: 813676, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250989

RESUMEN

Polygonatum sibiricum Red. has been used as a medicinal herb and nutritional food in traditional Chinese medicine for a long time. It must be processed prior to clinical use for safe and effective applications. However, the present studies mainly focused on crude Polygonatum sibiricum (PS). This study aimed to investigate the chemical properties, blood-enriching effects and mechanism of polysaccharide from the steam-processed Polygonatum sibiricum (SPS), which is a common form of PS in clinical applications. Instrumentation analyses and chemistry analyses revealed the structure of SPS polysaccharide (SPSP). A mice model of blood deficiency syndrome (BDS) was induced by acetylphenylhydrazine (APH) and cyclophosphamide (CTX). Blood routine test, spleen histopathological changes, serum cytokines, etc. were measured. The spleen transcriptome changes of BDS mice were detected by RNA sequencing (RNA-seq). The results showed that SPSP consists predominantly of Gal and GalA together with fewer amounts of Man, Glc, Ara, Rha and GlcN. It could significantly increase peripheral blood cells, restore the splenic trabecular structure, and reverse hematopoietic cytokines to normal levels. RNA-seq analysis showed that 122 differentially expressed genes (DEGs) were obtained after SPSP treatment. GO and KEGG analysis revealed that SPSP-regulated DEGs were mainly involved in hematopoiesis, immune regulation signaling pathways. The reliability of transcriptome profiling was validated by quantitative real-time PCR and Western blot, and the results indicated that the potential molecular mechanisms of the blood-enriching effects of SPSP might be associated with the regulating of JAK1-STAT1 pathway, and elevated the hematopoietic cytokines (EPO, G-CSF, TNF-α and IL-6). This work provides important information on the potential mechanisms of SPSP against BDS.


Asunto(s)
Enfermedades Hematológicas , Polygonatum , Polisacáridos , Animales , Citocinas/metabolismo , Enfermedades Hematológicas/inmunología , Enfermedades Hematológicas/metabolismo , Ratones , Polygonatum/química , Polygonatum/metabolismo , Polisacáridos/metabolismo , Polisacáridos/farmacología , Reproducibilidad de los Resultados , Vapor
15.
Nucleic Acids Res ; 50(D1): D231-D235, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34893873

RESUMEN

The MODOMICS database has been, since 2006, a manually curated and centralized resource, storing and distributing comprehensive information about modified ribonucleosides. Originally, it only contained data on the chemical structures of modified ribonucleosides, their biosynthetic pathways, the location of modified residues in RNA sequences, and RNA-modifying enzymes. Over the years, prompted by the accumulation of new knowledge and new types of data, it has been updated with new information and functionalities. In this new release, we have created a catalog of RNA modifications linked to human diseases, e.g., due to mutations in genes encoding modification enzymes. MODOMICS has been linked extensively to RCSB Protein Data Bank, and sequences of experimentally determined RNA structures with modified residues have been added. This expansion was accompanied by including nucleotide 5'-monophosphate residues. We redesigned the web interface and upgraded the database backend. In addition, a search engine for chemically similar modified residues has been included that can be queried by SMILES codes or by drawing chemical molecules. Finally, previously available datasets of modified residues, biosynthetic pathways, and RNA-modifying enzymes have been updated. Overall, we provide users with a new, enhanced, and restyled tool for research on RNA modification. MODOMICS is available at https://iimcb.genesilico.pl/modomics/.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Enzimas/genética , ARN/genética , Ribonucleósidos/genética , Interfaz Usuario-Computador , Secuencia de Bases , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Gráficos por Computador , Bases de Datos de Proteínas , Conjuntos de Datos como Asunto , Enzimas/metabolismo , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/patología , Humanos , Internet , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Enfermedades Musculoesqueléticas/genética , Enfermedades Musculoesqueléticas/metabolismo , Enfermedades Musculoesqueléticas/patología , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , ARN/metabolismo , Procesamiento Postranscripcional del ARN , Ribonucleósidos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
16.
FASEB J ; 35(11): e21955, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34613626

RESUMEN

Kabuki syndrome (KS) is a rare genetic disorder caused primarily by mutations in the histone modifier genes KMT2D and KDM6A. The genes have broad temporal and spatial expression in many organs, resulting in complex phenotypes observed in KS patients. Hypotonia is one of the clinical presentations associated with KS, yet detailed examination of skeletal muscle samples from KS patients has not been reported. We studied the consequences of loss of KMT2D function in both mouse and human muscles. In mice, heterozygous loss of Kmt2d resulted in reduced neuromuscular junction (NMJ) perimeter, decreased muscle cell differentiation in vitro and impaired myofiber regeneration in vivo. Muscle samples from KS patients of different ages showed presence of increased fibrotic tissue interspersed between myofiber fascicles, which was not seen in mouse muscles. Importantly, when Kmt2d-deficient muscle stem cells were transplanted in vivo in a physiologic non-Kabuki environment, their differentiation potential is restored to levels undistinguishable from control cells. Thus, the epigenetic changes due to loss of function of KMT2D appear reversible through a change in milieu, opening a potential therapeutic avenue.


Asunto(s)
Anomalías Múltiples/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Cara/anomalías , Enfermedades Hematológicas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/genética , Enfermedades Vestibulares/metabolismo , Anomalías Múltiples/genética , Adolescente , Animales , Niño , Preescolar , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Enfermedades Hematológicas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Masculino , Ratones , Ratones Transgénicos , Células Musculares/patología , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Enfermedades Vestibulares/genética
17.
Elife ; 102021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34463256

RESUMEN

Although each Mendelian Disorder of the Epigenetic Machinery (MDEM) has a different causative gene, there are shared disease manifestations. We hypothesize that this phenotypic convergence is a consequence of shared epigenetic alterations. To identify such shared alterations, we interrogate chromatin (ATAC-seq) and expression (RNA-seq) states in B cells from three MDEM mouse models (Kabuki [KS] type 1 and 2 and Rubinstein-Taybi type 1 [RT1] syndromes). We develop a new approach for the overlap analysis and find extensive overlap primarily localized in gene promoters. We show that disruption of chromatin accessibility at promoters often disrupts downstream gene expression, and identify 587 loci and 264 genes with shared disruption across all three MDEMs. Subtle expression alterations of multiple, IgA-relevant genes, collectively contribute to IgA deficiency in KS1 and RT1, but not in KS2. We propose that the joint study of MDEMs offers a principled approach for systematically mapping functional epigenetic variation in mammals.


Asunto(s)
Anomalías Múltiples/genética , Epigénesis Genética/genética , Cara/anomalías , Variación Genética/genética , Enfermedades Hematológicas/genética , Síndrome de Rubinstein-Taybi/genética , Transcriptoma/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/metabolismo , Animales , Cromatina/genética , Modelos Animales de Enfermedad , Femenino , Técnicas Genéticas , Enfermedades Hematológicas/metabolismo , Ratones , Fenotipo , Síndrome de Rubinstein-Taybi/metabolismo , Enfermedades Vestibulares/metabolismo
18.
Blood ; 138(24): 2455-2468, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33945606

RESUMEN

The BCL6 corepressor (BCOR) is a transcription factor involved in the control of embryogenesis, mesenchymal stem cells function, hematopoiesis, and lymphoid development. Recurrent somatic clonal mutations of the BCOR gene and its homolog BCORL1 have been detected in several hematologic malignancies and aplastic anemia. They are scattered across the whole gene length and mostly represent frameshifts (deletions, insertions), nonsense, and missence mutations. These disruptive events lead to the loss of full-length BCOR protein and to the lack or low expression of a truncated form of the protein, both consistent with the tumor suppressor role of BCOR.BCOR and BCORL1 mutations are similar to those causing 2 rare X-linked diseases: oculofaciocardiodental (OFCD) and Shukla-Vernon syndromes, respectively. Here, we focus on the structure and function of normal BCOR and BCORL1 in normal hematopoietic and lymphoid tissues and review the frequency and clinical significance of the mutations of these genes in malignant and nonmalignant hematologic diseases. Moreover, we discuss the importance of mouse models to better understand the role of Bcor loss, alone and combined with alterations of other genes (eg, Dnmt3a and Tet2), in promoting hematologic malignancies and in providing a useful platform for the development of new targeted therapies.


Asunto(s)
Enfermedades Hematológicas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Animales , Regulación Neoplásica de la Expresión Génica , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/patología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Mutación , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/análisis , Proteínas Represoras/metabolismo
19.
Cell Physiol Biochem ; 55(S3): 65-86, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33667332

RESUMEN

The family of two-pore domain potassium (K2P) channels is critically involved in central cellular functions such as ion homeostasis, cell development, and excitability. K2P channels are widely expressed in different human cell types and organs. It is therefore not surprising that aberrant expression and function of K2P channels are related to a spectrum of human diseases, including cancer, autoimmune, CNS, cardiovascular, and urinary tract disorders. Despite homologies in structure, expression, and stimulus, the functional diversity of K2P channels leads to heterogeneous influences on human diseases. The role of individual K2P channels in different disorders depends on expression patterns and modulation in cellular functions. However, an imbalance of potassium homeostasis and action potentials contributes to most disease pathologies. In this review, we provide an overview of current knowledge on the role of K2P channels in human diseases. We look at altered channel expression and function, the potential underlying molecular mechanisms, and prospective research directions in the field of K2P channels.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Gastrointestinales/metabolismo , Enfermedades Hematológicas/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Enfermedades Urológicas/metabolismo , Potenciales de Acción/fisiología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/patología , Expresión Génica , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/patología , Homeostasis/genética , Humanos , Transporte Iónico , Neoplasias/genética , Neoplasias/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Especificidad de Órganos , Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/clasificación , Canales de Potasio de Dominio Poro en Tándem/genética , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Enfermedades Urológicas/genética , Enfermedades Urológicas/patología
20.
PLoS One ; 16(2): e0247489, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33630943

RESUMEN

The gene SON is on human chromosome 21 (21q22.11) and is thought to be associated with hematopoietic disorders that accompany Down syndrome. Additionally, SON is an RNA splicing factor that plays a role in the transcription of leukemia-associated genes. Previously, we showed that mutations in SON cause malformations in human and zebrafish spines and brains during early embryonic development. To examine the role of SON in normal hematopoiesis, we reduced expression of the zebrafish homolog of SON in zebrafish at the single-cell developmental stage with specific morpholinos. In addition to the brain and spinal malformations we also observed abnormal blood cell levels upon son knockdown. We then investigated how blood production was altered when levels of son were reduced. Decreased levels of son resulted in lower amounts of red blood cells when visualized with lcr:GFP transgenic fish. There were also reduced thrombocytes seen with cd41:GFP fish, and myeloid cells when mpx:GFP fish were examined. We also observed a significant decrease in the quantity of T cells, visualized with lck:GFP fish. However, when we examined their hematopoietic stem and progenitor cells (HSPCs), we saw no difference in colony-forming capability. These studies indicate that son is essential for the proper differentiation of the innate and adaptive immune system, and further investigation determining the molecular pathways involved during blood development should elucidate important information about vertebrate HSPC generation, proliferation, and differentiation.


Asunto(s)
Embrión no Mamífero/citología , Hematopoyesis , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente/embriología , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/fisiología , Enfermedades Hematológicas/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Antígenos de Histocompatibilidad Menor/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...