Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 24(6): 944-957.e5, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31006621

RESUMEN

Stem cell heterogeneity is recognized as functionally relevant for tissue homeostasis and repair. The identity, context dependence, and regulation of skeletal muscle satellite cell (SC) subsets remains poorly understood. We identify a minor subset of Pax7+ SCs that is indelibly marked by an inducible Mx1-Cre transgene in vivo, is enriched for Pax3 expression, and has reduced ROS (reactive oxygen species) levels. Mx1+ SCs possess potent stem cell activity upon transplantation but minimally contribute to endogenous muscle repair, due to their relative low abundance. In contrast, a dramatic clonal expansion of Mx1+ SCs allows extensive contribution to muscle repair and niche repopulation upon selective pressure of radiation stress, consistent with reserve stem cell (RSC) properties. Loss of Pax3 in RSCs increased ROS content and diminished survival and stress tolerance. These observations demonstrate that the Pax7+ SC pool contains a discrete population of radiotolerant RSCs that undergo clonal expansion under severe stress.


Asunto(s)
Células Madre Adultas/fisiología , Daño del ADN/fisiología , Células Satélite del Músculo Esquelético/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Células Clonales , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Resistencia a Mixovirus/metabolismo , Factor de Transcripción PAX3/metabolismo , Factor de Transcripción PAX7/metabolismo , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Regeneración , Regulación hacia Arriba
2.
Nat Commun ; 10(1): 616, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728353

RESUMEN

Hematopoietic chimerism after allogeneic bone marrow transplantation may establish a state of donor antigen-specific tolerance. However, current allotransplantation protocols involve genotoxic conditioning which has harmful side-effects and predisposes to infection and cancer. Here we describe a non-genotoxic conditioning protocol for fully MHC-mismatched bone marrow allotransplantation in mice involving transient immunosuppression and selective depletion of recipient hematopoietic stem cells with a CD117-antibody-drug-conjugate (ADC). This protocol resulted in multilineage, high level (up to 50%), durable, donor-derived hematopoietic chimerism after transplantation of 20 million total bone marrow cells, compared with ≤ 2.1% hematopoietic chimerism from 50 million total bone marrow cells without conditioning. Moreover, long-term survival of bone marrow donor-type but not third party skin allografts is achieved in CD117-ADC-conditioned chimeric mice without chronic immunosuppression. The only observed adverse event is transient elevation of liver enzymes in the first week after conditioning. These results provide proof-of-principle for CD117-ADC as a non-genotoxic, highly-targeted conditioning agent in allotransplantation and tolerance protocols.


Asunto(s)
Trasplante de Médula Ósea/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoconjugados/farmacología , Proteínas Proto-Oncogénicas c-kit/inmunología , Tolerancia al Trasplante/efectos de los fármacos , Animales , Células de la Médula Ósea/efectos de los fármacos , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Células Madre Hematopoyéticas , Tolerancia Inmunológica , Terapia de Inmunosupresión/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Piel/patología , Trasplante de Piel/métodos , Quimera por Trasplante , Trasplante Homólogo
3.
Nat Commun ; 10(1): 617, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728354

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is a curative therapy for blood and immune diseases with potential for many settings beyond current standard-of-care. Broad HSCT application is currently precluded largely due to morbidity and mortality associated with genotoxic irradiation or chemotherapy conditioning. Here we show that a single dose of a CD117-antibody-drug-conjugate (CD117-ADC) to saporin leads to > 99% depletion of host HSCs, enabling rapid and efficient donor hematopoietic cell engraftment. Importantly, CD117-ADC selectively targets hematopoietic stem cells yet does not cause clinically significant side-effects. Blood counts and immune cell function are preserved following CD117-ADC treatment, with effective responses by recipients to both viral and fungal challenges. These results suggest that CD117-ADC-mediated HSCT pre-treatment could serve as a non-myeloablative conditioning strategy for the treatment of a wide range of non-malignant and malignant diseases, and might be especially suited to gene therapy and gene editing settings in which preservation of immunity is desired.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Inmunoconjugados/farmacología , Proteínas Proto-Oncogénicas c-kit/inmunología , Animales , Médula Ósea/efectos de los fármacos , Trasplante de Médula Ósea , Candida albicans/patogenicidad , Muerte Celular , Línea Celular , Femenino , Terapia Genética , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Neoplasias , Donantes de Tejidos
4.
Elife ; 72018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30561324

RESUMEN

A hallmark of adult hematopoiesis is the continuous replacement of blood cells with limited lifespans. While active hematopoietic stem cell (HSC) contribution to multilineage hematopoiesis is the foundation of clinical HSC transplantation, recent reports have questioned the physiological contribution of HSCs to normal/steady-state adult hematopoiesis. Here, we use inducible lineage tracing from genetically marked adult HSCs and reveal robust HSC-derived multilineage hematopoiesis. This commences via defined progenitor cells, but varies substantially in between different hematopoietic lineages. By contrast, adult HSC contribution to hematopoietic cells with proposed fetal origins is neglible. Finally, we establish that the HSC contribution to multilineage hematopoiesis declines with increasing age. Therefore, while HSCs are active contributors to native adult hematopoiesis, it appears that the numerical increase of HSCs is a physiologically relevant compensatory mechanism to account for their reduced differentiation capacity with age.


Asunto(s)
Envejecimiento/fisiología , Diferenciación Celular , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Factores de Edad , Animales , Linaje de la Célula , Ratones , Coloración y Etiquetado
5.
Elife ; 72018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30468428

RESUMEN

DNA methylation plays an essential role in mammalian genomes and expression of the responsible enzymes is tightly controlled. Deregulation of the de novo DNA methyltransferase DNMT3B is frequently observed across cancer types, yet little is known about its ectopic genomic targets. Here, we used an inducible transgenic mouse model to delineate rules for abnormal DNMT3B targeting, as well as the constraints of its activity across different cell types. Our results explain the preferential susceptibility of certain CpG islands to aberrant methylation and point to transcriptional state and the associated chromatin landscape as the strongest predictors. Although DNA methylation and H3K27me3 are usually non-overlapping at CpG islands, H3K27me3 can transiently co-occur with DNMT3B-induced DNA methylation. Our genome-wide data combined with ultra-deep locus-specific bisulfite sequencing suggest a distributive activity of ectopically expressed Dnmt3b that leads to discordant CpG island hypermethylation and provides new insights for interpreting the cancer methylome.


Asunto(s)
Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Metilación de ADN , Expresión Génica , Proteínas Recombinantes/biosíntesis , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica , Humanos , Ratones Transgénicos , Neoplasias/patología , Proteínas Recombinantes/genética , ADN Metiltransferasa 3B
6.
Cell ; 174(1): 156-171.e16, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29909984

RESUMEN

Extracellular proTGF-ß is covalently linked to "milieu" molecules in the matrix or on cell surfaces and is latent until TGF-ß is released by integrins. Here, we show that LRRC33 on the surface of microglia functions as a milieu molecule and enables highly localized, integrin-αVß8-dependent TGF-ß activation. Lrrc33-/- mice lack CNS vascular abnormalities associated with deficiency in TGF-ß-activating integrins but have microglia with a reactive phenotype and after 2 months develop ascending paraparesis with loss of myelinated axons and death by 5 months. Whole bone marrow transplantation results in selective repopulation of Lrrc33-/- brains with WT microglia and halts disease progression. The phenotypes of WT and Lrrc33-/- microglia in the same brain suggest that there is little spreading of TGF-ß activated from one microglial cell to neighboring microglia. Our results suggest that interactions between integrin-bearing cells and cells bearing milieu molecule-associated TGF-ß provide localized and selective activation of TGF-ß.


Asunto(s)
Proteínas Portadoras/metabolismo , Microglía/metabolismo , Sistema Nervioso/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Axones/metabolismo , Trasplante de Médula Ósea , Encéfalo/metabolismo , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Células Cultivadas , Integrinas/metabolismo , Estimación de Kaplan-Meier , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Mutagénesis Sitio-Dirigida , Enfermedades Neurodegenerativas/mortalidad , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Filogenia , Unión Proteica , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Factor de Crecimiento Transformador beta/genética
7.
Nat Cell Biol ; 20(4): 413-421, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29531308

RESUMEN

Ageing of haematopoietic stem cells (HSCs) contributes to deficits in the aged haematopoietic system. HSC decline is driven in part by DNA damage accumulation; yet, how ageing impacts the acute DNA damage response (DDR) of HSCs is poorly understood. We show that old HSCs exhibit diminished ATM activity and attenuated DDR, leading to elevated clonal survival in response to a range of genotoxins that was underwritten by diminished apoptotic priming. Distinct HSC subsets exhibited ageing-dependent and subtype-dependent differences in apoptotic priming and survival in response to DNA damage. The defective DDR of old HSCs was non-cell autonomous, as ATM signalling and clonal survival in response to DNA damage could be restored to levels observed in young HSCs post-transplantated into young recipients. These data indicate that defective DDR and diminished apoptotic priming provide a selective advantage to old HSCs that may contribute to mutation accrual and disease predisposition.


Asunto(s)
Apoptosis , Senescencia Celular , Daño del ADN , Reparación del ADN , Células Madre Hematopoyéticas/enzimología , Factores de Edad , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Masculino , Ratones Endogámicos C57BL , Transducción de Señal , Factores de Tiempo
8.
J Biol Chem ; 293(19): 7300-7314, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29593094

RESUMEN

In humans, six α(1,3)-fucosyltransferases (α(1,3)-FTs: FT3/FT4/FT5/FT6/FT7/FT9) reportedly fucosylate terminal lactosaminyl glycans yielding Lewis-X (LeX; CD15) and/or sialyl Lewis-X (sLeX; CD15s), structures that play key functions in cell migration, development, and immunity. Prior studies analyzing α(1,3)-FT specificities utilized either purified and/or recombinant enzymes to modify synthetic substrates under nonphysiological reaction conditions or molecular biology approaches wherein α(1,3)-FTs were expressed in mammalian cell lines, notably excluding investigations using primary human cells. Accordingly, although significant insights into α(1,3)-FT catalytic properties have been obtained, uncertainty persists regarding their human LeX/sLeX biosynthetic range across various glycoconjugates. Here, we undertook a comprehensive evaluation of the lactosaminyl product specificities of intracellularly expressed α(1,3)-FTs using a clinically relevant primary human cell type, mesenchymal stem cells. Cells were transfected with modified mRNA encoding each human α(1,3)-FT, and the resultant α(1,3)-fucosylated lactosaminyl glycoconjugates were analyzed using a combination of flow cytometry and MS. The data show that biosynthesis of sLeX is driven by FTs-3, -5, -6, and -7, with FT6 and FT7 having highest potency. FT4 and FT9 dominantly biosynthesize LeX, and, among all FTs, FT6 holds a unique capacity in creating sLeX and LeX determinants across protein and lipid glycoconjugates. Surprisingly, FT4 does not generate sLeX on glycolipids, and neither FT4, FT6, nor FT9 synthesizes the internally fucosylated sialyllactosamine VIM-2 (CD65s). These results unveil the relevant human lactosaminyl glycans created by human α(1,3)-FTs, providing novel insights on how these isoenzymes stereoselectively shape biosynthesis of vital glycoconjugates, thereby biochemically programming human cell migration and tuning human immunologic and developmental processes.


Asunto(s)
Fucosiltransferasas/metabolismo , Isoenzimas/metabolismo , Antígeno Lewis X/metabolismo , Células Madre Mesenquimatosas/enzimología , Amino Azúcares/metabolismo , Citometría de Flujo , Fucosiltransferasas/genética , Glicoconjugados/metabolismo , Glicómica , Humanos , Isoenzimas/genética , Antígeno Lewis X/genética , Espectrometría de Masas , Células Madre Mesenquimatosas/inmunología , ARN Mensajero/genética , Antígeno Sialil Lewis X
9.
Sci Adv ; 3(12): e1701211, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29226242

RESUMEN

Recent evidence indicates that hematopoietic stem and progenitor cells (HSPCs) can serve as vehicles for therapeutic molecular delivery to the brain by contributing to the turnover of resident myeloid cell populations. However, such engraftment needs to be fast and efficient to exert its therapeutic potential for diseases affecting the central nervous system. Moreover, the nature of the cells reconstituted after transplantation and whether they could comprise bona fide microglia remain to be assessed. We demonstrate that transplantation of HSPCs in the cerebral lateral ventricles provides rapid engraftment of morphologically, antigenically, and transcriptionally dependable microglia-like cells. We show that the cells comprised within the hematopoietic stem cell compartment and enriched early progenitor fractions generate this microglia-like population when injected in the brain ventricles in the absence of engraftment in the bone marrow. This delivery route has therapeutic relevance because it increases the delivery of therapeutic molecules to the brain, as shown in a humanized animal model of a prototypical lysosomal storage disease affecting the central nervous system.


Asunto(s)
Ventrículos Cerebrales/citología , Trasplante de Células Madre Hematopoyéticas/métodos , Microglía/citología , Animales , Antígenos CD34 , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucodistrofia Metacromática/etiología , Leucodistrofia Metacromática/terapia , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología
10.
Blood ; 130(5): 619-624, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28615219

RESUMEN

The concept that tumor-initiating cells can co-opt the self-renewal program of endogenous stem cells as a means of enforcing their unlimited proliferative potential is widely accepted, yet identification of specific factors that regulate self-renewal of normal and cancer stem cells remains limited. Using a comparative transcriptomic approach, we identify ZNF521/Zfp521 as a conserved hematopoietic stem cell (HSC)-enriched transcription factor in human and murine hematopoiesis whose function in HSC biology remains elusive. Competitive serial transplantation assays using Zfp521-deficient mice revealed that ZFP521 regulates HSC self-renewal and differentiation. In contrast, ectopic expression of ZFP521 in HSCs led to a robust maintenance of progenitor activity in vitro. Transcriptional analysis of human acute myeloid leukemia (AML) patient samples revealed that ZNF521 is highly and specifically upregulated in AMLs with MLL translocations. Using an MLL-AF9 murine leukemia model and serial transplantation studies, we show that ZFP521 is not required for leukemogenesis, although its absence leads to a significant delay in leukemia onset. Furthermore, knockdown of ZNF521 reduced proliferation in human leukemia cell lines possessing MLL-AF9 translocations. Taken together, these results identify ZNF521/ZFP521 as a critical regulator of HSC function, which facilitates MLL-AF9-mediated leukemic disease in mice.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Neoplasias Experimentales/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/patología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Factores de Transcripción/genética , Translocación Genética
11.
Stem Cell Reports ; 8(6): 1563-1572, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28479303

RESUMEN

Recent evidence points to the embryonic emergence of some tissue-resident innate immune cells, such as B-1a lymphocytes, prior to and independently of hematopoietic stem cells (HSCs). However, whether the full hematopoietic repertoire of embryonic HSCs initially includes these unique lineages of innate immune cells has been difficult to assess due to lack of clonal assays that identify and assess HSC precursor (pre-HSC) potential. Here, by combining index sorting of single embryonic hemogenic precursors with in vitro HSC maturation and transplantation assays, we analyze emerging pre-HSCs at the single-cell level, revealing their unique stage-specific properties and clonal lineage potential. Remarkably, clonal pre-HSCs detected between E9.5 and E11.5 contribute to the complete B cell repertoire, including B-1a lymphocytes, revealing a previously unappreciated common precursor for all B cell lineages at the pre-HSC stage and a second embryonic origin for B-1a lymphocytes.


Asunto(s)
Linfocitos B/metabolismo , Células Madre Hematopoyéticas/citología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/metabolismo , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/citología , Cadherinas/metabolismo , Proteínas de Unión al Calcio , Células Cultivadas , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Femenino , Citometría de Flujo , Factores de Intercambio de Guanina Nucleótido/genética , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL
12.
J Clin Invest ; 127(6): 2433-2437, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28481220

RESUMEN

Generation of functional hematopoietic stem and progenitor cells (HSPCs) from human pluripotent stem cells (PSCs) has been a long-sought-after goal for use in hematopoietic cell production, disease modeling, and eventually transplantation medicine. Homing of HSPCs from bloodstream to bone marrow (BM) is an important aspect of HSPC biology that has remained unaddressed in efforts to derive functional HSPCs from human PSCs. We have therefore examined the BM homing properties of human induced pluripotent stem cell-derived HSPCs (hiPS-HSPCs). We found that they express molecular effectors of BM extravasation, such as the chemokine receptor CXCR4 and the integrin dimer VLA-4, but lack expression of E-selectin ligands that program HSPC trafficking to BM. To overcome this deficiency, we expressed human fucosyltransferase 6 using modified mRNA. Expression of fucosyltransferase 6 resulted in marked increases in levels of cell surface E-selectin ligands. The glycoengineered cells exhibited enhanced tethering and rolling interactions on E-selectin-bearing endothelium under flow conditions in vitro as well as increased BM trafficking and extravasation when transplanted into mice. However, glycoengineered hiPS-HSPCs did not engraft long-term, indicating that additional functional deficiencies exist in these cells. Our results suggest that strategies toward increasing E-selectin ligand expression could be applicable as part of a multifaceted approach to optimize the production of HSPCs from human PSCs.


Asunto(s)
Movimiento Celular , Células Madre Hematopoyéticas/fisiología , ARN Mensajero/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Selectina E , Fucosiltransferasas/fisiología , Glicosilación , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Ratones Endogámicos NOD , Ratones SCID , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética
13.
Nat Biomed Eng ; 1(11): 878-888, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-31015609

RESUMEN

Gene disruption by clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) is highly efficient and relies on the error-prone non-homologous end-joining pathway. Conversely, precise gene editing requires homology-directed repair (HDR), which occurs at a lower frequency than non-homologous end-joining in mammalian cells. Here, by testing whether manipulation of DNA repair factors improves HDR efficacy, we show that transient ectopic co-expression of RAD52 and a dominant-negative form of tumour protein p53-binding protein 1 (dn53BP1) synergize to enable efficient HDR using a single-stranded oligonucleotide DNA donor template at multiple loci in human cells, including patient-derived induced pluripotent stem cells. Co-expression of RAD52 and dn53BP1 improves multiplexed HDR-mediated editing, whereas expression of RAD52 alone enhances HDR with Cas9 nickase. Our data show that the frequency of non-homologous end-joining-mediated double-strand break repair in the presence of these two factors is not suppressed and suggest that dn53BP1 competitively antagonizes 53BP1 to augment HDR in combination with RAD52. Importantly, co-expression of RAD52 and dn53BP1 does not alter Cas9 off-target activity. These findings support the use of RAD52 and dn53BP1 co-expression to overcome bottlenecks that limit HDR in precision genome editing.


Asunto(s)
Sistemas CRISPR-Cas , Reparación del ADN , Edición Génica/métodos , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Expresión Génica Ectópica , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Reparación del ADN por Recombinación
14.
Sci Transl Med ; 8(347): 347ra93, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27412785

RESUMEN

C9ORF72 mutations are found in a significant fraction of patients suffering from amyotrophic lateral sclerosis and frontotemporal dementia, yet the function of the C9ORF72 gene product remains poorly understood. We show that mice harboring loss-of-function mutations in the ortholog of C9ORF72 develop splenomegaly, neutrophilia, thrombocytopenia, increased expression of inflammatory cytokines, and severe autoimmunity, ultimately leading to a high mortality rate. Transplantation of mutant mouse bone marrow into wild-type recipients was sufficient to recapitulate the phenotypes observed in the mutant animals, including autoimmunity and premature mortality. Reciprocally, transplantation of wild-type mouse bone marrow into mutant mice improved their phenotype. We conclude that C9ORF72 serves an important function within the hematopoietic system to restrict inflammation and the development of autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/genética , Proteína C9orf72/genética , Animales , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/genética , Autoinmunidad/fisiología , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiología , Citocinas/metabolismo , Leucemia/genética , Leucemia/metabolismo , Ratones , Mutación/genética , Esplenomegalia/genética , Esplenomegalia/inmunología , Trombocitopenia/genética , Trombocitopenia/inmunología
15.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27424808

RESUMEN

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/enzimología , Isocitrato Deshidrogenasa/genética , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Regulación hacia Abajo , Células Madre Hematopoyéticas/citología , Humanos , Isocitrato Deshidrogenasa/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas/metabolismo
16.
Nat Biotechnol ; 34(7): 738-45, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27272386

RESUMEN

Hematopoietic stem cell transplantation (HSCT) offers curative therapy for patients with hemoglobinopathies, congenital immunodeficiencies, and other conditions, possibly including AIDS. Autologous HSCT using genetically corrected cells would avoid the risk of graft-versus-host disease (GVHD), but the genotoxicity of conditioning remains a substantial barrier to the development of this approach. Here we report an internalizing immunotoxin targeting the hematopoietic-cell-restricted CD45 receptor that effectively conditions immunocompetent mice. A single dose of the immunotoxin, CD45-saporin (SAP), enabled efficient (>90%) engraftment of donor cells and full correction of a sickle-cell anemia model. In contrast to irradiation, CD45-SAP completely avoided neutropenia and anemia, spared bone marrow and thymic niches, enabling rapid recovery of T and B cells, preserved anti-fungal immunity, and had minimal overall toxicity. This non-genotoxic conditioning method may provide an attractive alternative to current conditioning regimens for HSCT in the treatment of non-malignant blood diseases.


Asunto(s)
Daño del ADN/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Antígenos Comunes de Leucocito/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Daño del ADN/genética , Femenino , Mejoramiento Genético/métodos , Fenómenos Inmunogenéticos/genética , Inmunotoxinas , Ratones , Ratones Endogámicos C57BL , Saporinas
17.
Stem Cells ; 34(10): 2501-2511, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27335219

RESUMEN

Human mesenchymal stem cells (MSCs) hold great promise in cellular therapeutics for skeletal diseases but lack expression of E-selectin ligands that direct homing of blood-borne cells to bone marrow. Previously, we described a method to engineer E-selectin ligands on the MSC surface by exofucosylating cells with fucosyltransferase VI (FTVI) and its donor sugar, GDP-Fucose, enforcing transient surface expression of the potent E-selectin ligand HCELL with resultant enhanced osteotropism of intravenously administered cells. Here, we sought to determine whether E-selectin ligands created via FTVI-exofucosylation are distinct in identity and function to those created by FTVI expressed intracellularly. To this end, we introduced synthetic modified mRNA encoding FTVI (FUT6-modRNA) into human MSCs. FTVI-exofucosylation (i.e., extracellular fucosylation) and FUT6-modRNA transfection (i.e., intracellular fucosylation) produced similar peak increases in cell surface E-selectin ligand levels, and shear-based functional assays showed comparable increases in tethering/rolling on human endothelial cells expressing E-selectin. However, biochemical analyses revealed that intracellular fucosylation induced expression of both intracellular and cell surface E-selectin ligands and also induced a more sustained expression of E-selectin ligands compared to extracellular fucosylation. Notably, live imaging studies to assess homing of human MSC to mouse calvarium revealed more osteotropism following intravenous administration of intracellularly-fucosylated cells compared to extracellularly-fucosylated cells. This study represents the first direct analysis of E-selectin ligand expression programmed on human MSCs by FTVI-mediated intracellular versus extracellular fucosylation. The observed differential biologic effects of FTVI activity in these two contexts may yield new strategies for improving the efficacy of human MSCs in clinical applications. Stem Cells 2016;34:2501-2511.


Asunto(s)
Huesos/citología , Movimiento Celular , Selectina E/metabolismo , Fucosa/metabolismo , Células Madre Mesenquimatosas/citología , Ingeniería Metabólica/métodos , Animales , Médula Ósea/metabolismo , Línea Celular , Membrana Celular/metabolismo , Espacio Extracelular/metabolismo , Extravasación de Materiales Terapéuticos y Diagnósticos/patología , Fucosiltransferasas/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Humanos , Espacio Intracelular/metabolismo , Cinética , Ligandos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Cráneo/metabolismo , Transfección , Trasplante Heterólogo
18.
Trends Mol Med ; 22(8): 635-637, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27345866

RESUMEN

The hematopoietic system undergoes many changes during aging, but the causes and molecular mechanisms behind these changes are not well understood. Wang et al. have recently implicated a circadian rhythm gene, Per2, as playing a role in the DNA damage response and in the expression of lymphoid genes in aged hematopoietic stem cells.


Asunto(s)
Envejecimiento , Ritmo Circadiano , Daño del ADN , Hematopoyesis , Animales , Regulación de la Expresión Génica , Humanos , Proteínas Circadianas Period/genética
19.
Cell Rep ; 14(12): 2809-18, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26997272

RESUMEN

Homeostasis of short-lived blood cells is dependent on rapid proliferation of immature precursors. Using a conditional histone 2B-mCherry-labeling mouse model, we characterize hematopoietic stem cell (HSC) and progenitor proliferation dynamics in steady state and following several types of induced stress. HSC proliferation following HSC transplantation into lethally irradiated mice is fundamentally different not only from native hematopoiesis but also from other stress contexts. Whereas transplantation promoted sustained, long-term proliferation of HSCs, both cytokine-induced mobilization and acute depletion of selected blood cell lineages elicited very limited recruitment of HSCs to the proliferative pool. By coupling mCherry-based analysis of proliferation history with multiplex gene expression analyses on single cells, we have found that HSCs can be stratified into four distinct subtypes. These subtypes have distinct molecular signatures and differ significantly in their reconstitution potentials, showcasing the power of tracking proliferation history when resolving functional heterogeneity of HSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Mitosis , Animales , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Doxorrubicina/toxicidad , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Plásmidos/genética , Plásmidos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Trasplante Homólogo , Proteína Fluorescente Roja
20.
Exp Hematol ; 43(9): 756-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26143581

RESUMEN

Transplantation of hematopoietic stem cells (HSCs) to treat hematologic disorders is routinely used in the clinic. However, HSC therapy is hindered by the requirements of finding human leukocyte antigen (HLA)-matched donors and attaining sufficient numbers of long-term HSCs in the graft. Therefore, ex vivo expansion of transplantable HSCs remains one of the "holy grails" of hematology. Without the ability to maintain and expand human HSCs in vitro, two complementary approaches involving cellular reprogramming to generate transplantable HSCs have emerged. Reprogrammed HSCs represent a potentially inexhaustible supply of autologous tissue. On March 18th, 2015, Dr. George Q. Daley and Dr. Derrick J. Rossi, two pioneers in the field, presented and discussed their most recent research on these topics in a webinar organized by the International Society for Experimental Hematology (ISEH). Here, we summarize these seminars and discuss the possibilities and challenges in the field of hematopoietic specification.


Asunto(s)
Enfermedades Hematológicas , Células Madre Hematopoyéticas , Células Madre Pluripotentes , Animales , Reprogramación Celular , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/patología , Enfermedades Hematológicas/terapia , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA