Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Genet ; 20(7): 377-388, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30737492

RESUMEN

The derivation of induced pluripotent stem cells (iPSCs) over a decade ago sparked widespread enthusiasm for the development of new models of human disease, enhanced platforms for drug discovery and more widespread use of autologous cell-based therapy. Early studies using directed differentiation of iPSCs frequently uncovered cell-level phenotypes in monogenic diseases, but translation to tissue-level and organ-level diseases has required development of more complex, 3D, multicellular systems. Organoids and human-rodent chimaeras more accurately mirror the diverse cellular ecosystems of complex tissues and are being applied to iPSC disease models to recapitulate the pathobiology of a broad spectrum of human maladies, including infectious diseases, genetic disorders and cancer.


Asunto(s)
Enfermedades Transmisibles/terapia , Enfermedades Genéticas Congénitas/terapia , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Neoplasias/terapia , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Quimera/genética , Quimera/inmunología , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/patología , Descubrimiento de Drogas/métodos , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Enfermedades Genéticas Congénitas/patología , Terapia Genética/métodos , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/trasplante , Modelos Animales , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Organoides/citología , Organoides/efectos de los fármacos , Organoides/inmunología , Trasplante de Tejidos/métodos , Trasplante Heterólogo
2.
Nature ; 545(7655): 432-438, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514439

RESUMEN

A variety of tissue lineages can be differentiated from pluripotent stem cells by mimicking embryonic development through stepwise exposure to morphogens, or by conversion of one differentiated cell type into another by enforced expression of master transcription factors. Here, to yield functional human haematopoietic stem cells, we perform morphogen-directed differentiation of human pluripotent stem cells into haemogenic endothelium followed by screening of 26 candidate haematopoietic stem-cell-specifying transcription factors for their capacity to promote multi-lineage haematopoietic engraftment in mouse hosts. We recover seven transcription factors (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1 and SPI1) that are sufficient to convert haemogenic endothelium into haematopoietic stem and progenitor cells that engraft myeloid, B and T cells in primary and secondary mouse recipients. Our combined approach of morphogen-driven differentiation and transcription-factor-mediated cell fate conversion produces haematopoietic stem and progenitor cells from pluripotent stem cells and holds promise for modelling haematopoietic disease in humanized mice and for therapeutic strategies in genetic blood disorders.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Animales , Reprogramación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Endotelio/citología , Femenino , Trasplante de Células Madre Hematopoyéticas , Proteínas Homeobox A10 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Regulador Transcripcional ERG/metabolismo
3.
Annu Rev Cell Dev Biol ; 25: 567-95, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19575644

RESUMEN

A seminal event in cancer progression is the ability of the neoplastic cell to mobilize the necessary machinery to breach surrounding extracellular matrix barriers while orchestrating a host stromal response that ultimately supports tissue-invasive and metastatic processes. With over 500 proteolytic enzymes identified in the human genome, interconnecting webs of protease-dependent and protease-independent processes have been postulated to drive the cancer cell invasion program via schemes of daunting complexity. Increasingly, however, a body of evidence has begun to emerge that supports a unifying model wherein a small group of membrane-tethered enzymes, termed the membrane-type matrix metalloproteinases (MT-MMPs), plays a dominant role in regulating cancer cell, as well as stromal cell, traffic through the extracellular matrix barriers assembled by host tissues in vivo. Understanding the mechanisms that underlie the regulation and function of these metalloenzymes as host cell populations traverse the dynamic extracellular matrix assembled during neoplastic states should provide new and testable theories regarding cancer invasion and metastasis.


Asunto(s)
Matriz Extracelular/patología , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Neoplasias/patología , Animales , Membrana Basal/patología , Humanos , Células del Estroma/patología
4.
Proc Natl Acad Sci U S A ; 117(9): 4653-4663, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32060122

RESUMEN

The LIN28:pre-let-7:TUTase ternary complex regulates pluripotency and oncogenesis by controlling processing of the let-7 family of microRNAs. The complex oligouridylates the 3' ends of pre-let-7 molecules, leading to their degradation via the DIS3L2 exonuclease. Previous studies suggest that components of this complex are potential therapeutic targets in malignancies that aberrantly express LIN28. In this study we developed a functional epitope selection approach to identify nanobody inhibitors of the LIN28:pre-let-7:TUT4 complex. We demonstrate that one of the identified nanobodies, Nb-S2A4, targets the 106-residue LIN28:let-7 interaction (LLI) fragment of TUT4. Nb-S2A4 can effectively inhibit oligouridylation and monouridylation of pre-let-7g in vitro. Expressing Nb-S2A4 allows maturation of the let-7 species in cells expressing LIN28, highlighting the therapeutic potential of targeting the LLI fragment.


Asunto(s)
Proteínas de Unión al ADN/inmunología , MicroARNs/metabolismo , Procesamiento de Término de ARN 3' , Anticuerpos de Dominio Único/inmunología , Animales , Sitios de Unión , Proteínas de Unión al ADN/química , Células HEK293 , Células HeLa , Humanos , Ratones , MicroARNs/genética , Unión Proteica , Estabilidad del ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células Sf9 , Spodoptera
5.
Development ; 143(21): 3956-3968, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27633994

RESUMEN

During late embryogenesis, mammary epithelial cells initiate migration programs that drive ductal invasion into the surrounding adipose-rich mesenchyme. Currently, branching morphogenesis is thought to depend on the mobilization of the membrane-anchored matrix metalloproteinases MMP14 (MT1-MMP) and MMP15 (MT2-MMP), which drive epithelial cell invasion by remodeling the extracellular matrix and triggering associated signaling cascades. However, the roles that these proteinases play during mammary gland development in vivo remain undefined. Here, we characterize the impact of global Mmp14 and Mmp15 targeting on early postnatal mammary gland development in mice. Unexpectedly, both Mmp14-/- and Mmp15-/- mammary glands retain the ability to generate intact ductal networks. Although neither proteinase is required for branching morphogenesis, transcriptome profiling reveals a key role for MMP14 and MMP15 in regulating mammary gland adipocyte differentiation. Whereas MMP14 promotes the generation of white fat depots crucial for energy storage, MMP15 differentially controls the formation of thermogenic brown fat. Taken together, these data not only indicate that current paradigms relevant to proteinase-dependent morphogenesis need be revisited, but also identify new roles for the enzymes in regulating adipocyte fate determination in the developing mammary gland.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Metaloproteinasa 14 de la Matriz/fisiología , Metaloproteinasa 15 de la Matriz/fisiología , Morfogénesis/genética , Adipocitos/fisiología , Adipogénesis/genética , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Metabolismo Energético/genética , Femenino , Metaloproteinasa 14 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Termogénesis/genética
6.
Pediatr Blood Cancer ; 65(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29251406

RESUMEN

BACKGROUND: In osteosarcoma, patient survival has not changed in over 30 years. Multiple phase II trials have been conducted in osteosarcoma using the Response Evaluation Criteria in Solid Tumors (RECIST) as a primary endpoint; however, none of these have revealed new treatment strategies. We investigated RECIST in newly diagnosed patients who received neoadjuvant chemotherapy proven to be beneficial. METHODS: Patients treated from 1986 to 2011 for newly diagnosed osteosarcoma with paired tumor imaging before and after adequate neoadjuvant chemotherapy were included in this retrospective study. Two radiologists performed independent, blinded (to image timing) RECIST measurements of primary tumor and lung metastases at diagnosis and post-neoadjuvant chemotherapy. Association between RECIST and histological necrosis and outcome were assessed. RESULTS: Seventy-four patients met inclusion criteria. Five-year overall survival and progression-free survival (PFS) were 77 ± 7% and 61 ± 8%, respectively. No patients had RECIST partial or complete response in the primary tumor. Sixty-four patients (86%) had stable disease, and 10 (14%) had progressive disease (PD). PD in the primary tumor was associated with significantly worse PFS in localized disease patients (P = 0.02). There was no association between RECIST in the primary tumor and necrosis. There were an insufficient number of patients with lung nodules ≥1 cm at diagnosis to evaluate RECIST in pulmonary metastases. CONCLUSIONS: PD by RECIST predicts poor outcome in localized disease patients. In bone lesions, chemotherapy proven to improve overall survival does not result in radiographic responses as measured by RECIST. Further investigation of RECIST in pulmonary metastatic disease in osteosarcoma is needed.


Asunto(s)
Neoplasias Óseas , Neoplasias Pulmonares , Terapia Neoadyuvante , Osteosarcoma , Adolescente , Adulto , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Metástasis de la Neoplasia , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/mortalidad , Osteosarcoma/patología , Estudios Retrospectivos , Tasa de Supervivencia
7.
Nature ; 541(7636): 166-167, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-27974805
8.
Biol Blood Marrow Transplant ; 22(7): 1275-1283, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27090959

RESUMEN

Cytomegalovirus (CMV) infection is a significant source of morbidity and mortality in allogeneic stem cell transplantation (SCT). We identified a cohort of 91 pediatric SCT patients at risk (defined as either donor and/or recipient seropositivity) for CMV infection at our institution. We retrospectively categorized at-risk SCT recipients as those who (1) were at risk of CMV infection in the post-SCT period, (2) had documented CMV infection before SCT, (3) experienced recurrence of post-SCT CMV viremia, or (4) experienced late post-SCT CMV viremia; categories were not mutually exclusive. We analyzed the impact of SCT-related factors on incidence of CMV infection and outcome, and we described the outcome of each of these cohorts. In univariate analysis, recipient CMV seropositivity, use of umbilical cord blood graft, and acute graft-versus-host disease (GVHD) predicted post-SCT CMV viremia, and the effects of acute GVHD (odds ratio, 4.018; 95% confidence interval, 1.032 to 15.643) and CMV seropositivity (odds ratio, 16.525; 95% confidence interval, 2.041 to 133.803) were confirmed in multivariate analysis. Patients with recurrence of post-SCT CMV viremia had a 50% all-cause mortality rate, compared with 12% in all 91 patients. Patients with pre-SCT CMV infection had a high incidence of post-SCT CMV infection but could successfully undergo SCT with antiviral prophylaxis and pre-emptive CMV treatment. All patients with late CMV infection had prior GVHD. Theses findings identify risk factors for post-SCT CMV infection and provide novel descriptions of childhood SCT recipients with pre-SCT, recurrent, and late CMV infection, which may contribute to risk stratification strategies for CMV at-risk patients in pediatric allogeneic SCT.


Asunto(s)
Infecciones por Citomegalovirus/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adolescente , Adulto , Antivirales/uso terapéutico , Niño , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Infecciones por Citomegalovirus/prevención & control , Femenino , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Lactante , Masculino , Premedicación/métodos , Recurrencia , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Viremia/etiología , Viremia/prevención & control , Adulto Joven
9.
Pediatr Blood Cancer ; 63(5): 928-30, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26713672

RESUMEN

We present an 8-year-old male with metastatic alveolar rhabdomyosarcoma (ARMS) who developed precipitous cardiopulmonary collapse with severe tumor lysis syndrome (TLS) 48 hr after initiation of chemotherapy. Despite no detectable pulmonary metastases, acute hypoxemic respiratory failure developed, requiring extracorporeal membrane oxygenation (ECMO). Although TLS has been reported in disseminated ARMS, this singular case of life-threatening respiratory deterioration developing after initiation of chemotherapy presented unique therapeutic dilemmas. We review the clinical aspects of this case, including possible mechanisms of respiratory failure, and discuss the role of ECMO utilization in pediatric oncology.


Asunto(s)
Oxigenación por Membrana Extracorpórea , Edema Pulmonar , Rabdomiosarcoma Alveolar/tratamiento farmacológico , Síndrome de Lisis Tumoral , Enfermedad Aguda , Niño , Humanos , Masculino , Metástasis de la Neoplasia , Edema Pulmonar/etiología , Edema Pulmonar/patología , Edema Pulmonar/terapia , Rabdomiosarcoma Alveolar/patología , Síndrome de Lisis Tumoral/etiología , Síndrome de Lisis Tumoral/patología , Síndrome de Lisis Tumoral/terapia
10.
Leukemia ; 38(3): 521-529, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38245602

RESUMEN

Constitutional trisomy 21 (T21) is a state of aneuploidy associated with high incidence of childhood acute myeloid leukemia (AML). T21-associated AML is preceded by transient abnormal myelopoiesis (TAM), which is triggered by truncating mutations in GATA1 generating a short GATA1 isoform (GATA1s). T21-associated AML emerges due to secondary mutations in hematopoietic clones bearing GATA1s. Since aneuploidy generally impairs cellular fitness, the paradoxically elevated risk of myeloid malignancy in T21 is not fully understood. We hypothesized that individuals with T21 bear inherent genome instability in hematopoietic lineages that promotes leukemogenic mutations driving the genesis of TAM and AML. We found that individuals with T21 show increased chromosomal copy number variations (CNVs) compared to euploid individuals, suggesting that genome instability could be underlying predisposition to TAM and AML. Acquisition of GATA1s enforces myeloid skewing and maintenance of the hematopoietic progenitor state independently of T21; however, GATA1s in T21 hematopoietic progenitor cells (HPCs) further augments genome instability. Increased dosage of the chromosome 21 (chr21) gene DYRK1A impairs homology-directed DNA repair as a mechanism of elevated mutagenesis. These results posit a model wherein inherent genome instability in T21 drives myeloid malignancy in concert with GATA1s mutations.


Asunto(s)
Síndrome de Down , Leucemia Mieloide Aguda , Reacción Leucemoide , Trastornos Mieloproliferativos , Humanos , Niño , Síndrome de Down/complicaciones , Variaciones en el Número de Copia de ADN , Trastornos Mieloproliferativos/genética , Inestabilidad Genómica , Leucemia Mieloide Aguda/patología , Aneuploidia , Trisomía , Factor de Transcripción GATA1/genética
11.
Am J Pathol ; 180(5): 1863-78, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22464947

RESUMEN

The myocardial extracellular matrix (ECM), an interwoven meshwork of proteins, glycoproteins, proteoglycans, and glycosaminoglycans that is dominated by polymeric fibrils of type I collagen, serves as the mechanical scaffold on which myocytes are arrayed for coordinated and synergistic force transduction. Following ischemic injury, cardiac ECM remodeling is initiated via localized proteolysis, the bulk of which has been assigned to matrix metalloproteinase (MMP) family members. Nevertheless, the key effector(s) of myocardial type I collagenolysis both in vitro and in vivo have remained unidentified. In this study, using cardiac explants from mice deficient in each of the major type I collagenolytic MMPs, including MMP-13, MMP-8, MMP-2, MMP-9, or MT1-MMP, we identify the membrane-anchored MMP, MT1-MMP, as the dominant collagenase that is operative within myocardial tissues in vitro. Extending these observations to an in vivo setting, mice heterozygous for an MT1-MMP-null allele display a distinct survival advantage and retain myocardial function relative to wild-type littermates in an experimental model of myocardial infarction, effects associated with preservation of the myocardial type I collagen network as a consequence of the decreased collagenolytic potential of cardiac fibroblasts. This study identifies MT1-MMP as a key MMP responsible for effecting postinfarction cardiac ECM remodeling and cardiac dysfunction.


Asunto(s)
Matriz Extracelular/enzimología , Metaloproteinasa 14 de la Matriz/fisiología , Infarto del Miocardio/enzimología , Remodelación Ventricular/fisiología , Animales , Colágeno Tipo I/metabolismo , Matriz Extracelular/fisiología , Femenino , Fibroblastos/enzimología , Hibridación in Situ , Metaloproteinasa 14 de la Matriz/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/metabolismo , Técnicas de Cultivo de Órganos , Ultrasonografía
12.
Res Sq ; 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37886538

RESUMEN

Abundant macrophage infiltration and altered tumor metabolism are two key hallmarks of glioblastoma. By screening a cluster of metabolic small-molecule compounds, we show that inhibiting glioblastoma cell glycolysis impairs macrophage migration and lactate dehydrogenase (LDH) inhibitor stiripentol (an FDA-approved anti-seizure drug for Dravet Syndrome) emerges as the top hit. Combined profiling and functional studies demonstrate that LDHA-directed ERK pathway activates YAP1/STAT3 transcriptional co-activators in glioblastoma cells to upregulate CCL2 and CCL7, which recruit macrophages into the tumor microenvironment. Reciprocally, infiltrating macrophages produce LDHA-containing extracellular vesicles to promote glioblastoma cell glycolysis, proliferation, and survival. Genetic and pharmacological inhibition of LDHA-mediated tumor-macrophage symbiosis markedly suppresses tumor progression and macrophage infiltration in glioblastoma mouse models. Analysis of tumor and plasma samples of glioblastoma patients confirms that LDHA and its downstream signals are potential biomarkers correlating positively with macrophage density. Thus, LDHA-mediated tumor-macrophage symbiosis provides therapeutic targets for glioblastoma.

13.
Leukemia ; 37(8): 1698-1708, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37391485

RESUMEN

Many inherited bone marrow failure syndromes (IBMFSs) present a high risk of transformation to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). During transformation of IBMFSs, hematopoietic stem and progenitor cells (HSPCs) with poor fitness gain ectopic, dysregulated self-renewal secondary to somatic mutations via undefined mechanisms. Here, in the context of the prototypical IBMFS Fanconi anemia (FA), we performed multiplexed gene editing of mutational hotspots in MDS-associated genes in human induced pluripotent stem cells (iPSCs) followed by hematopoietic differentiation. We observed aberrant self-renewal and impaired differentiation of HSPCs with enrichment of RUNX1 insertions and deletions (indels), generating a model of IBMFS-associated MDS. We observed that compared to the failure state, FA MDS cells show mutant RUNX1-mediated blunting of the G1/S cell cycle checkpoint that is normally activated in FA in response to DNA damage. RUNX1 indels also lead to activation of innate immune signaling, which stabilizes the homologous recombination (HR) effector BRCA1, and this pathway can be targeted to abrogate viability and restore sensitivity to genotoxins in FA MDS. Together, these studies develop a paradigm for modeling clonal evolution in IBMFSs, provide basic understanding of the pathogenesis of MDS, and uncover a therapeutic target in FA-associated MDS.


Asunto(s)
Anemia de Fanconi , Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Humanos , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Anemia de Fanconi/terapia , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Pluripotentes Inducidas/patología , Síndromes Mielodisplásicos/patología , Mutación , Leucemia Mieloide Aguda/patología
15.
Blood ; 115(2): 221-9, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19901267

RESUMEN

Human mesenchymal stem cells (hMSCs) localized to bone marrow, nonhematopoietic organs, as well as perivascular niches are postulated to traffic through type I collagen-rich stromal tissues to first infiltrate sites of tissue damage, inflammation, or neoplasia and then differentiate. Nevertheless, the molecular mechanisms supporting the ability of hMSCs to remodel 3-dimensional (3D) collagenous barriers during trafficking or differentiation remain undefined. Herein, we demonstrate that hMSCs degrade and penetrate type I collagen networks in tandem with the expression of a 5-member set of collagenolytic matrix metalloproteinases (MMPs). Specific silencing of each of these proteases reveals that only a single membrane-tethered metalloenzyme, termed MT1-MMP, plays a required role in hMSC-mediated collagenolysis, 3D invasion, and intravasation. Further, once confined within type I collagen-rich tissue, MT1-MMP also controls hMSC differentiation in a 3D-specific fashion. Together, these data demonstrate that hMSC invasion and differentiation programs fall under the control of the pericellular collagenase, MT1-MMP.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Colágeno Tipo I/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Metaloproteinasa 14 de la Matriz/biosíntesis , Células Madre Mesenquimatosas/enzimología , Células Cultivadas , Silenciador del Gen , Humanos , Células Madre Mesenquimatosas/citología
16.
Cell Rep ; 39(1): 110587, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385744

RESUMEN

Hematopoiesis changes over life to meet the demands of maturation and aging. Here, we find that the definitive hematopoietic stem and progenitor cell (HSPC) compartment is remodeled from gestation into adulthood, a process regulated by the heterochronic Lin28b/let-7 axis. Native fetal and neonatal HSPCs distribute with a pro-lymphoid/erythroid bias with a shift toward myeloid output in adulthood. By mining transcriptomic data comparing juvenile and adult HSPCs and reconstructing coordinately activated gene regulatory networks, we uncover the Polycomb repressor complex 1 (PRC1) component Cbx2 as an effector of Lin28b/let-7's control of hematopoietic maturation. We find that juvenile Cbx2-/- hematopoietic tissues show impairment of B-lymphopoiesis, a precocious adult-like myeloid bias, and that Cbx2/PRC1 regulates developmental timing of expression of key hematopoietic transcription factors. These findings define a mechanism of regulation of HSPC output via chromatin modification as a function of age with potential impact on age-biased pediatric and adult blood disorders.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , MicroARNs , Complejo Represivo Polycomb 1 , Proteínas de Unión al ARN , Adulto , Animales , Niño , Redes Reguladoras de Genes , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Recién Nacido , Linfopoyesis , Ratones , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
17.
Cell Rep ; 39(4): 110752, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476984

RESUMEN

High-risk forms of B-acute lymphoblastic leukemia (B-ALL) remain a therapeutic challenge. Leukemia-initiating cells (LICs) self-renew and spark relapse and therefore have been the subject of intensive investigation; however, the properties of LICs in high-risk B-ALL are not well understood. Here, we use single-cell transcriptomics and quantitative xenotransplantation to understand LICs in MLL-rearranged (MLL-r) B-ALL. Compared with reported LIC frequencies in acute myeloid leukemia (AML), engraftable LICs in MLL-r B-ALL are abundant. Although we find that multipotent, self-renewing LICs are enriched among phenotypically undifferentiated B-ALL cells, LICs with the capacity to replenish the leukemic cellular diversity can emerge from more mature fractions. While inhibiting oxidative phosphorylation blunts blast proliferation, this intervention promotes LIC emergence. Conversely, inhibiting hypoxia and glycolysis impairs MLL-r B-ALL LICs, providing a therapeutic benefit in xenotransplantation systems. These findings provide insight into the aggressive nature of MLL-r B-ALL and provide a rationale for therapeutic targeting of hypoxia and glycolysis.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Glucólisis , Humanos , Hipoxia , Leucemia Mieloide Aguda/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
18.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L683-92, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21840960

RESUMEN

In acute and chronic lung disease, widespread disruption of tissue architecture underlies compromised pulmonary function. Pulmonary fibroblasts have been implicated as critical effectors of tissue-destructive extracellular matrix (ECM) remodeling by mobilizing a spectrum of proteolytic enzymes. Although efforts to date have focused on the catabolism of type I collagen, the predominant component of the lung interstitial matrix, the key collagenolytic enzymes employed by pulmonary fibroblasts remain unidentified. Herein, membrane type-1 matrix metalloprotease (MT1-MMP) is identified as the dominant and direct-acting protease responsible for the type I collagenolytic activity mediated by both mouse and human pulmonary fibroblasts. Furthermore, MT1-MMP is shown to be essential for pulmonary fibroblast migration within three-dimensional (3-D) hydrogels of cross-linked type I collagen that recapitulate ECM barriers encountered in the in vivo environment. Together, these findings demonstrate that MT1-MMP serves as a key effector of type I collagenolytic activity in pulmonary fibroblasts and earmark this pericellular collagenase as a potential target for therapeutic intervention.


Asunto(s)
Asma/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos , Pulmón/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Fibrosis Pulmonar/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/patología , Asma/fisiopatología , Movimiento Celular , Enfermedad Crónica , Colágeno Tipo I , Matriz Extracelular/patología , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Hidrogeles , Pulmón/patología , Pulmón/fisiopatología , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Cultivo Primario de Células , Unión Proteica , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Biol Chem ; 284(34): 23001-11, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19542530

RESUMEN

Fibroblasts degrade type I collagen, the major extracellular protein found in mammals, during events ranging from bulk tissue resorption to invasion through the three-dimensional extracellular matrix. Current evidence suggests that type I collagenolysis is mediated by secreted as well as membrane-anchored members of the matrix metalloproteinase (MMP) gene family. However, the roles played by these multiple and possibly redundant, degradative systems during fibroblast-mediated matrix remodeling is undefined. Herein, we use fibroblasts isolated from Mmp13(-/-), Mmp8(-/-), Mmp2(-/-), Mmp9(-/-), Mmp14(-/-) and Mmp16(-/-) mice to define the functional roles for secreted and membrane-anchored collagenases during collagen-resorptive versus collagen-invasive events. In the presence of a functional plasminogen activator-plasminogen axis, secreted collagenases arm cells with a redundant collagenolytic potential that allows fibroblasts harboring single deficiencies for either MMP-13, MMP-8, MMP-2, or MMP-9 to continue to degrade collagen comparably to wild-type fibroblasts. Likewise, Mmp14(-/-) or Mmp16(-/-) fibroblasts retain near-normal collagenolytic activity in the presence of plasminogen via the mobilization of secreted collagenases, but only Mmp14 (MT1-MMP) plays a required role in the collagenolytic processes that support fibroblast invasive activity. Furthermore, by artificially tethering a secreted collagenase to the surface of Mmp14(-/-) fibroblasts, we demonstrate that localized pericellular collagenolytic activity differentiates the collagen-invasive phenotype from bulk collagen degradation. Hence, whereas secreted collagenases arm fibroblasts with potent matrix-resorptive activity, only MT1-MMP confers the focal collagenolytic activity necessary for supporting the tissue-invasive phenotype.


Asunto(s)
Colágeno Tipo I/metabolismo , Colagenasas/metabolismo , Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Becaplermina , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/fisiología , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/fisiología , Metaloproteinasa 16 de la Matriz/genética , Metaloproteinasa 16 de la Matriz/fisiología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 8 de la Matriz/genética , Metaloproteinasa 8 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Plasminógeno/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-sis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/fisiología
20.
Blood Adv ; 4(19): 4679-4692, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33002135

RESUMEN

Fanconi anemia (FA) is a disorder of DNA repair that manifests as bone marrow (BM) failure. The lack of accurate murine models of FA has refocused efforts toward differentiation of patient-derived induced pluripotent stem cells (IPSCs) to hematopoietic progenitor cells (HPCs). However, an intact FA DNA repair pathway is required for efficient IPSC derivation, hindering these efforts. To overcome this barrier, we used inducible complementation of FANCA-deficient IPSCs, which permitted robust maintenance of IPSCs. Modulation of FANCA during directed differentiation to HPCs enabled the production of FANCA-deficient human HPCs that recapitulated FA genotoxicity and hematopoietic phenotypes relative to isogenic FANCA-expressing HPCs. FANCA-deficient human HPCs underwent accelerated terminal differentiation driven by activation of p53/p21. We identified growth arrest specific 6 (GAS6) as a novel target of activated p53 in FANCA-deficient HPCs and modulate GAS6 signaling to rescue hematopoiesis in FANCA-deficient cells. This study validates our strategy to derive a sustainable, highly faithful human model of FA, uncovers a mechanism of HPC exhaustion in FA, and advances toward future cell therapy in FA.


Asunto(s)
Anemia de Fanconi , Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Humanos , Ratones , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA