Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000374

ABSTRACT

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.


Subject(s)
Hematologic Diseases , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , Hematologic Diseases/metabolism , Lutheran Blood-Group System/metabolism , Cell Adhesion , Animals , Cell Adhesion Molecules/metabolism , Cell Movement
2.
Genome Res ; 34(5): 696-710, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38702196

ABSTRACT

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.


Subject(s)
Abnormalities, Multiple , Aging , Chromatin , CpG Islands , Face , Hematologic Diseases , Neurons , Vestibular Diseases , Animals , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Mice , Face/abnormalities , Chromatin/metabolism , Chromatin/genetics , Vestibular Diseases/genetics , Neurons/metabolism , Aging/genetics , Abnormalities, Multiple/genetics , Disease Models, Animal , Epigenesis, Genetic , T-Lymphocytes/metabolism , B-Lymphocytes/metabolism
3.
EBioMedicine ; 104: 105156, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768529

ABSTRACT

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).


Subject(s)
DNA-Binding Proteins , Diet, Ketogenic , Face , Hematologic Diseases , Proteomics , Ribosomal Proteins , Vestibular Diseases , Vestibular Diseases/genetics , Vestibular Diseases/metabolism , Vestibular Diseases/diet therapy , Humans , Face/abnormalities , Male , Hematologic Diseases/metabolism , Hematologic Diseases/genetics , Hematologic Diseases/etiology , Hematologic Diseases/diet therapy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Child , Proteomics/methods , Female , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Gene Expression Regulation , Mutation , Transcriptome , Abnormalities, Multiple
4.
Biomed Pharmacother ; 173: 116334, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422658

ABSTRACT

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.


Subject(s)
Extracellular Traps , Hematologic Diseases , Hematologic Neoplasms , Neoplasms , Humans , Neutrophils/metabolism , Histones/metabolism , DNA/metabolism , Hematologic Neoplasms/metabolism , Neoplasms/pathology , Hematologic Diseases/metabolism
5.
Exp Hematol ; 128: 10-18, 2023 12.
Article in English | MEDLINE | ID: mdl-37816445

ABSTRACT

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.


Subject(s)
Hematologic Diseases , Hematopoietic Stem Cells , Humans , Hematopoietic Stem Cells/metabolism , Aging/physiology , Hematologic Diseases/metabolism
6.
Exp Hematol ; 127: 8-13, 2023 11.
Article in English | MEDLINE | ID: mdl-37647982

ABSTRACT

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.


Subject(s)
Hematologic Diseases , Hematopoietic Stem Cells , Humans , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Signal Transduction , Hematologic Diseases/metabolism , Inflammation/pathology
7.
Exp Hematol ; 125-126: 1-5, 2023.
Article in English | MEDLINE | ID: mdl-37433369

ABSTRACT

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.


Subject(s)
Aging , Hematologic Diseases , Humans , Aged , Aging/pathology , Hematopoietic Stem Cells/metabolism , Cellular Senescence/genetics , Hematologic Diseases/metabolism
8.
Front Immunol ; 13: 1041010, 2022.
Article in English | MEDLINE | ID: mdl-36561751

ABSTRACT

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.


Subject(s)
Hematologic Diseases , Neoplasms , Humans , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Neoplasms/metabolism , Myelopoiesis , Hematologic Diseases/metabolism , Disease Progression
9.
Biomed Pharmacother ; 153: 113519, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36076604

ABSTRACT

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.


Subject(s)
Hematologic Neoplasms , Osteonectin , Albumins , Cell Adhesion , Extracellular Matrix/metabolism , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Osteonectin/genetics , Osteonectin/metabolism
10.
Biomolecules ; 12(6)2022 06 08.
Article in English | MEDLINE | ID: mdl-35740926

ABSTRACT

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.


Subject(s)
Cell-Derived Microparticles , Exosomes , Extracellular Vesicles , Hematologic Diseases , Blood Platelets , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Extracellular Vesicles/metabolism , Hematologic Diseases/metabolism , Humans
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(4): 374-377, 2022 Apr.
Article in Chinese | MEDLINE | ID: mdl-35583068

ABSTRACT

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.


Subject(s)
Extracellular Traps , Hematologic Diseases , Leukemia , Extracellular Traps/metabolism , Hematologic Diseases/metabolism , Humans , NADP/metabolism , Neutrophils/metabolism
12.
Front Immunol ; 13: 813676, 2022.
Article in English | MEDLINE | ID: mdl-35250989

ABSTRACT

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.


Subject(s)
Hematologic Diseases , Polygonatum , Polysaccharides , Animals , Cytokines/metabolism , Hematologic Diseases/immunology , Hematologic Diseases/metabolism , Mice , Polygonatum/chemistry , Polygonatum/metabolism , Polysaccharides/metabolism , Polysaccharides/pharmacology , Reproducibility of Results , Steam
13.
Nucleic Acids Res ; 50(D1): D231-D235, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34893873

ABSTRACT

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/.


Subject(s)
Databases, Nucleic Acid , Enzymes/genetics , RNA/genetics , Ribonucleosides/genetics , User-Computer Interface , Base Sequence , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Computer Graphics , Databases, Protein , Datasets as Topic , Enzymes/metabolism , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Humans , Internet , Mental Disorders/genetics , Mental Disorders/metabolism , Mental Disorders/pathology , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/metabolism , Musculoskeletal Diseases/pathology , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , RNA/metabolism , RNA Processing, Post-Transcriptional , Ribonucleosides/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
14.
FASEB J ; 35(11): e21955, 2021 11.
Article in English | MEDLINE | ID: mdl-34613626

ABSTRACT

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.


Subject(s)
Abnormalities, Multiple/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/metabolism , Face/abnormalities , Hematologic Diseases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Muscle Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/genetics , Vestibular Diseases/metabolism , Abnormalities, Multiple/genetics , Adolescent , Animals , Child , Child, Preschool , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Hematologic Diseases/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Infant , Male , Mice , Mice, Transgenic , Muscle Cells/pathology , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Vestibular Diseases/genetics
15.
Elife ; 102021 08 31.
Article in English | MEDLINE | ID: mdl-34463256

ABSTRACT

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.


Subject(s)
Abnormalities, Multiple/genetics , Epigenesis, Genetic/genetics , Face/abnormalities , Genetic Variation/genetics , Hematologic Diseases/genetics , Rubinstein-Taybi Syndrome/genetics , Transcriptome/genetics , Vestibular Diseases/genetics , Abnormalities, Multiple/metabolism , Animals , Chromatin/genetics , Disease Models, Animal , Female , Genetic Techniques , Hematologic Diseases/metabolism , Mice , Phenotype , Rubinstein-Taybi Syndrome/metabolism , Vestibular Diseases/metabolism
16.
Blood ; 138(24): 2455-2468, 2021 12 16.
Article in English | MEDLINE | ID: mdl-33945606

ABSTRACT

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.


Subject(s)
Hematologic Diseases/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Animals , Gene Expression Regulation, Neoplastic , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Mutation , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/analysis , Repressor Proteins/metabolism
17.
Cell Physiol Biochem ; 55(S3): 65-86, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33667332

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/metabolism , Cardiovascular Diseases/metabolism , Gastrointestinal Diseases/metabolism , Hematologic Diseases/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Urologic Diseases/metabolism , Action Potentials/physiology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/pathology , Gene Expression , Hematologic Diseases/genetics , Hematologic Diseases/pathology , Homeostasis/genetics , Humans , Ion Transport , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Organ Specificity , Potassium/metabolism , Potassium Channels, Tandem Pore Domain/classification , Potassium Channels, Tandem Pore Domain/genetics , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Urologic Diseases/genetics , Urologic Diseases/pathology
18.
PLoS One ; 16(2): e0247489, 2021.
Article in English | MEDLINE | ID: mdl-33630943

ABSTRACT

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.


Subject(s)
Embryo, Nonmammalian/cytology , Hematopoiesis , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/physiology , Hematologic Diseases/metabolism , Hematopoietic Stem Cells/cytology , Humans , Minor Histocompatibility Antigens/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
19.
Sci Rep ; 11(1): 89, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420217

ABSTRACT

Current models to study the hematopoietic syndrome largely rely on the uniform whole-body exposures. However, in the radio-nuclear accidents or terrorist events, exposure can be non-uniform. The data available on the non-uniform exposures is limited. Thus, we have developed a mice model for studying the hematopoietic syndrome in the non-uniform or partial body exposure scenarios using the localized cobalt60 gamma radiation exposure. Femur region of Strain 'A' male mice was exposed to doses ranging from 7 to 20 Gy. The 30 day survival assay showed 19 Gy as LD100 and 17 Gy as LD50. We measured an array of cytokines and important stem cell markers such as IFN-γ, IL-3, IL-6, GM-CSF, TNF-α, G-CSF, IL-1α, IL-1ß, CD 34 and Sca 1. We found significant changes in IL-6, GM-CSF, TNF-α, G-CSF, and IL-1ß levels compared to untreated groups and amplified levels of CD 34 and Sca 1 positive population in the irradiated mice compared to the untreated controls. Overall, we have developed a mouse model of the hematopoietic acute radiation syndrome that might be useful for understanding of the non-uniform body exposure scenarios. This may also be helpful in the screening of drugs intended for individuals suffering from radiation induced hematopoietic syndrome.


Subject(s)
Acute Radiation Syndrome/etiology , Disease Models, Animal , Hematologic Diseases/etiology , Radiation Exposure/adverse effects , Acute Radiation Syndrome/genetics , Acute Radiation Syndrome/metabolism , Animals , Cobalt Radioisotopes/adverse effects , Cobalt Radioisotopes/chemistry , Cytokines/genetics , Cytokines/metabolism , Femur/metabolism , Femur/radiation effects , Gamma Rays/adverse effects , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Humans , Male , Mice
20.
Asian Pac J Cancer Prev ; 21(10): 2817-2821, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33112535

ABSTRACT

OBJECTIVE: Bone marrow fluid (BMF) consists of various components that establishes a microenvironment for cell differentiation and remodeling. MicroRNA-21 (miR-21) levels have recently emerged as novel biomarkers for different diseases. However, the conventional RNU6B (U6), used as the reference for intracellular miRNA, may not be appropriate for the normalization of circulating miRNAs. METHODS: We measured the levels of U6, spiked-in RNA, and miR-21 in the BMF of 13 healthy controls and 37 patients with hematological disorders to investigate the reliability of either U6 or spike-in RNA as an endogenous reference and also to study the correlation between miR-21, hematological disorders and mortality. RESULTS: Notably, the levels of U6 demonstrated a high variability in BMF of healthy controls and patients. In contrast, the levels of spiked-in RNA displayed a significantly higher stability in both cohorts. Compared with controls, the levels of miR-21 were significantly upregulated in BMF of patients with leukemia but not lymphoma. Also, using 21 as the cut-off value of miR-21, it differentiated the mortality of patients with hematologic disorders. CONCLUSIONS: Collectively, using spiked-in RNA as a reference the upregulated miR-21 levels in BMF could be an indicator of the diagnosis of leukemia and a predictor of mortality.


Subject(s)
Biomarkers, Tumor/genetics , Bone Marrow/metabolism , Hematologic Diseases/diagnosis , Hematologic Diseases/mortality , MicroRNAs/genetics , Aged , Bone Marrow/pathology , Case-Control Studies , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Hematologic Diseases/metabolism , Humans , Male , Middle Aged , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...