Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
bioRxiv ; 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38895208

RESUMEN

A defined number of hematopoietic stem cell (HSC) clones are born during development and expand to form the pool of adult stem cells. An intricate balance between self-renewal and differentiation of these HSCs supports hematopoiesis for life. HSC fate is determined by complex transcription factor networks that drive cell-type specific gene programs. The transcription factor RUNX1 is required for definitive hematopoiesis, and mutations in Runx1 have been shown to reduce clonal diversity. The RUNX1 cofactor, CBFý, stabilizes RUNX1 binding to DNA, and disruption of their interaction alters downstream gene expression. Chemical screening for modulators of Runx1 and HSC expansion in zebrafish led us to identify a new mechanism for the RUNX1 inhibitor, Ro5-3335. We found that Ro5-3335 increased HSC divisions in zebrafish, and animals transplanted with Ro5-3335 treated cells had enhanced chimerism compared to untreated cells. Using human CD34+ cells, we show that Ro5-3335 remodels the RUNX1 transcription complex by binding to ELF1, independent of CBFý. This allows specific expression of cell cycle and hematopoietic genes that enhance HSC self-renewal and prevent differentiation. Furthermore, we provide the first evidence to show that it is possible to pharmacologically increase the number of stem cell clones in vivo , revealing a previously unknown mechanism for enhancing clonal diversity. Our studies have revealed a mechanism by which binding partners of RUNX1 determine cell fate, with ELF transcription factors guiding cell division. This information could lead to treatments that enhance clonal diversity for blood diseases.

3.
Cell Rep ; 42(5): 112528, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37209097

RESUMEN

Altered hematopoietic stem cell (HSC) fate underlies primary blood disorders but microenvironmental factors controlling this are poorly understood. Genetically barcoded genome editing of synthetic target arrays for lineage tracing (GESTALT) zebrafish were used to screen for factors expressed by the sinusoidal vascular niche that alter the phylogenetic distribution of the HSC pool under native conditions. Dysregulated expression of protein kinase C delta (PKC-δ, encoded by prkcda) increases the number of HSC clones by up to 80% and expands polyclonal populations of immature neutrophil and erythroid precursors. PKC agonists such as cxcl8 augment HSC competition for residency within the niche and expand defined niche populations. CXCL8 induces association of PKC-δ with the focal adhesion complex, activating extracellular signal-regulated kinase (ERK) signaling and expression of niche factors in human endothelial cells. Our findings demonstrate the existence of reserve capacity within the niche that is controlled by CXCL8 and PKC and has significant impact on HSC phylogenetic and phenotypic fate.


Asunto(s)
Células Endoteliales , Pez Cebra , Animales , Humanos , Células Endoteliales/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Filogenia , Proteína Quinasa C-delta/metabolismo , Nicho de Células Madre , Interleucina-8/metabolismo
4.
Trends Cell Biol ; 33(8): 695-707, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36593155

RESUMEN

Acquired genetic or cytogenetic alterations in a blood stem cell that confer clonal fitness promote its relative expansion leading to clonal hematopoiesis (CH). Despite a largely intact hematopoietic output, CH is associated with a heightened risk of progression to hematologic malignancies and with non-hematologic health manifestations, including cardiovascular disease and overall mortality. We focus on the evidence for the role of inflammation in establishing, maintaining and reciprocally being affected by CH. We describe the known pro-inflammatory signals associated with CH and preclinical studies that elucidated the cellular mechanisms involved. We review the evolving literature on early-onset CH in germline predisposition conditions and the possible role of immune dysregulation in this context.


Asunto(s)
Hematopoyesis Clonal , Hematopoyesis , Humanos , Hematopoyesis Clonal/genética , Factores de Riesgo , Mutación , Inflamación
5.
Front Oncol ; 12: 813149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35356204

RESUMEN

Pediatric myelodysplastic syndromes (MDS) often raise concern for an underlying germline predisposition to hematologic malignancies, referred to as germline predisposition herein. With the availability of genetic testing, it is now clear that syndromic features may be lacking in patients with germline predisposition. Many genetic lesions underlying germline predisposition may also be mutated somatically in de novo MDS and leukemias, making it critical to distinguish their germline origin. The verification of a suspected germline predisposition informs therapeutic considerations, guides monitoring pre- and post-treatment, and allows for family counseling. Presentation of MDS due to germline predisposition is not limited to children and spans a wide age range. In fact, the risk of MDS may increase with age in many germline predisposition conditions and can present in adults who lack classical stigmata in their childhood. Furthermore, germline predisposition associated with DDX41 mutations presents with older adult-onset MDS. Although a higher proportion of pediatric patients with MDS will have a germline predisposition, the greater number of MDS diagnoses in adult patients may result in a larger overall number of those with an underlying germline predisposition. In this review, we present a framework for the evaluation of germline predisposition to MDS across all ages. We discuss characteristics of personal and family history, clinical exam and laboratory findings, and integration of genetic sequencing results to assist in the diagnostic evaluation. We address the implications of a diagnosis of germline predisposition for the individual, for their care after MDS therapy, and for family members. Studies on MDS with germline predisposition have provided unique insights into the pathogenesis of hematologic malignancies and mechanisms of somatic genetic rescue vs. disease progression. Increasing recognition in adult patients will inform medical management and may provide potential opportunities for the prevention or interception of malignancy.

6.
J Clin Invest ; 131(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623329

RESUMEN

Germline RUNX1 variants have been identified in relation to myeloid malignancy predisposition, with lymphoid hematological malignancies present at a lower frequency in families. In this issue of the JCI, Li and Yang et al. examined the frequency and type of germline RUNX1 variants in pediatric patients with acute lymphoblastic leukemia (ALL). Patients with T cell ALL (T-ALL) harbored rare, damaging RUNX1 mutations that were not seen in patients with B cell ALL (B-ALL). Further, several of the T-ALL-associated RUNX1 variants had potential dominant-negative activity. RUNX1-mutated T-ALL cases were also associated with somatic JAK3 mutations and enriched for the early T cell precursor (ETP) leukemia subtype, a finding that was validated when RUNX1 and JAK3 mutations were combined in mice. This study confirms germline RUNX1 predisposition beyond myeloid malignancy, demonstrates the importance of examining both germline and somatic mutations in malignancy cohorts, and demarcates the ETP ALL subtype as a flag for germline predisposition in patients.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Niño , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Germinativas , Mutación de Línea Germinal , Humanos , Ratones , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
7.
Blood Adv ; 5(16): 3199-3202, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34424323

RESUMEN

Germline RUNX1 mutations underlie a syndrome, RUNX1-familial platelet disorder (RUNX1-FPD), characterized by bleeding symptoms that result from quantitative and/or qualitative defect in platelets and a significantly increased risk for developing hematologic malignancies. Myeloid neoplasms are the most commonly diagnosed hematologic malignancies, followed by lymphoid malignancies of T-cell origin. Here, we describe the first 2 cases of B-cell acute lymphoblastic leukemia (B-ALL) in patients with confirmed germline RUNX1 mutations. While 1 of the patients had a known diagnosis of RUNX1-FPD with a RUNX1 p.P240Hfs mutation, the other was the index patient of a kindred with a novel RUNX1 variant, RUNX1 c.587C>T (p.T196I), noted on a targeted genetic testing of the B-ALL diagnostic sample. We discuss the clinical course, treatment approaches, and the outcome for the 2 patients. Additionally, we describe transient resolution of the mild thrombocytopenia and bleeding symptoms during therapy, as well as the finding of clonal hematopoiesis with a TET2 mutant clone in 1 of the patients. It is critical to consider testing for germline RUNX1 mutations in patients presenting with B-ALL who have a personal or family history of thrombocytopenia, bleeding symptoms, or RUNX1 variants identified on genetic testing at diagnosis.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Linfocitos B , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Germinativas , Humanos , Mutación
8.
Blood Adv ; 5(13): 2673-2686, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34170284

RESUMEN

Germline heterozygous mutations in GATA2 are associated with a syndrome characterized by cytopenias, atypical infections, and increased risk of hematologic malignancies. Here, we generated a zebrafish mutant of gata2b that recapitulated the myelomonocytopenia and B-cell lymphopenia of GATA2 deficiency syndrome. Using single-cell assay for transposase accessible chromatin with sequencing of marrow cells, we showed that loss of gata2b led to contrasting alterations in chromosome accessibility in early myeloid and lymphoid progenitors, associated with defects in gene expression. Within the myeloid lineage in gata2b mutant zebrafish, we identified an attenuated myeloid differentiation with reduced transcriptional priming and skewing away from the monocytic program. In contrast, in early lymphoid progenitors, gata2b loss led to accumulation of B-lymphoid transcription factor accessibility coupled with increased expression of the B-cell lineage-specification program. However, gata2b mutant zebrafish had incomplete B-cell lymphopoiesis with loss of lineage-specific transcription factor accessibility in differentiating B cells, in the context of aberrantly reduced oxidative metabolic pathways. Our results establish that transcriptional events in early progenitors driven by Gata2 are required to complete normal differentiation.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Deficiencia GATA2 , Animales , Factor de Transcripción GATA2 , Linfopoyesis , Factores de Transcripción/genética , Proteínas de Xenopus , Pez Cebra
9.
Biol Blood Marrow Transplant ; 26(6): 1124-1130, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32088370

RESUMEN

Germline mutations in GATA2 are associated with an inherited predisposition to bone marrow failure (BMF), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). Hematopoietic stem cell transplantation (HSCT) remains the only curative therapy. However, patients may be at an increased risk for transplant-related toxicity (TRT) and transplant-related mortality (TRM) due to their underlying disease biology. We performed a retrospective case-control study of pediatric patients with BMF/MDS/AML with germline GATA2 mutations, comparing HSCT outcomes to randomly selected patients without germline GATA2 mutations and BMF/MDS (control A) and acute leukemia (control B). The 5-year overall and disease-free survival rates in the GATA2 cohort (65%, 51%) were similar to control A (58%, 49%) and B (45%, 43%) cohorts. In contrast, the 5-year event-free survival rate was significantly lower in the GATA2 cohort (7% ± 6%, 28% ± 10%, and 33% ± 8% for GATA2, A, and B, respectively), due to an increased number of unique TRTs. Specifically, neurologic toxicities occurred significantly more frequently in GATA2 patients than in the control groups, and post-HSCT thrombotic events occurred only in the GATA2 cohort. There was no difference in TRM, infections, or graft-versus-host disease across groups. The higher incidence of thrombotic and neurologic events specific to GATA2 patients warrants further investigation and has potential treatment ramifications.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Trastornos de Fallo de la Médula Ósea , Estudios de Casos y Controles , Niño , Factor de Transcripción GATA2/genética , Células Germinativas , Mutación de Línea Germinal , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/terapia , Estudios Retrospectivos
10.
Zebrafish ; 14(4): 379-382, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28557653

RESUMEN

The establishment of in vitro cultures of zebrafish cancer cells has expanded the potential of zebrafish as a disease model. However, the lack of effective methods for gene delivery and genetic manipulation has limited the experimental applications of these cultures. To overcome this barrier, we tested and optimized vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped lentiviral and retroviral vector transduction protocols. We show that lentivirus successfully and efficiently transduced zebrafish melanoma cell lines in vitro, allowing antibiotic selection, fluorescence-based sorting, and in vivo allotransplantation. In addition, injection of concentrated lentiviral particles into embryos and tumors established the feasibility of in vivo gene delivery.


Asunto(s)
Vectores Genéticos/administración & dosificación , Lentivirus/genética , Melanoma/genética , Retroviridae/genética , Transducción Genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Melanoma/patología , Glicoproteínas de Membrana/genética , Células Tumorales Cultivadas , Proteínas del Envoltorio Viral/genética , Pez Cebra/crecimiento & desarrollo
11.
FEBS Lett ; 590(22): 4016-4033, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27616157

RESUMEN

Hematopoietic stem cells (HSCs) give rise to all differentiated blood cells. Understanding the mechanisms that regulate self-renewal and lineage specification of HSCs is key for developing treatments for many human diseases. Zebrafish have emerged as an excellent model for studying vertebrate hematopoiesis. This review will highlight the unique strengths of zebrafish and important findings that have emerged from studies of blood development and disorders using this system. We discuss recent advances in our understanding of hematopoiesis, including the origin of HSCs, molecular control of their development, and key signaling pathways involved in their regulation. We highlight significant findings from zebrafish models of blood disorders and discuss their application for investigating stem cell dysfunction in disease and for the development of new therapeutics.


Asunto(s)
Enfermedades Hematológicas/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Pez Cebra/genética , Animales , Enfermedades Hematológicas/patología , Células Madre Hematopoyéticas/metabolismo , Humanos , Pez Cebra/crecimiento & desarrollo
12.
Mol Cell ; 62(1): 34-46, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058786

RESUMEN

Studying cancer metabolism gives insight into tumorigenic survival mechanisms and susceptibilities. In melanoma, we identify HEXIM1, a transcription elongation regulator, as a melanoma tumor suppressor that responds to nucleotide stress. HEXIM1 expression is low in melanoma. Its overexpression in a zebrafish melanoma model suppresses cancer formation, while its inactivation accelerates tumor onset in vivo. Knockdown of HEXIM1 rescues zebrafish neural crest defects and human melanoma proliferation defects that arise from nucleotide depletion. Under nucleotide stress, HEXIM1 is induced to form an inhibitory complex with P-TEFb, the kinase that initiates transcription elongation, to inhibit elongation at tumorigenic genes. The resulting alteration in gene expression also causes anti-tumorigenic RNAs to bind to and be stabilized by HEXIM1. HEXIM1 plays an important role in inhibiting cancer cell-specific gene transcription while also facilitating anti-cancer gene expression. Our study reveals an important role for HEXIM1 in coupling nucleotide metabolism with transcriptional regulation in melanoma.


Asunto(s)
Melanoma/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Pirimidinas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Melanoma Experimental , Proteínas Oncogénicas/genética , Factores de Transcripción , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Hum Gene Ther ; 27(4): 287-94, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27018965

RESUMEN

Since its introduction in early 1980s, the zebrafish (Danio rerio) has become an invaluable vertebrate animal model system to study many human disorders in almost all systems, from hepatic and brain pathology, to autoimmune and psychiatric disorders. Hematopoiesis between zebrafish and mammals is highly conserved, making the zebrafish an attractive model to study hematopoietic development and blood disorders. Unique attributes of the zebrafish include the ability to perform large-scale genetic and chemical screens in vivo, study development at the cellular level, and use transgenic fish to dissect mechanisms of disease or drug effects. This review summarizes major discoveries that helped define molecular control of hematopoiesis in vertebrates and specific contributions from studies in zebrafish.


Asunto(s)
Sangre/metabolismo , Enfermedades Hematológicas/patología , Hematopoyesis , Pez Cebra/genética , Pez Cebra/fisiología , Animales , Pruebas Genéticas , Humanos , Genética Inversa , Pez Cebra/sangre
14.
Blood ; 124(10): 1622-5, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25075129

RESUMEN

XRCC4-like factor (XLF/Cernunnos) is a component of the nonhomologous end-joining (NHEJ) pathway of double-strand DNA break repair. XLF-deficient patients develop a severe progressive lymphocytopenia. Although NHEJ is required for V(D)J recombination and lymphocyte development, XLF-deficient mice have normal V(D)J recombination, highlighting the need for an alternative mechanism for the lymphocytopenia. Here, we report that XLF-deficient mice recapitulate the age-dependent lymphocytopenia of patients. We show that XLF deficiency leads to premature aging of hematopoietic stem cells (HSCs), measured by decreased functional capacity in transplantation assays, preferential myeloid reconstitution, and reduced self-renewal at a young age. We propose that premature aging of HSCs, together with previously reported defects in class-switch recombination and memory immune response, underlies the progressive and severe lymphocytopenia in XLF-deficient patients in the absence of measurable V(D)J recombination defects.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/fisiología , Linfopenia/genética , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Células Cultivadas , Senescencia Celular/genética , Senescencia Celular/inmunología , Progresión de la Enfermedad , Linfopenia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
J Pediatr Gastroenterol Nutr ; 56(3): e17-20, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22331019
16.
Blood ; 118(23): 6078-86, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21967974

RESUMEN

Hematopoiesis is the process whereby BM HSCs renew to maintain their number or to differentiate into committed progenitors to generate all blood cells. One approach to gain mechanistic insight into this complex process is the investigation of quantitative genetic variation in hematopoietic function among inbred mouse strains. We previously showed that TGF-ß2 is a genetically determined positive regulator of hematopoiesis. In the presence of unknown nonprotein serum factors TGF-ß2, but not TGF-ß1 or -ß3, enhances progenitor proliferation in vitro, an effect that is subject to mouse strain-dependent variation mapping to a locus on chr.4, Tb2r1. TGF-ß2-deficient mice show hematopoietic defects, demonstrating the physiologic role of this cytokine. Here, we show that TGF-ß2 specifically and predominantly cell autonomously enhances signaling by FLT3 in vitro and in vivo. A coding polymorphism in Prdm16 (PR-domain-containing 16) underlies Tb2r1 and differentially regulates transcriptional activity of peroxisome proliferator-activated receptor-γ (PPARγ), identifying lipid PPAR ligands as the serum factors required for regulation of FLT3 signaling by TGF-ß2. We furthermore show that PPARγ agonists play a FLT3-dependent role in stress responses of progenitor cells. These observations identify a novel regulatory axis that includes PPARs, Prdm16, and TGF-ß2 in hematopoiesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , PPAR gamma/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta2/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Células COS , Diferenciación Celular/fisiología , División Celular/fisiología , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , PPAR gamma/agonistas , PPAR gamma/metabolismo , Polimorfismo Genético/fisiología , Sitios de Carácter Cuantitativo/fisiología , Estrés Fisiológico/fisiología , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo
17.
Blood ; 117(19): 5057-66, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21343612

RESUMEN

Fetal liver and adult bone marrow hematopoietic stem cells (HSCs) renew or differentiate into committed progenitors to generate all blood cells. PRDM16 is involved in human leukemic translocations and is expressed highly in some karyotypically normal acute myeloblastic leukemias. As many genes involved in leukemogenic fusions play a role in normal hematopoiesis, we analyzed the role of Prdm16 in the biology of HSCs using Prdm16-deficient mice. We show here that, within the hematopoietic system, Prdm16 is expressed very selectively in the earliest stem and progenitor compartments, and, consistent with this expression pattern, is critical for the establishment and maintenance of the HSC pool during development and after transplantation. Prdm16 deletion enhances apoptosis and cycling of HSCs. Expression analysis revealed that Prdm16 regulates a remarkable number of genes that, based on knockout models, both enhance and suppress HSC function, and affect quiescence, cell cycling, renewal, differentiation, and apoptosis to various extents. These data suggest that Prdm16 may be a critical node in a network that contains negative and positive feedback loops and integrates HSC renewal, quiescence, apoptosis, and differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Factores de Transcripción/metabolismo , Animales , Apoptosis/fisiología , Separación Celular , Proteínas de Unión al ADN/genética , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Genotipo , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética
18.
Methods Enzymol ; 476: 429-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20691879

RESUMEN

Hematopoietic stem cells (HSCs) can self-renew and give rise to all the cells of the blood and the immune system. As they differentiate, HSCs progressively lose their self-renewal capacity and generate lineage-restricted multipotential progenitor cells that in turn give rise to mature cells. The development of rigorous quantitative in vivo assays for HSC activity combined with multicolor flow cytometry and high-speed sorting have resulted in the phenotypic definition of HSCs to virtual purity. Here, we describe the isolation and identification of HSCs by flow cytometry and the use of competitive repopulation to assess HSC number and function.


Asunto(s)
Separación Celular , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Separación Celular/instrumentación , Separación Celular/métodos , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Fenotipo
19.
Science ; 328(5986): 1689-93, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20488992

RESUMEN

Elevated leukocyte cell numbers (leukocytosis), and monocytes in particular, promote atherosclerosis; however, how they become increased is poorly understood. Mice deficient in the adenosine triphosphate-binding cassette (ABC) transporters ABCA1 and ABCG1, which promote cholesterol efflux from macrophages and suppress atherosclerosis in hypercholesterolemic mice, displayed leukocytosis, a transplantable myeloproliferative disorder, and a dramatic expansion of the stem and progenitor cell population containing Lin(-)Sca-1(+)Kit+ (LSK) in the bone marrow. Transplantation of Abca1(-/-) Abcg1(-/-) bone marrow into apolipoprotein A-1 transgenic mice with elevated levels of high-density lipoprotein (HDL) suppressed the LSK population, reduced leukocytosis, reversed the myeloproliferative disorder, and accelerated atherosclerosis. The findings indicate that ABCA1, ABCG1, and HDL inhibit the proliferation of hematopoietic stem and multipotential progenitor cells and connect expansion of these populations with leukocytosis and accelerated atherosclerosis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Aterosclerosis/fisiopatología , Colesterol/metabolismo , Células Madre Hematopoyéticas/fisiología , Leucocitosis/fisiopatología , Lipoproteínas HDL/metabolismo , Lipoproteínas/metabolismo , Células Progenitoras Mieloides/fisiología , Transportador 1 de Casete de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/terapia , Trasplante de Médula Ósea , Proliferación Celular , Células Cultivadas , Hipercolesterolemia/metabolismo , Leucocitosis/metabolismo , Leucocitosis/terapia , Lipoproteínas/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Células Madre Multipotentes/fisiología , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/fisiopatología , Trastornos Mieloproliferativos/terapia , Fenotipo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Interleucina-3/metabolismo , Transducción de Señal
20.
J Gerontol A Biol Sci Med Sci ; 65(6): 620-5, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20371546

RESUMEN

The mechanisms underlying age-associated thymic involution are unknown. In mice, thymic involution shows mouse strain-dependent genetic variation. Identification of the underlying genes would provide mechanistic insight into this elusive process. We previously showed that responsiveness of hematopoietic stem and progenitor cells (HSPCs) to transforming growth factor-beta 2, a positive regulator of HSPC proliferation, is regulated by a quantitative trait locus (QTL) on chr. 4, Tb2r1. Interestingly, Tgfb2(+/-) mice have delayed thymic involution. Therefore, we tested the hypothesis that a QTL on chr. 4 might regulate thymic involution. Aged, but not young, B6.D2-chr.4 congenic mice, where the telomeric region of chr. 4 was introgressed from DBA/2 to C57BL/6 mice, had larger thymi, and better maintenance of early thymic precursors than C57BL/6 control mice. These observations unequivocally demonstrate that the telomeric region of chr. 4 contains a QTL, Ti1 (thymic involution 1) that regulates thymic involution, and suggest the possibility that Ti1 may be identical to Tb2r1.


Asunto(s)
Envejecimiento/fisiología , Cromosomas de los Mamíferos/genética , Sitios de Carácter Cuantitativo/fisiología , Timo/fisiología , Factor de Crecimiento Transformador beta2/fisiología , Animales , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Madre/citología , Timo/anatomía & histología , Timo/citología , Factores de Tiempo , Factor de Crecimiento Transformador beta2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...