Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Blood ; 142(18): 1529-1542, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37584437

RESUMEN

The cross talk between extrinsic niche-derived and intrinsic hematopoietic stem cell (HSC) factors controlling HSC maintenance remains elusive. Here, we demonstrated that amphiregulin (AREG) from bone marrow (BM) leptin receptor (LepR+) niche cells is an important factor that mediates the cross talk between the BM niche and HSCs in stem cell maintenance. Mice deficient of the DNA repair gene Brca2, specifically in LepR+ cells (LepR-Cre;Brca2fl/fl), exhibited increased frequencies of total and myeloid-biased HSCs. Furthermore, HSCs from LepR-Cre;Brca2fl/fl mice showed compromised repopulation, increased expansion of donor-derived, myeloid-biased HSCs, and increased myeloid output. Brca2-deficient BM LepR+ cells exhibited persistent DNA damage-inducible overproduction of AREG. Ex vivo treatment of wild-type HSCs or systemic treatment of C57BL/6 mice with recombinant AREG impaired repopulation, leading to HSC exhaustion. Conversely, inhibition of AREG by an anti-AREG-neutralizing antibody or deletion of the Areg gene in LepR-Cre;Brca2fl/fl mice rescued HSC defects caused by AREG. Mechanistically, AREG activated the phosphoinositide 3-kinases (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, promoted HSC cycling, and compromised HSC quiescence. Finally, we demonstrated that BM LepR+ niche cells from other DNA repair-deficient and aged mice also showed persistent DNA damage-associated overexpression of AREG, which exerts similar negative effects on HSC maintenance. Therefore, we identified an important factor that regulates HSCs function under conditions of DNA repair deficiency and aging.


Asunto(s)
Trastornos por Deficiencias en la Reparación del ADN , Receptores de Leptina , Ratones , Animales , Anfirregulina/genética , Anfirregulina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Ratones Endogámicos C57BL , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/genética , Trastornos por Deficiencias en la Reparación del ADN/metabolismo , Nicho de Células Madre/genética , Mamíferos/metabolismo
2.
Haematologica ; 107(11): 2576-2588, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35443564

RESUMEN

The immune receptor TREM1 (Triggering receptor expressed on myeloid cells 1) is a master regulator of inflammatory response. Compelling evidence suggests important pathological roles for TREM1 in various types of solid tumors. However, the role of TREM1 in hematologic malignancies is not known. Our previous study demonstrated that TREM1 cooperates with diminished DNA damage response to induce expansion of pre-leukemic hematopoietic stem cells (HSC) in mice deficient for the Fanconi anemia gene Fanca. Here we investigated TREM1 in leukemogenesis using mouse models of the DNA repair-deficient Fanca-/- and the oncogenic MLL-AF9 or KrasG12D. We found that Trem1 was highly expressed in preleukemic HSC and leukemia stem cells (LSC). By selective deletion of the Trem1 gene in the hematopoietic compartment, we showed that ablation of Trem1 reduced leukemogenic activity of the pre-leukemic HSC and LSC in mice. Trem1 was required for the proliferation of the pre-leukemic HSC and LSC. Further analysis revealed that Trem1 expression in preleukemic HSC and LSC was associated with persistent DNA damage, prolonged oncogenic stress, and a strong inflammatory signature. Targeting several top Trem1 inflammatory signatures inhibited the proliferation of pre-leukemic HSC and LSC. Collectively, our observations uncover previously unknown expression and function of TREM1 in malignant stem cells, and identify TREM1 as a driver of leukemogenesis.


Asunto(s)
Anemia de Fanconi , Leucemia Mieloide Aguda , Ratones , Animales , Receptor Activador Expresado en Células Mieloides 1/genética , Células Madre Neoplásicas/metabolismo , Leucemia Mieloide Aguda/genética , Daño del ADN , Anemia de Fanconi/patología , Carcinogénesis/metabolismo
3.
Stem Cells ; 34(4): 960-71, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676373

RESUMEN

Fanconi anemia (FA) is an inherited bone marrow (BM) failure syndrome, presumably resulting from defects in hematopoietic stem cells (HSCs). Normal HSCs depend more on glycolysis than on oxidative phosphorylation (OXPHOS) for energy production. Here, we show that FA HSCs are more sensitive to the respiration inhibitor NaN3 treatment than to glycolytic inhibitor 2-deoxy-d-glucose (2-DG), indicating more dependence on OXPHOS. FA HSCs undergo glycolysis-to-OXPHOS switch in response to oxidative stress through a p53-dependent mechanism. Metabolic stresses induce upregulation of p53 metabolic targets in FA HSCs. Inactivation of p53 in FA HSCs prevents glycolysis-to-OXPHOS switch. Furthermore, p53-deficient FA HSCs are more sensitive to 2-DG-mediated metabolic stress. Finally, oxidative stress-induced glycolysis-to-OXPHOS switch is mediated by synthesis of cytochrome c oxidase 2 (SCO2). These findings demonstrate p53-mediated OXPHOS function as a compensatory alteration in FA HSCs to ensure a functional but mildly impaired energy metabolism and suggest a cautious approach to manipulating p53 signaling in FA.


Asunto(s)
Proteínas Portadoras/biosíntesis , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Mitocondriales/biosíntesis , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Proteínas Portadoras/genética , Desoxiglucosa/administración & dosificación , Metabolismo Energético , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Glucólisis/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Proteínas Mitocondriales/genética , Chaperonas Moleculares , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Azida Sódica/administración & dosificación , Proteína p53 Supresora de Tumor/metabolismo
4.
Stem Cells ; 33(7): 2320-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25917546

RESUMEN

20-kDa FANCA-associated protein (FAAP20) is a recently identified protein that associates with the Fanconi anemia (FA) core complex component, FANCA. FAAP20 contains a conserved ubiquitin-binding zinc-finger domain and plays critical roles in the FA-BRCA pathway of DNA repair and genome maintenance. The function of FAAP20 in animals has not been explored. Here, we report that deletion of Faap20 in mice led to a mild FA-like phenotype with defects in the reproductive and hematopoietic systems. Specifically, hematopoietic stem and progenitor cells (HSPCs) from Faap20(-) (/) (-) mice showed defects in long-term multilineage reconstitution in lethally irradiated recipient mice, with milder phenotype as compared to HSPCs from Fanca(-) (/) (-) or Fancc(-) (/) (-) mice. Faap20(-) (/) (-) mice are susceptible to mitomycin C (MMC)-induced pancytopenia. That is, acute MMC stress induced a significant progenitor loss especially the erythroid progenitors and megakaryocyte-erythrocyte progenitors in Faap20(-) (/) (-) mice. Furthermore, Faap20(-) (/) (-) HSPCs displayed aberrant cell cycle pattern during chronic MMC treatment. Finally, using Faap20(-) (/) (-) Fanca(-) (/) (-) double-knockout mice, we demonstrated a possible dominant effect of FANCA in the interaction between FAAP20 and FANCA. This novel Faap20 mouse model may be valuable in studying the regulation of the FA pathway during bone marrow failure progress in FA patients.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Aminoacil-ARNt Sintetasas/genética , Animales , Diferenciación Celular , Daño del ADN , Anemia de Fanconi/mortalidad , Humanos , Ratones , Ratones Endogámicos C57BL , Células Madre , Análisis de Supervivencia
6.
Int J Stem Cells ; 12(3): 457-462, 2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31474030

RESUMEN

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure and high risk of cancer particularly leukemia. Here we show that inactivation of the non-homologous end-joining (NHEJ) activity of DNA-PKcs prevented DNA damage-induced expansion of FA pre-leukemic hematopoietic stem cells (HSCs). Furthermore, we performed serial BM transplantation to demonstrate that the DNA damage-induced expanded FA HSC compartment contained pre-leukemic stem cells that required the NHEJ activity of DNA-PKcs to induce leukemia in the secondary recipients. These results suggest that NHEJ may collaborate with FA deficiency to promote DNA damage-induced expansion of pre-leukemic HSCs.

7.
Stem Cell Res ; 40: 101550, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31472450

RESUMEN

Members of the Fanconi anemia (FA) protein family are involved in multiple cellular processes including response to DNA damage and oxidative stress. Here we show that a major FA protein, Fancd2, plays a role in mitochondrial biosynthesis through regulation of mitochondrial translation. Fancd2 interacts with Atad3 and Tufm, which are among the most frequently identified components of the mitochondrial nucleoid complex essential for mitochondrion biosynthesis. Deletion of Fancd2 in mouse hematopoietic stem and progenitor cells (HSPCs) leads to increase in mitochondrial number, and enzyme activity of mitochondrion-encoded respiratory complexes. Fancd2 deficiency increases mitochondrial protein synthesis and induces mitonuclear protein imbalance. Furthermore, Fancd2-deficient HSPCs show increased mitochondrial respiration and mitochondrial reactive oxygen species. By using a cell-free assay with mitochondria isolated from WT and Fancd2-KO HSPCs, we demonstrate that the increased mitochondrial protein synthesis observed in Fancd2-KO HSPCs was directly linked to augmented mitochondrial translation. Finally, Fancd2-deficient HSPCs are selectively sensitive to mitochondrial translation inhibition and depend on augmented mitochondrial translation for survival and proliferation. Collectively, these results suggest that Fancd2 restricts mitochondrial activity through regulation of mitochondrial translation, and that augmented mitochondrial translation and mitochondrial respiration may contribute to HSC defect and bone marrow failure in FA.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mitocondrias/metabolismo , Biosíntesis de Proteínas , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Anemia de Fanconi/genética , Anemia de Fanconi/fisiopatología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Técnicas de Inactivación de Genes , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Factor Tu de Elongación Peptídica/genética , Factor Tu de Elongación Peptídica/metabolismo , Unión Proteica
8.
Stem Cell Res Ther ; 10(1): 114, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30925933

RESUMEN

BACKGROUND: Recent studies have shown that deficiency in the Fanconi anemia (FA) DNA repair pathway enhances the error-prone non-homologous end-joining (NHEJ) repair, leading to increased genomic instability, and that genetic or pharmacological inhibition of the NHEJ pathway could rescue the FA phenotype. METHODS: First, we exposed LSK cells from WT and Fanca-/- mice to DNA-PKcs inhibitor NU7026 or Ku70 knockdown to examine whether inhibition of NHEJ sensitizes Fanca-/- HSPCs to PARP inhibitor (PARPi)- or interstrand crosslinking (ICL)-induced cell death and genomic instability. We then generated DNA-PKcs3A/3AFanca-/- mice to investigate the effect of specific inactivation of NHEJ on fetal HSCs. Lastly, we used two p53 mutant models to test whether specific inactivation of the p53 function in apoptosis is sufficient to rescue embryonic lethality and fetal HSC depletion in Fanca-/- DNA-PKcs3A/3A mice. RESULTS: Inhibition of NHEJ sensitizes HSPCs from Fanca-/- mice to PARP inhibition- and ICL-induced cell death and genomic instability and further decreases Fanca-/- HSPC proliferation and hematopoietic repopulation in irradiated transplant recipients. Specific inactivation of NHEJ activity by the knockin DNA-PKcs3A/3A mutation in two FA mouse models, Fanca-/- and Fancc-/-, leads to embryonic lethality. DNA-PKcs3A/3A causes fetal HSC depletion in developing Fanca-/- embryos due to increased HSC apoptosis and cycling. Both p53-/- and a knockin p53515C mutation, which selectively impairs the p53 function in apoptosis, can rescue embryonic lethality and fetal HSC depletion in Fanca-/- DNA-PKcs3A/3A mice. CONCLUSION: These results demonstrate that the NHEJ pathway functions to maintain Fanconi anemia fetal HSCs.


Asunto(s)
Apoptosis , Reparación del ADN por Unión de Extremidades , Anemia de Fanconi , Feto , Inestabilidad Genómica , Células Madre Hematopoyéticas , Animales , Modelos Animales de Enfermedad , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Feto/metabolismo , Feto/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Ratones , Ratones Noqueados
9.
Stem Cell Reports ; 10(2): 339-346, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29307578

RESUMEN

Overactive p53 has been proposed as an important pathophysiological factor for bone marrow failure syndromes, including Fanconi anemia (FA). Here, we report a p53-dependent effect on hematopoietic stem and progenitor cell (HSPC) proliferation in mice deficient for the FA gene Fanca. Deletion of p53 in Fanca-/- mice leads to replicative exhaustion of the hematopoietic stem cell (HSC) in transplant recipients. Using Fanca-/- HSCs expressing the separation-of-function mutant p53515C transgene, which selectively impairs the p53 function in apoptosis but keeps its cell-cycle checkpoint activities intact, we show that the p53 cell-cycle function is specifically required for the regulation of Fanca-/- HSC proliferation. Our results demonstrate that p53 plays a compensatory role in preventing FA HSCs from replicative exhaustion and suggest a cautious approach to manipulating p53 signaling as a therapeutic utility in FA.


Asunto(s)
Anemia Aplásica/genética , Enfermedades de la Médula Ósea/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Hemoglobinuria Paroxística/genética , Proteína p53 Supresora de Tumor/genética , Anemia Aplásica/patología , Animales , Enfermedades de la Médula Ósea/patología , Trastornos de Fallo de la Médula Ósea , Puntos de Control del Ciclo Celular/genética , Proliferación Celular/genética , Anemia de Fanconi/patología , Regulación del Desarrollo de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/patología , Humanos , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transgenes/genética
10.
Stem Cell Res Ther ; 9(1): 145, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29784053

RESUMEN

Although p53 mutations are common in solid tumors, such mutations are found at a lower frequency in hematologic malignancies. In the genetic disorder Fanconi anemia (FA), p53 has been proposed as an important pathophysiological factor for two important hematologic hallmarks of the disease: bone marrow failure and leukemogenesis. Here we show that low levels of the p53 protein enhance the capacity of leukemic stem cells from FA patients to repopulate immunodeficient mice. Furthermore, boosting p53 protein levels with the use of the small molecule Nutlin-3 reduced leukemia burden in recipient mice. These results demonstrate that the level of p53 protein plays a crucial role in FA leukemogenesis.


Asunto(s)
Anemia de Fanconi/genética , Proteína p53 Supresora de Tumor/genética , Proliferación Celular , Humanos
11.
Cell Cycle ; 16(12): 1201-1209, 2017 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-28475398

RESUMEN

Oxidative stress is considered as an important pathogenic factor in many human diseases including Fanconi anemia (FA), an inherited bone marrow failure syndrome with extremely high risk of leukemic transformation. Members of the FA protein family are involved in DNA damage and other cellular stress responses. Loss of FA proteins renders cells hypersensitive to oxidative stress and cancer transformation. However, how FA cells respond to oxidative DNA damage remains unclear. By using an in vivo stress-response mouse strain expressing the Gadd45ß-luciferase transgene, we show here that haematopoietic stem and progenitor cells (HSPCs) from mice deficient for the FA gene Fanca or Fancc persistently responded to oxidative stress. Mechanistically, we demonstrated that accumulation of unrepaired DNA damage, particularly in oxidative damage-sensitive genes, was responsible for the long-lasting response in FA HSPCs. Furthermore, genetic correction of Fanca deficiency almost completely abolished the persistent oxidative stress-induced G2/M arrest and DNA damage response in vivo. Our study suggests that FA pathway is an integral part of a versatile cellular mechanism by which HSPCs respond to oxidative stress.


Asunto(s)
Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Estrés Oxidativo , Animales , Células Cultivadas , Daño del ADN , Anemia de Fanconi/patología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Ratones Noqueados
12.
Sci Rep ; 7: 45626, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28378742

RESUMEN

Fancd2 is a component of the Fanconi anemia (FA) DNA repair pathway, which is frequently found defective in human cancers. The full repertoire of Fancd2 functions in normal development and tumorigenesis remains to be determined. Here we developed a Flag- and hemagglutinin-tagged Fancd2 knock-in mouse strain that allowed a high throughput mass spectrometry approach to search for Fancd2-binding proteins in different mouse organs. In addition to DNA repair partners, we observed that many Fancd2-interacting proteins are mitochondrion-specific. Fancd2 localizes in the mitochondrion and associates with the nucleoid complex components Atad3 and Tufm. The Atad3-Tufm complex is disrupted in Fancd2-/- mice and those deficient for the FA core component Fanca. Fancd2 mitochondrial localization requires Atad3. Collectively, these findings provide evidence for Fancd2 as a crucial regulator of mitochondrion biosynthesis, and of a molecular link between FA and mitochondrial homeostasis.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , Mapas de Interacción de Proteínas , Animales , Técnicas de Sustitución del Gen , Espectrometría de Masas , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica
13.
Sci Rep ; 6: 22167, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26916217

RESUMEN

The prominent role of Fanconi anemia (FA) proteins involves homologous recombination (HR) repair. Poly[ADP-ribose] polymerase1 (PARP1) functions in multiple cellular processes including DNA repair and PARP inhibition is an emerging targeted therapy for cancer patients deficient in HR. Here we show that PARP1 activation in hematopoietic stem and progenitor cells (HSPCs) in response to genotoxic or oxidative stress attenuates HSPC exhaustion. Mechanistically, PARP1 controls the balance between HR and non-homologous end joining (NHEJ) in double strand break (DSB) repair by preventing excessive NHEJ. Disruption of the FA core complex skews PARP1 function in DSB repair and led to hyper-active NHEJ in Fanca(-/-) or Fancc(-/-) HSPCs. Re-expression of PARP1 rescues the hyper-active NHEJ phenotype in Brca1(-/-)Parp1(-/-) but less effective in Fanca(-/-)Parp1(-/-) cells. Inhibition of NHEJ prevents myeloid/erythroid pathologies associated with synthetic lethality. Our results suggest that hyper-active NHEJ may select for "synthetic lethality" resistant and pathological HSPCs.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Células Madre Hematopoyéticas/citología , Recombinación Homóloga/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Mutaciones Letales Sintéticas/genética , Animales , Proteína BRCA1 , Línea Celular , Roturas del ADN de Doble Cadena , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA