Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Hematol ; 99(7): 1220-1229, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38629639

ABSTRACT

Polycythemia vera (PV) is a clonal disorder arising from the acquired somatic mutations of the JAK2 gene, including JAK2V617F or several others in exon 12. A 38-year-old female had a stroke at age 32 and found to have elevated hemoglobin, normal leukocytes, normal platelets, and tested negative for JAK2V617F and exon 12 mutations. Next generation sequencing revealed a novel mutation: JAK2R715T in the pseudokinase domain (JH2) at 47.5%. Its presence in her nail DNA confirmed a germline origin. Her mother and her son similarly had erythrocytosis and a JAK2R715T mutation. Computer modeling indicated gain-of-function JAK2 activity. The propositus and her mother had polyclonal myelopoiesis, ruling out another somatic mutation-derived clonal hematopoiesis. Some erythroid progenitors of all three generations grew without erythropoietin, a hallmark of PV. The in vitro reporter assay confirmed increased activity of the JAK2R715T kinase. Similar to PV, the JAK2R715T native cells have increased STAT5 phosphorylation, augmented transcripts of prothrombotic and inflammatory genes, and decreased KLF2 transcripts. The propositus was not controlled by hydroxyurea, and JAK2 inhibitors were not tolerated; however, Ropeginterferon-alfa-2b (Ropeg-IFN-α) induced a remission. Ropeg-IFN-α treatment also reduced JAK2 activity in the propositus, her mother and JAK2V617F PV subjects. We report dominantly inherited erythrocytosis secondary to a novel germline JAK2R715T gain-of-function mutation with many but not all comparable molecular features to JAK2V617F PV. We also document a previously unreported inhibitory mechanism of JAK2 signaling by Ropeg-IFN-α.


Subject(s)
Germ-Line Mutation , Janus Kinase 2 , Polycythemia , Adult , Female , Humans , Gain of Function Mutation , Interferon-alpha/therapeutic use , Janus Kinase 2/genetics , Pedigree , Polycythemia/genetics , Polycythemia/drug therapy , Polycythemia Vera/genetics , Polycythemia Vera/drug therapy
2.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348919

ABSTRACT

Molecular pathophysiology of Diamond-Blackfan anemia (DBA) involves disrupted erythroid-lineage proliferation, differentiation and apoptosis; with the activation of p53 considered as a key component. Recently, oxidative stress was proposed to play an important role in DBA pathophysiology as well. CRISPR/Cas9-created Rpl5- and Rps19-deficient murine erythroleukemia (MEL) cells and DBA patients' samples were used to evaluate proinflammatory cytokines, oxidative stress, DNA damage and DNA damage response. We demonstrated that the antioxidant defense capacity of Rp-mutant cells is insufficient to meet the greater reactive oxygen species (ROS) production which leads to oxidative DNA damage, cellular senescence and activation of DNA damage response signaling in the developing erythroblasts and altered characteristics of mature erythrocytes. We also showed that the disturbed balance between ROS formation and antioxidant defense is accompanied by the upregulation of proinflammatory cytokines. Finally, the alterations detected in the membrane of DBA erythrocytes may cause their enhanced recognition and destruction by reticuloendothelial macrophages, especially during infections. We propose that the extent of oxidative stress and the ability to activate antioxidant defense systems may contribute to high heterogeneity of clinical symptoms and response to therapy observed in DBA patients.


Subject(s)
Anemia, Diamond-Blackfan/pathology , DNA Damage , Erythrocytes/pathology , Inflammation Mediators/metabolism , Inflammation/pathology , Oxidative Stress , Adult , Anemia, Diamond-Blackfan/immunology , Anemia, Diamond-Blackfan/metabolism , Animals , Case-Control Studies , Child , Erythrocytes/metabolism , Female , Follow-Up Studies , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Middle Aged , Prognosis , Young Adult
3.
Genes (Basel) ; 10(10)2019 10 15.
Article in English | MEDLINE | ID: mdl-31618985

ABSTRACT

Myeloproliferative neoplasms (MPN) are genetically very complex and heterogeneous diseases in which the acquisition of a somatic driver mutation triggers three main myeloid cytokine receptors, and phenotypically expresses as polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The course of the diseases may be influenced by germline predispositions, modifying mutations, their order of acquisition and environmental factors such as aging and inflammation. Deciphering these contributory elements, their mutual interrelationships, and their contribution to MPN pathogenesis brings important insights into the diseases. Animal models (mainly mouse and zebrafish) have already significantly contributed to understanding the role of several acquired and germline mutations in MPN oncogenic signaling. Novel technologies such as induced pluripotent stem cells (iPSCs) and precise genome editing (using CRISPR/Cas9) contribute to the emerging understanding of MPN pathogenesis and clonal architecture, and form a convenient platform for evaluating drug efficacy. In this overview, the genetic landscape of MPN is briefly described, with an attempt to cover the main discoveries of the last 15 years. Mouse and zebrafish models of the driver mutations are discussed and followed by a review of recent progress in modeling MPN with patient-derived iPSCs and CRISPR/Cas9 gene editing.


Subject(s)
Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/physiopathology , Animals , Calreticulin/genetics , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism , Janus Kinase 2/genetics , Mice , Mutation , Neoplasms/genetics , Phenotype , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Receptors, Thrombopoietin/genetics , Signal Transduction , Thrombocythemia, Essential/genetics , Zebrafish
4.
J Cell Mol Med ; 23(11): 7785-7795, 2019 11.
Article in English | MEDLINE | ID: mdl-31517438

ABSTRACT

The patients with mantle cell lymphoma (MCL) have translocation t(11;14) associated with cyclin D1 overexpression. We observed that iron (an essential cofactor of dioxygenases including prolyl hydroxylases [PHDs]) depletion by deferoxamine blocked MCL cells' proliferation, increased expression of DNA damage marker γH2AX, induced cell cycle arrest and decreased cyclin D1 level. Treatment of MCL cell lines with dimethyloxalylglycine, which blocks dioxygenases involving PHDs by competing with their substrate 2-oxoglutarate, leads to their decreased proliferation and the decrease of cyclin D1 level. We then postulated that loss of EGLN2/PHD1 in MCL cells may lead to down-regulation of cyclin D1 by blocking the degradation of FOXO3A, a cyclin D1 suppressor. However, the CRISPR/Cas9-based loss-of-function of EGLN2/PHD1 did not affect cyclin D1 expression and the loss of FOXO3A did not restore cyclin D1 levels after iron chelation. These data suggest that expression of cyclin D1 in MCL is not controlled by ENGL2/PHD1-FOXO3A pathway and that chelation- and 2-oxoglutarate competition-mediated down-regulation of cyclin D1 in MCL cells is driven by yet unknown mechanism involving iron- and 2-oxoglutarate-dependent dioxygenases other than PHD1. These data support further exploration of the use of iron chelation and 2-oxoglutarate-dependent dioxygenase inhibitors as a novel therapy of MCL.


Subject(s)
Cyclin D1/metabolism , Dioxygenases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Iron Chelating Agents/pharmacology , Ketoglutaric Acids/pharmacology , Lymphoma, Mantle-Cell/enzymology , Amino Acids, Dicarboxylic/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , DNA Damage , Deferoxamine/pharmacology , Dioxygenases/metabolism , Down-Regulation/drug effects , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Iron Deficiencies , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Genes (Basel) ; 9(9)2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30200414

ABSTRACT

T-cell factor 4 (TCF4), together with ß-catenin coactivator, functions as the major transcriptional mediator of the canonical wingless/integrated (Wnt) signaling pathway in the intestinal epithelium. The pathway activity is essential for both intestinal homeostasis and tumorigenesis. To date, several mouse models and cellular systems have been used to analyze TCF4 function. However, some findings were conflicting, especially those that were related to the defects observed in the mouse gastrointestinal tract after Tcf4 gene deletion, or to a potential tumor suppressive role of the gene in intestinal cancer cells or tumors. Here, we present the results obtained using a newly generated conditional Tcf4 allele that allows inactivation of all potential Tcf4 isoforms in the mouse tissue or small intestinal and colon organoids. We also employed the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system to disrupt the TCF4 gene in human cells. We showed that in adult mice, epithelial expression of Tcf4 is indispensable for cell proliferation and tumor initiation. However, in human cells, the TCF4 role is redundant with the related T-cell factor 1 (TCF1) and lymphoid enhancer-binding factor 1 (LEF1) transcription factors.

7.
Oncotarget ; 8(7): 11739-11747, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28036300

ABSTRACT

Tibetans existed in high altitude for ~25 thousand years and have evolutionary selected unique haplotypes assumed to be beneficial to hypoxic adaptation. EGLN1/PHD2 and EPAS1/HIF-2α, both crucial components of hypoxia sensing, are the two best-established loci contributing to high altitude adaptation. The co-adapted Tibetan-specific haplotype encoding for PHD2:p.[D4E/C127S] promotes increased HIF degradation under hypoxic conditions. The Tibetan-specific 200 kb EPAS1 haplotype introgressed from an archaic human population related to Denisovans which underwent evolutionary decay; however, the functional variant(s) responsible for high-altitude adaptation at EPAS1/HIF-2α have not yet been identified. Since HIF modulates the behavior of cancer cells, we hypothesized that these Tibetan selected genomic variants may modify cancer risk predisposition. Here, we ascertained the frequencies of EGLN1D4E/C127S and EGLN1C127S variants and ten EPAS1/HIF-2α variants in lung cancer patients and controls in Nepal, whose population consists of people with Indo-Aryan origin and Tibetan-related Mongoloid origin. We observed a significant association between the selected Tibetan EGLN1/PHD2 haplotype and lung cancer (p=0.0012 for D4E, p=0.0002 for C127S), corresponding to a two-fold increase in lung cancer risk. We also observed a two-fold or greater increased risk for two of the ten EPAS1/HIF-2α variants, although the association was not significant after correcting for multiple comparisons (p=0.12). Although these data cannot address the role of these genetic variants on lung cancer initiation or progression, we conclude that some selected Tibetan variants are strongly associated with a modified risk of lung cancer.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Acclimatization , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Tibet
9.
Hum Mutat ; 37(11): 1153-1156, 2016 11.
Article in English | MEDLINE | ID: mdl-27492747

ABSTRACT

We report an infant with sickle cell disease phenotype by biochemical analysis whose ß-globin gene (HBB) sequencing showed sickle cell mutation (HBBS ) heterozygosity. The proband has a unique head-to-tail duplication of the ß-globin gene cluster having wild-type (HBBA ) and HBBS alleles inherited from her father; constituting her HBBS /HBBS -HBBA genotype. Further analyses revealed that proband's duplicated ß-globin gene cluster (∼650 kb) encompassing HBBA does not include the immediate upstream locus control region (LCR) or 3' DNase I hypersensitivity (HS) element. The LCR interacts with ß-globin gene cluster involving long range DNA interactions mediated by various transcription factors to drive the regulation of globin genes expression. However, a low level of HBBA transcript was clearly detected by digital PCR. In this patient, the observed transcription from the duplicated, distally displaced HBBA cluster demonstrates that the loss of LCR and flanking 3'HS sites do not lead to complete silencing of HBB transcription.


Subject(s)
Anemia, Sickle Cell/genetics , Genes, Duplicate , beta-Globins/genetics , 3' Flanking Region , Female , Gene Silencing , Humans , Infant , Locus Control Region , Mutation , Transcription, Genetic
10.
Transl Oncol ; 9(2): 99-107, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27084425

ABSTRACT

Neoplastic growth is frequently associated with genomic DNA methylation that causes transcriptional silencing of tumor suppressor genes. We used a collection of colorectal polyps and carcinomas in combination with bioinformatics analysis of large datasets to study the expression and methylation of Hypermethylated in cancer 1 (HIC1), a tumor suppressor gene inactivated in many neoplasms. In premalignant stages, HIC1 expression was decreased, and the decrease was linked to methylation of a specific region in the HIC1 locus. However, in carcinomas, the HIC1 expression was variable and, in some specimens, comparable to healthy tissue. Importantly, high HIC1 production distinguished a specific type of chemotherapy-responsive tumors.

11.
Pediatr Blood Cancer ; 61(11): 2104-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24729484

ABSTRACT

von Hippel-Lindau (VHL) protein is the principal negative regulator of hypoxia sensing mediated by transcription factors. Mutations in exon 3 of the VHL gene lead to Chuvash (VHL(R200W)) and Croatian (VHL(H191D)) polycythemias. Here, we describe an infant of Bangladesh ethnicity with a novel homozygous VHL(D126N) mutation with congenital polycythemia and dramatically elevated erythropoietin (EPO) levels, who developed severe fatal pulmonary hypertension. In contrast to Chuvash polycythemia, erythroid progenitors (BFU-Es) did not reveal a marked EPO hypersensitivity. Further, NF-E2 and RUNX1 transcripts that correlate with BFU-Es EPO hypersensitivity in polycythemic mutations were not elevated.


Subject(s)
Erythropoietin/blood , Hypertension, Pulmonary/genetics , Mutation, Missense , Polycythemia/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Humans , Infant , Male
12.
Blood ; 123(3): 391-4, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24297870

ABSTRACT

Overexpression of transcription factors runt-related transcription factor 1 (RUNX1) and nuclear factor, erythroid-derived 2 (NF-E2) was reported in granulocytes of patients with polycythemia vera and other myeloproliferative neoplasms (MPNs). Further, a transgenic mouse overexpressing the NF-E2 transgene was reported to be a model of MPN. We hypothesized that increased transcripts of RUNX1 and NF-E2 might characterize other polycythemic states with primary polycythemic features, that is, those with exaggerated erythropoiesis due to augmented erythropoietin (EPO) sensitivity. We tested the expression of RUNX1 and NF-E2 in polycythemic patients of diverse phenotypes and molecular causes. We report that RUNX1 and NF-E2 overexpression is not specific for MPN; these transcripts were also significantly elevated in polycythemias with augmented hypoxia-inducible factor activity whose erythroid progenitors were hypersensitive to EPO. RUNX1 and NF-E2 overexpression was not detected in patients with EPO receptor (EPOR) gain-of-function, suggesting distinct mechanisms by which erythroid progenitors in polycythemias with defects of hypoxia sensing and EPOR mutations exert their EPO hypersensitivity.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloproliferative Disorders/metabolism , NF-E2 Transcription Factor, p45 Subunit/metabolism , Polycythemia/metabolism , Animals , Cell Hypoxia , Core Binding Factor Alpha 2 Subunit/genetics , Erythropoietin/metabolism , Gene Expression Regulation , Granulocytes/cytology , Humans , Leukocytes, Mononuclear/cytology , Mice , Mice, Transgenic , Mutation , Myeloproliferative Disorders/genetics , NF-E2 Transcription Factor, p45 Subunit/genetics , Polycythemia/genetics , Signal Transduction
13.
Stem Cells ; 32(1): 269-78, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24105986

ABSTRACT

Disease-specific induced pluripotent stem cells (iPSCs) provide an unprecedented opportunity to establish novel disease models and accelerate drug development using distinct tissue target cells generated from isogenic iPSC lines with and without disease-causing mutations. To realize the potential of iPSCs in modeling acquired diseases which are usually heterogeneous, we have generated multiple iPSC lines including two lines that are JAK2-wild-type and four lines homozygous for JAK2-V617F somatic mutation from a single polycythemia vera (PV) patient blood. In vitro differentiation of the same patient-derived iPSC lines have demonstrated the differential contributions of their parental hematopoietic clones to the abnormal erythropoiesis including the formation of endogenous erythroid colonies. This iPSC approach thus may provide unique and valuable insights into the genetic events responsible for disease development. To examine the potential of iPSCs in drug testing, we generated isogenic hematopoietic progenitors and erythroblasts from the same iPSC lines derived from PV patients and normal donors. Their response to three clinical JAK inhibitors, INCB018424 (Ruxolitinib), TG101348 (SAR302503), and the more recent CYT387 was evaluated. All three drugs similarly inhibited erythropoiesis from normal and PV iPSC lines containing the wild-type JAK2 genotype, as well as those containing a homozygous or heterozygous JAK2-V617F activating mutation that showed increased erythropoiesis without a JAK inhibitor. However, the JAK inhibitors had less inhibitory effect on the self-renewal of CD34+ hematopoietic progenitors. The iPSC-mediated disease modeling thus underlies the ineffectiveness of the current JAK inhibitors and provides a modeling system to develop better targeted therapies for the JAK2 mutated hematopoiesis.


Subject(s)
Erythroblasts/drug effects , Hematopoietic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Differentiation/drug effects , Erythroblasts/enzymology , Erythropoiesis/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/enzymology , Janus Kinase 2/genetics
14.
Hum Mutat ; 34(10): 1361-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878091

ABSTRACT

We describe the molecular etiology of ß(+)-thalassemia that is caused by the insertion of the full-length transposable element LINE-1 (L1) into the intron-2 of the ß-globin gene (HBB). The transcript level of the affected ß-globin gene was severely reduced. The remaining transcripts consisted of full-length, correctly processed ß-globin mRNA and a minute amount of three aberrantly spliced transcripts with a decreased half-life due to activation of the nonsense-mediated decay pathway. The lower steady-state amount of mRNA produced by the ß-globin(L1) allele also resulted from a reduced rate of transcription and decreased production of full-length ß-globin primary transcripts. The promoter and enhancer sequences of the ß-globin(L1) allele were hypermethylated; however, treatment with a demethylating agent did not restore the impaired transcription. A histone deacetylase inhibitor partially reactivated the ß-globin(L1) transcription despite permanent ß-globin(L1) promoter CpG methylation. This result indicates that the decreased rate of transcription from the ß-globin(L1) allele is associated with an altered chromatin structure. Therefore, the molecular defect caused by intronic L1 insertion in the ß-globin gene represents a novel etiology of ß-thalassemia.


Subject(s)
Introns , Long Interspersed Nucleotide Elements , Mutagenesis, Insertional , beta-Globins/genetics , beta-Thalassemia/genetics , Adult , Alleles , Alternative Splicing , CpG Islands , DNA Methylation , Female , Gene Expression Regulation , Gene Order , Gene Silencing , Humans , Promoter Regions, Genetic , RNA Stability , Transcription, Genetic
16.
Blood ; 121(19): 3918-24, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23538339

ABSTRACT

Germline von Hippel-Lindau (VHL) gene mutations underlie dominantly inherited familial VHL tumor syndrome comprising a predisposition for renal cell carcinoma, pheochromocytoma/paraganglioma, cerebral hemangioblastoma, and endolymphatic sac tumors. However, recessively inherited congenital polycythemia, exemplified by Chuvash polycythemia, has been associated with 2 separate 3' VHL gene mutations in exon 3. It was proposed that different positions of loss-of-function VHL mutations are associated with VHL syndrome cancer predisposition and only C-terminal domain-encoding VHL mutations would cause polycythemia. However, now we describe a new homozygous VHL exon 2 mutation of the VHL gene:(c.413C>T):P138L, which is associated in the affected homozygote with congenital polycythemia but not in her, or her-heterozygous relatives, with cancer or other VHL syndrome tumors. We show that VHL(P138L) has perturbed interaction with hypoxia-inducible transcription factor (HIF)1α. Further, VHL(P138L) protein has decreased stability in vitro. Similarly to what was reported in Chuvash polycythemia and some other instances of HIFs upregulation, VHL(P138L) erythroid progenitors are hypersensitive to erythropoietin. Interestingly, the level of RUNX1/AML1 and NF-E2 transcripts that are specifically upregulated in acquired polycythemia vera were also upregulated in VHL(P138L) granulocytes.


Subject(s)
Mutation , Polycythemia/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adolescent , Exons/genetics , Female , Homozygote , Humans , Models, Molecular , Mutation/physiology , Neoplasms/genetics , Polycythemia/congenital , Protein Structure, Secondary , Von Hippel-Lindau Tumor Suppressor Protein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...