Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Nature ; 590(7846): 457-462, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33568812

RESUMO

In contrast to nearly all other tissues, the anatomy of cell differentiation in the bone marrow remains unknown. This is owing to a lack of strategies for examining myelopoiesis-the differentiation of myeloid progenitors into a large variety of innate immune cells-in situ in the bone marrow. Such strategies are required to understand differentiation and lineage-commitment decisions, and to define how spatial organizing cues inform tissue function. Here we develop approaches for imaging myelopoiesis in mice, and generate atlases showing the differentiation of granulocytes, monocytes and dendritic cells. The generation of granulocytes and dendritic cells-monocytes localizes to different blood-vessel structures known as sinusoids, and displays lineage-specific spatial and clonal architectures. Acute systemic infection with Listeria monocytogenes induces lineage-specific progenitor clusters to undergo increased self-renewal of progenitors, but the different lineages remain spatially separated. Monocyte-dendritic cell progenitors (MDPs) map with nonclassical monocytes and conventional dendritic cells; these localize to a subset of blood vessels expressing a major regulator of myelopoiesis, colony-stimulating factor 1 (CSF1, also known as M-CSF)1. Specific deletion of Csf1 in endothelium disrupts the architecture around MDPs and their localization to sinusoids. Subsequently, there are fewer MDPs and their ability to differentiate is reduced, leading to a loss of nonclassical monocytes and dendritic cells during both homeostasis and infection. These data indicate that local cues produced by distinct blood vessels are responsible for the spatial organization of definitive blood cell differentiation.


Assuntos
Rastreamento de Células/métodos , Células Mieloides/citologia , Mielopoese , Coloração e Rotulagem/métodos , Animais , Atlas como Assunto , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Linhagem da Célula , Autorrenovação Celular , Células Dendríticas/citologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Granulócitos/citologia , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Fator Estimulador de Colônias de Macrófagos/deficiência , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Masculino , Camundongos , Monócitos/citologia , Células Mieloides/metabolismo
2.
Genes Dev ; 32(23-24): 1537-1549, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30463901

RESUMO

Human globin gene production transcriptionally "switches" from fetal to adult synthesis shortly after birth and is controlled by macromolecular complexes that enhance or suppress transcription by cis elements scattered throughout the locus. The DRED (direct repeat erythroid-definitive) repressor is recruited to the ε-globin and γ-globin promoters by the orphan nuclear receptors TR2 (NR2C1) and TR4 (NR2C2) to engender their silencing in adult erythroid cells. Here we found that nuclear receptor corepressor-1 (NCoR1) is a critical component of DRED that acts as a scaffold to unite the DNA-binding and epigenetic enzyme components (e.g., DNA methyltransferase 1 [DNMT1] and lysine-specific demethylase 1 [LSD1]) that elicit DRED function. We also describe a potent new regulator of γ-globin repression: The deubiquitinase BRCA1-associated protein-1 (BAP1) is a component of the repressor complex whose activity maintains NCoR1 at sites in the ß-globin locus, and BAP1 inhibition in erythroid cells massively induces γ-globin synthesis. These data provide new mechanistic insights through the discovery of novel epigenetic enzymes that mediate γ-globin gene repression.


Assuntos
Regulação da Expressão Gênica/genética , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , gama-Globinas/genética , Sítios de Ligação , Linhagem Celular , Ativação Enzimática/genética , Epigênese Genética/genética , Células Eritroides/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Células K562 , Membro 1 do Grupo C da Subfamília 2 de Receptores Nucleares/metabolismo , Domínios Proteicos , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo
3.
Development ; 148(17)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34383890

RESUMO

Neural crest cells (NCCs) within the mandibular and maxillary prominences of the first pharyngeal arch are initially competent to respond to signals from either region. However, mechanisms that are only partially understood establish developmental tissue boundaries to ensure spatially correct patterning. In the 'hinge and caps' model of facial development, signals from both ventral prominences (the caps) pattern the adjacent tissues whereas the intervening region, referred to as the maxillomandibular junction (the hinge), maintains separation of the mandibular and maxillary domains. One cap signal is GATA3, a member of the GATA family of zinc-finger transcription factors with a distinct expression pattern in the ventral-most part of the mandibular and maxillary portions of the first arch. Here, we show that disruption of Gata3 in mouse embryos leads to craniofacial microsomia and syngnathia (bony fusion of the upper and lower jaws) that results from changes in BMP4 and FGF8 gene regulatory networks within NCCs near the maxillomandibular junction. GATA3 is thus a crucial component in establishing the network of factors that functionally separate the upper and lower jaws during development.


Assuntos
Padronização Corporal , Face/embriologia , Fator de Transcrição GATA3/metabolismo , Animais , Região Branquial/citologia , Região Branquial/embriologia , Região Branquial/metabolismo , Morte Celular , Proliferação de Células , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Embrião de Mamíferos , Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica no Desenvolvimento , Mandíbula/citologia , Mandíbula/embriologia , Maxila/citologia , Maxila/embriologia , Camundongos , Morfogênese , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo
4.
Blood ; 138(18): 1691-1704, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34324630

RESUMO

Histone H3 lysine 4 methylation (H3K4Me) is most often associated with chromatin activation, and removing H3K4 methyl groups has been shown to be coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a therapeutic target for multiple diseases, including for the potential treatment of ß-globinopathies (sickle cell disease and ß-thalassemia), because it is a component of γ-globin repressor complexes, and LSD1 inactivation leads to robust induction of the fetal globin genes. The effects of LSD1 inhibition in definitive erythropoiesis are not well characterized, so we examined the consequences of conditional inactivation of Lsd1 in adult red blood cells using a new Gata1creERT2 bacterial artificial chromosome transgene. Erythroid-specific loss of Lsd1 activity in mice led to a block in erythroid progenitor differentiation and to the expansion of granulocyte-monocyte progenitor-like cells, converting hematopoietic differentiation potential from an erythroid fate to a myeloid fate. The analogous phenotype was also observed in human hematopoietic stem and progenitor cells, coincident with the induction of myeloid transcription factors (eg, PU.1 and CEBPα). Finally, blocking the activity of the transcription factor PU.1 or RUNX1 at the same time as LSD1 inhibition rescued myeloid lineage conversion to an erythroid phenotype. These data show that LSD1 promotes erythropoiesis by repressing myeloid cell fate in adult erythroid progenitors and that inhibition of the myeloid-differentiation pathway reverses the lineage switch induced by LSD1 inactivation.


Assuntos
Células Eritroides/citologia , Eritropoese , Histona Desmetilases/metabolismo , Células Mieloides/citologia , Animais , Linhagem Celular , Células Cultivadas , Células Eritroides/metabolismo , Deleção de Genes , Histona Desmetilases/genética , Humanos , Camundongos , Células Mieloides/metabolismo
5.
Genes Dev ; 29(18): 1930-41, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26385963

RESUMO

Protein abundance must be precisely regulated throughout life, and nowhere is the stringency of this requirement more evident than during T-cell development: A twofold increase in the abundance of transcription factor GATA3 results in thymic lymphoma, while reduced GATA3 leads to diminished T-cell production. GATA3 haploinsufficiency also causes human HDR (hypoparathyroidism, deafness, and renal dysplasia) syndrome, often accompanied by immunodeficiency. Here we show that loss of one Gata3 allele leads to diminished expansion (and compromised development) of immature T cells as well as aberrant induction of myeloid transcription factor PU.1. This effect is at least in part mediated transcriptionally: We discovered that Gata3 is monoallelically expressed in a parent of origin-independent manner in hematopoietic stem cells and early T-cell progenitors. Curiously, half of the developing cells switch to biallelic Gata3 transcription abruptly at midthymopoiesis. We show that the monoallelic-to-biallelic transcriptional switch is stably maintained and therefore is not a stochastic phenomenon. This unique mechanism, if adopted by other regulatory genes, may provide new biological insights into the rather prevalent phenomenon of monoallelic expression of autosomal genes as well as into the variably penetrant pathophysiological spectrum of phenotypes observed in many human syndromes that are due to haploinsufficiency of the affected gene.


Assuntos
Alelos , Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica/genética , Linfócitos T/metabolismo , Animais , Medula Óssea/metabolismo , Proliferação de Células/genética , Células Cultivadas , Fator de Transcrição GATA3/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/genética , Timócitos/citologia , Timócitos/metabolismo , Transativadores/genética
6.
Development ; 146(21)2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31582413

RESUMO

Mutations in the transcription factor GATA2 cause lymphedema. GATA2 is necessary for the development of lymphatic valves and lymphovenous valves, and for the patterning of lymphatic vessels. Here, we report that GATA2 is not necessary for valvular endothelial cell (VEC) differentiation. Instead, GATA2 is required for VEC maintenance and morphogenesis. GATA2 is also necessary for the expression of the cell junction molecules VE-cadherin and claudin 5 in lymphatic vessels. We identified miR-126 as a target of GATA2, and miR-126-/- embryos recapitulate the phenotypes of mice lacking GATA2. Primary human lymphatic endothelial cells (HLECs) lacking GATA2 (HLECΔGATA2) have altered expression of claudin 5 and VE-cadherin, and blocking miR-126 activity in HLECs phenocopies these changes in expression. Importantly, overexpression of miR-126 in HLECΔGATA2 significantly rescues the cell junction defects. Thus, our work defines a new mechanism of GATA2 activity and uncovers miR-126 as a novel regulator of mammalian lymphatic vascular development.


Assuntos
Células Endoteliais/metabolismo , Fator de Transcrição GATA2/metabolismo , MicroRNAs/metabolismo , Mutação , Angiopoietina-2/metabolismo , Animais , Sistemas CRISPR-Cas , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , Claudina-5/metabolismo , Família de Proteínas EGF/metabolismo , Endotélio Vascular/metabolismo , Feminino , Deleção de Genes , Humanos , Vasos Linfáticos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA-Seq
7.
Genes Cells ; 25(9): 607-614, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32562431

RESUMO

Catecholamine synthesized in the sympathoadrenal system, including sympathetic neurons and adrenal chromaffin cells, is vital for cardiovascular homeostasis. It has been reported that GATA2, a zinc finger transcription factor, is expressed in murine sympathoadrenal progenitor cells. However, a physiological role for GATA2 in adrenal chromaffin cells has not been established. In this study, we demonstrate that GATA2 is specifically expressed in adrenal chromaffin cells. We examined the consequences of Gata2 loss-of-function mutations, exploiting a Gata2 conditional knockout allele crossed to neural crest-specific Wnt1-Cre transgenic mice (Gata2 NC-CKO). The vast majority of Gata2 NC-CKO embryos died by embryonic day 14.5 (e14.5) and exhibited a decrease in catecholamine-producing adrenal chromaffin cells, implying that a potential catecholamine defect might lead to the observed embryonic lethality. When intercrossed pregnant dams were fed with synthetic adrenaline analogs, the lethality of the Gata2 NC-CKO embryos was partially rescued, indicating that placental transfer of the adrenaline analogs complements the lethal catecholamine deficiency in the Gata2 NC-CKO embryos. These results demonstrate that GATA2 participates in the development of neuroendocrine adrenaline biosynthesis, which is essential for fetal survival.


Assuntos
Células Cromafins/metabolismo , Fator de Transcrição GATA2/fisiologia , Glândulas Suprarrenais/anatomia & histologia , Medula Suprarrenal/metabolismo , Animais , Epinefrina/fisiologia , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Genes Letais , Camundongos , Camundongos Transgênicos , Crista Neural
8.
Genes Cells ; 24(8): 534-545, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31141264

RESUMO

Zinc-finger transcription factors GATA2 and GATA3 are both expressed in the developing inner ear, although their overlapping versus distinct activities in adult definitive inner ear are not well understood. We show here that GATA2 and GATA3 are co-expressed in cochlear spiral ganglion cells and redundantly function in the maintenance of spiral ganglion cells and auditory neural circuitry. Notably, Gata2 and Gata3 compound heterozygous mutant mice had a diminished number of spiral ganglion cells due to enhanced apoptosis, which resulted in progressive hearing loss. The decrease in spiral ganglion cellularity was associated with lowered expression of neurotrophin receptor TrkC that is an essential factor for spiral ganglion cell survival. We further show that Gata2 null mutants that additionally bear a Gata2 YAC (yeast artificial chromosome) that counteracts the lethal hematopoietic deficiency due to complete Gata2 loss nonetheless failed to complement the deficiency in neonatal spiral ganglion neurons. Furthermore, cochlea-specific Gata2 deletion mice also had fewer spiral ganglion cells and resultant hearing impairment. These results show that GATA2 and GATA3 redundantly function to maintain spiral ganglion cells and hearing. We propose possible mechanisms underlying hearing loss in human GATA2- or GATA3-related genetic disorders.


Assuntos
Surdez/etiologia , Fatores de Transcrição GATA/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Animais , Apoptose/genética , Contagem de Células , Cóclea/metabolismo , Cóclea/patologia , Surdez/metabolismo , Surdez/fisiopatologia , Modelos Animais de Doenças , Fatores de Transcrição GATA/genética , Expressão Gênica , Genes Reporter , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Gânglio Espiral da Cóclea/patologia
9.
Blood ; 132(12): 1279-1292, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30076146

RESUMO

Notch1 signaling must elevate to high levels in order to drive the proliferation of CD4-CD8- double-negative (DN) thymocytes and progression to the CD4+CD8+ double-positive (DP) stage through ß-selection. During this critical phase of pre-T-cell development, which is also known as the DN-DP transition, it is unclear whether the Notch1 transcriptional complex strengthens its signal output as a discrete unit or through cofactors. We previously showed that the protein inhibitor of activated STAT-like coactivator Zmiz1 is a context-dependent cofactor of Notch1 in T-cell leukemia. We also showed that withdrawal of Zmiz1 generated an early T-lineage progenitor (ETP) defect. Here, we show that this early defect seems inconsistent with loss-of-Notch1 function. In contrast, at the later pre-T-cell stage, withdrawal of Zmiz1 impaired the DN-DP transition by inhibiting proliferation, like withdrawal of Notch. In pre-T cells, but not ETPs, Zmiz1 cooperatively regulated Notch1 target genes Hes1, Lef1, and Myc. Enforced expression of either activated Notch1 or Myc partially rescued the Zmiz1-deficient DN-DP defect. We identified residues in the tetratricopeptide repeat (TPR) domain of Zmiz1 that bind Notch1. Mutating only a single residue impaired the Zmiz1-Notch1 interaction, Myc induction, the DN-DP transition, and leukemic proliferation. Similar effects were seen using a dominant-negative TPR protein. Our studies identify stage-specific roles of Zmiz1. Zmiz1 is a context-specific cofactor for Notch1 during Notch/Myc-dependent thymocyte proliferation, whether normal or malignant. Finally, we highlight a vulnerability in leukemic cells that originated from a developmentally important Zmiz1-Notch1 interaction that is hijacked during transformation from normal pre-T cells.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia de Células T/patologia , Receptor Notch1/metabolismo , Linfócitos T/patologia , Timo/patologia , Animais , Proliferação de Células , Deleção de Genes , Regulação Leucêmica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Camundongos , Modelos Moleculares , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA , Receptor Notch1/genética , Linfócitos T/metabolismo , Timo/metabolismo
10.
Ann Hematol ; 99(9): 2027-2036, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32567028

RESUMO

Increased expression of fetal hemoglobin (HbF) improves the clinical severity of ß-thalassemia patients. EHMT1/2 histone methyltransferases are epigenetic modifying enzymes that are responsible for catalyzing addition of the repressive histone mark H3K9me2 at silenced genes, including the γ-globin genes. UNC0638, a chemical inhibitor of EHMT1/2, has been shown to induce HbF expression in human erythroid progenitor cell cultures. Here, we report the HbF-inducing activity of UNC0638 in erythroid progenitor cells from ß-thalassemia/HbE patients. UNC0638 treatment led to significant increases in γ-globin mRNA, HbF expression, and HbF-containing cells in the absence of significant cytotoxicity. Moreover, UNC0638 showed additive effects on HbF induction in combination with the immunomodulatory drug pomalidomide and the DNMT1 inhibitor decitabine. These studies provide a scientific proof of concept that a small molecule targeting EHMT1/2 epigenetic enzymes, used alone or in combination with pomalidomide or decitabine, is a potential therapeutic approach for HbF induction. Further development of structural analogs of UNC0638 with similar biological effects but improved pharmacokinetic properties may lead to promising therapies and possible clinical application for the treatment of ß-thalassemia.


Assuntos
Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biossíntese , Hemoglobina E/metabolismo , Quinazolinas/farmacologia , Talassemia beta/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Células Precursoras Eritroides/efeitos dos fármacos , Hemoglobina Fetal/genética , Expressão Gênica , Humanos , Talassemia beta/genética
11.
J Immunol ; 201(11): 3465-3470, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30381480

RESUMO

Allelic exclusion is a vital mechanism for the generation of monospecificity to foreign Ags in B and T lymphocytes. In this study, we developed a high-throughput barcoded method to simultaneously analyze the VDJ recombination status of both mouse TCR-ß alleles in hundreds of single cells using next-generation sequencing.


Assuntos
Linfócitos B/imunologia , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Linfócitos T/imunologia , Alelos , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Endogâmicos C57BL , Análise de Célula Única , Recombinação V(D)J
12.
Blood ; 130(23): 2537-2547, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29018082

RESUMO

The orphan nuclear receptors TR4 (NR2C2) and TR2 (NR2C1) are the DNA-binding subunits of the macromolecular complex, direct repeat erythroid-definitive, which has been shown to repress ε- and γ-globin transcription during adult definitive erythropoiesis. Previous studies implied that TR2 and TR4 act largely in a redundant manner during erythroid differentiation; however, during the course of routine genetic studies, we observed multiple variably penetrant phenotypes in the Tr4 mutants, suggesting that indirect effects of the mutation might be masked by multiple modifying genes. To test this hypothesis, Tr4+/- mutant mice were bred into a congenic C57BL/6 background and their phenotypes were reexamined. Surprisingly, we found that homozygous Tr4 null mutant mice expired early during embryogenesis, around embryonic day 7.0, and well before erythropoiesis commences. We further found that Tr4+/- erythroid cells failed to fully differentiate and exhibited diminished proliferative capacity. Analysis of Tr4+/- mutant erythroid cells revealed that reduced TR4 abundance resulted in decreased expression of genes required for heme biosynthesis and erythroid differentiation (Alad and Alas2), but led to significantly increased expression of the proliferation inhibitory factor, cyclin dependent kinase inhibitor (Cdkn1c) These studies support a vital role for TR4 in promoting erythroid maturation and proliferation, and demonstrate that TR4 and TR2 execute distinct, individual functions during embryogenesis and erythroid differentiation.


Assuntos
Diferenciação Celular/genética , Células Eritroides/citologia , Células Eritroides/metabolismo , Membro 2 do Grupo C da Subfamília 2 de Receptores Nucleares/genética , Anemia/sangue , Anemia/genética , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proliferação de Células/genética , Eritropoese/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Heterozigoto , Homozigoto , Linfopoese/genética , Camundongos , Camundongos Knockout , Mutação , Mielopoese/genética , Membro 2 do Grupo C da Subfamília 2 de Receptores Nucleares/metabolismo
13.
Blood ; 130(7): 908-919, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28630119

RESUMO

Chromosomal rearrangements between 3q21 and 3q26 induce inappropriate EVI1 expression by recruiting a GATA2-distal hematopoietic enhancer (G2DHE) to the proximity of the EVI1 gene, leading to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). The acquisition of G2DHE by the EVI1 gene reciprocally deprives this enhancer of 1 of the 2 GATA2 alleles, resulting in a loss-of-function genetic reduction in GATA2 abundance. Because GATA2 haploinsufficiency is strongly associated with MDS and AML, we asked whether EVI1 misexpression and GATA2 haploinsufficiency both contributed to the observed leukemogenesis by using a 3q21q26 mouse model that recapitulates the G2DHE-driven EVI1 misexpression, but in this case, it was coupled to a Gata2 heterozygous germ line deletion. Of note, the Gata2 heterozygous deletion promoted the EVI1-provoked leukemic transformation, resulting in early onset of leukemia. The 3q21q26 mice suffered from leukemia in which B220+ cells and/or Gr1+ leukemic cells occupied their bone marrows. We found that the B220+Gr1-c-Kit+ population contained leukemia-initiating cells and supplied Gr1+ leukemia cells in the 3q21q26 leukemia. When Gata2 expression levels in the B220+Gr1-c-Kit+ cells were decreased as a result of Gata2 heterozygous deletion or spontaneous phenomenon, myeloid differentiation of the B220+Gr1-c-Kit+ cells was suppressed, and the cells acquired induced proliferation as well as B-lymphoid-primed characteristics. Competitive transplantation analysis revealed that Gata2 heterozygous deletion confers selective advantage to EVI1-expressing leukemia cell expansion in recipient mice. These results demonstrate that both the inappropriate stimulation of EVI1 and the loss of 1 allele equivalent of Gata2 expression contribute to the acceleration of leukemogenesis.


Assuntos
Carcinogênese/patologia , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição GATA2/genética , Haploinsuficiência/genética , Leucemia/patologia , Fatores de Transcrição/metabolismo , Alelos , Animais , Carcinogênese/genética , Diferenciação Celular , Proliferação de Células , Cromossomos de Mamíferos/genética , Metabolismo Energético/genética , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Leucemia/genética , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proto-Oncogenes , Estresse Fisiológico/genética
14.
Nucleic Acids Res ; 45(2): 657-671, 2017 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28123038

RESUMO

The first intronic mutations in the intron 1 GATA site (int-1-GATA) of 5-aminolevulinate synthase 2 (ALAS2) have been identified in X-linked sideroblastic anemia (XLSA) pedigrees, strongly suggesting it could be causal mutations of XLSA. However, the function of this int-1-GATA site during in vivo development remains largely unknown. Here, we generated mice lacking a 13 bp fragment, including this int-1-GATA site (T AGATAA: AGCCCC) and found that hemizygous deletion led to an embryonic lethal phenotype due to severe anemia resulting from a lack of ALAS2 expression, indicating that this non-coding sequence is indispensable for ALAS2 expression in vivo Further analyses revealed that this int-1-GATA site anchored the GATA site in intron 8 (int-8-GATA) and the proximal promoter, forming a long-range loop to enhance ALAS2 expression by an enhancer complex including GATA1, TAL1, LMO2, LDB1 and Pol II at least, in erythroid cells. However, compared with the int-8-GATA site, the int-1-GATA site is more essential for regulating ALAS2 expression through CRISPR/Cas9-mediated site-specific deletion. Therefore, the int-1-GATA site could serve as a valuable site for diagnosing XLSA in cases with unknown mutations.


Assuntos
5-Aminolevulinato Sintetase/genética , Sítios de Ligação , Diferenciação Celular , Células Eritroides/citologia , Células Eritroides/metabolismo , Fator de Transcrição GATA1/metabolismo , Íntrons , Anemia Sideroblástica/genética , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Genes Letais , Doenças Genéticas Ligadas ao Cromossomo X/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Hemizigoto , Humanos , Células K562 , Masculino , Mutação , Linhagem , Regiões Promotoras Genéticas , Deleção de Sequência
15.
Proc Natl Acad Sci U S A ; 112(39): 12169-74, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26371321

RESUMO

Sickle cell disease (SCD) is an inherited disorder caused by a point mutation in the ß-globin gene, leading to the production of abnormally shaped red blood cells. Sickle cells are prone to hemolysis and thereby release free heme into plasma, causing oxidative stress and inflammation that in turn result in damage to multiple organs. The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) is a master regulator of the antioxidant cell-defense system. Here we show that constitutive Nrf2 activation by ablation of its negative regulator Keap1 (kelch-like ECH-associated protein 1) significantly improves symptoms in SCD model mice. SCD mice exhibit severe liver damage and lung inflammation associated with high expression levels of proinflammatory cytokines and adhesion molecules compared with normal mice. Importantly, these symptoms subsided after Nrf2 activation. Although hemolysis and stress erythropoiesis did not change substantially in the Nrf2-activated SCD mice, Nrf2 promoted the elimination of plasma heme released by sickle cells' hemolysis and thereby reduced oxidative stress and inflammation, demonstrating that Nrf2 activation reduces organ damage and segregates inflammation from prevention of hemolysis in SCD mice. Furthermore, administration of the Nrf2 inducer CDDO-Im (2-cyano-3, 12 dioxooleana-1, 9 diene-28-imidazolide) also relieved inflammation and organ failure in SCD mice. These results support the contention that Nrf2 induction may be an important means to protect organs from the pathophysiology of sickle cell-induced damage.


Assuntos
Anemia Falciforme/complicações , Inflamação/genética , Inflamação/terapia , Fígado/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/genética , Ativação Transcricional/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Anemia Falciforme/terapia , Animais , Proteínas do Citoesqueleto/genética , Primers do DNA/genética , Citometria de Fluxo , Técnicas de Inativação de Genes , Immunoblotting , Proteína 1 Associada a ECH Semelhante a Kelch , Luciferases , Camundongos , Fator 2 Relacionado a NF-E2/genética , Reação em Cadeia da Polimerase em Tempo Real
16.
Dev Biol ; 409(1): 218-233, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26542011

RESUMO

Lymph is returned to the blood circulation exclusively via four lymphovenous valves (LVVs). Despite their vital importance, the architecture and development of LVVs is poorly understood. We analyzed the formation of LVVs at the molecular and ultrastructural levels during mouse embryogenesis and identified three critical steps. First, LVV-forming endothelial cells (LVV-ECs) differentiate from PROX1(+) progenitors and delaminate from the luminal side of the veins. Second, LVV-ECs aggregate, align perpendicular to the direction of lymph flow and establish lympho-venous connections. Finally, LVVs mature with the recruitment of mural cells. LVV morphogenesis is disrupted in four different mouse models of primary lymphedema and the severity of LVV defects correlate with that of lymphedema. In summary, we have provided the first and the most comprehensive analysis of LVV development. Furthermore, our work suggests that aberrant LVVs contribute to lymphedema.


Assuntos
Vasos Linfáticos/embriologia , Linfedema/embriologia , Linfedema/patologia , Válvulas Venosas/embriologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , Vasos Linfáticos/ultraestrutura , Camundongos Endogâmicos C57BL , Morfogênese , Penetrância , Fenótipo , Válvulas Venosas/ultraestrutura
17.
PLoS Med ; 14(9): e1002382, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28880867

RESUMO

BACKGROUND: Sickle cell disease (SCD), a congenital hemolytic anemia that exacts terrible global morbidity and mortality, is driven by polymerization of mutated sickle hemoglobin (HbS) in red blood cells (RBCs). Fetal hemoglobin (HbF) interferes with this polymerization, but HbF is epigenetically silenced from infancy onward by DNA methyltransferase 1 (DNMT1). METHODS AND FINDINGS: To pharmacologically re-induce HbF by DNMT1 inhibition, this first-in-human clinical trial (NCT01685515) combined 2 small molecules-decitabine to deplete DNMT1 and tetrahydrouridine (THU) to inhibit cytidine deaminase (CDA), the enzyme that otherwise rapidly deaminates/inactivates decitabine, severely limiting its half-life, tissue distribution, and oral bioavailability. Oral decitabine doses, administered after oral THU 10 mg/kg, were escalated from a very low starting level (0.01, 0.02, 0.04, 0.08, or 0.16 mg/kg) to identify minimal doses active in depleting DNMT1 without cytotoxicity. Patients were SCD adults at risk of early death despite standard-of-care, randomized 3:2 to THU-decitabine versus placebo in 5 cohorts of 5 patients treated 2X/week for 8 weeks, with 4 weeks of follow-up. The primary endpoint was ≥ grade 3 non-hematologic toxicity. This endpoint was not triggered, and adverse events (AEs) were not significantly different in THU-decitabine-versus placebo-treated patients. At the decitabine 0.16 mg/kg dose, plasma concentrations peaked at approximately 50 nM (Cmax) and remained elevated for several hours. This dose decreased DNMT1 protein in peripheral blood mononuclear cells by >75% and repetitive element CpG methylation by approximately 10%, and increased HbF by 4%-9% (P < 0.001), doubling fetal hemoglobin-enriched red blood cells (F-cells) up to approximately 80% of total RBCs. Total hemoglobin increased by 1.2-1.9 g/dL (P = 0.01) as reticulocytes simultaneously decreased; that is, better quality and efficiency of HbF-enriched erythropoiesis elevated hemoglobin using fewer reticulocytes. Also indicating better RBC quality, biomarkers of hemolysis, thrombophilia, and inflammation (LDH, bilirubin, D-dimer, C-reactive protein [CRP]) improved. As expected with non-cytotoxic DNMT1-depletion, platelets increased and neutrophils concurrently decreased, but not to an extent requiring treatment holds. As an early phase study, limitations include small patient numbers at each dose level and narrow capacity to evaluate clinical benefits. CONCLUSION: Administration of oral THU-decitabine to patients with SCD was safe in this study and, by targeting DNMT1, upregulated HbF in RBCs. Further studies should investigate clinical benefits and potential harms not identified to date. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01685515.


Assuntos
Anemia Falciforme/tratamento farmacológico , Azacitidina/análogos & derivados , Inibidores Enzimáticos/administração & dosagem , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Tetra-Hidrouridina/administração & dosagem , Adulto , Anemia Falciforme/genética , Azacitidina/administração & dosagem , Azacitidina/farmacologia , Decitabina , Quimioterapia Combinada , Inibidores Enzimáticos/farmacologia , Feminino , Hemoglobina Fetal/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Tetra-Hidrouridina/farmacologia , Resultado do Tratamento , Adulto Jovem
18.
Blood ; 125(9): 1477-87, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25561507

RESUMO

The orphan nuclear receptors TR2 and TR4 have been shown to play key roles in repressing the embryonic and fetal globin genes in erythroid cells. However, combined germline inactivation of Tr2 and Tr4 leads to periimplantation lethal demise in inbred mice. Hence, we have previously been unable to examine the consequences of their dual loss of function in adult definitive erythroid cells. To circumvent this issue, we generated conditional null mutants in both genes and performed gene inactivation in vitro in adult bone marrow cells. Compound Tr2/Tr4 loss of function led to induced expression of the embryonic εy and ßh1 globins (murine counterparts of the human ε- and γ-globin genes). Additionally, TR2/TR4 function is required for terminal erythroid cell maturation. Loss of TR2/TR4 abolished their occupancy on the εy and ßh1 gene promoters, and concurrently impaired co-occupancy by interacting corepressors. These data strongly support the hypothesis that the TR2/TR4 core complex is an adult stage-specific, gene-selective repressor of the embryonic globin genes. Detailed mechanistic understanding of the roles of TR2/TR4 and their cofactors in embryonic and fetal globin gene repression may ultimately enhance the discovery of novel therapeutic agents that can effectively inhibit their transcriptional activity and be safely applied to the treatment of ß-globinopathies.


Assuntos
Embrião de Mamíferos/metabolismo , Células Eritroides/citologia , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Membro 1 do Grupo C da Subfamília 2 de Receptores Nucleares/fisiologia , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/fisiologia , Globinas beta/metabolismo , Animais , Western Blotting , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , Células Eritroides/metabolismo , Citometria de Fluxo , Inativação Gênica , Humanos , Integrases/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Globinas beta/genética
19.
Blood ; 126(3): 386-96, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26031919

RESUMO

Inhibition of lysine-specific demethylase 1 (LSD1) has been shown to induce fetal hemoglobin (HbF) levels in cultured human erythroid cells in vitro. Here we report the in vivo effects of LSD1 inactivation by a selective and more potent inhibitor, RN-1, in a sickle cell disease (SCD) mouse model. Compared with untreated animals, RN-1 administration leads to induced HbF synthesis and to increased frequencies of HbF-positive cells and mature erythrocytes, as well as fewer reticulocytes and sickle cells, in the peripheral blood of treated SCD mice. In keeping with these observations, histologic analyses of the liver and spleen of treated SCD mice verified that they do not exhibit the necrotic lesions that are usually associated with SCD. These data indicate that RN-1 can effectively induce HbF levels in red blood cells and reduce disease pathology in SCD mice, and may therefore offer new therapeutic possibilities for treating SCD.


Assuntos
Anemia Falciforme/prevenção & controle , Hemoglobina Fetal/biossíntese , Histona Desmetilases/antagonistas & inibidores , Rodaminas/farmacologia , Compostos de Espiro/farmacologia , Esplenomegalia/prevenção & controle , Tiofenos/farmacologia , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Hemoglobina Fetal/efeitos dos fármacos , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Camundongos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esplenomegalia/sangue , Esplenomegalia/patologia , Globinas beta/genética , Globinas beta/metabolismo
20.
Adv Exp Med Biol ; 1013: 177-202, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29127681

RESUMO

Reactivation of fetal hemoglobin (HbF) in adult hematopoietic cells has the potential for great clinical benefit in patients bearing deleterious mutations in the ß-globin gene, such as ß-thalassemia and sickle cell disease (SCD), since increasing the production of HbF can compensate for underproduction of ß-globin chains (in ß-thalassemia) and it can also disrupt sickle hemoglobin polymerization (in SCD). Thus for the past few decades, concerted efforts have been made to identify an effective way to induce the synthesis of HbF in adult erythroid cells for potential therapeutic relief from the effects of these ß-globinopathies. Chemical inducers of HbF as well as a number of transcription factors that are able to reactivate HbF synthesis in vitro and in vivo in adult erythroid cells have been identified. However, there has been only limited success in attempts to manipulate either the drugs or regulatory proteins, and in only a fraction of patients, and there is wide variation in individual response to these drugs or transcription factors. These studies highlight the importance for understanding the molecular mechanisms underlying hemoglobin switching so that future studies can be designed to treat these disorders.


Assuntos
Anemia Falciforme/terapia , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Ativação Transcricional , Talassemia beta/terapia , Adulto , Anemia Falciforme/genética , Antidrepanocíticos/farmacologia , Células Eritroides/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Hidroxiureia/farmacologia , Fatores de Transcrição/metabolismo , Globinas beta/genética , Talassemia beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA