Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Int J Hematol ; 119(3): 231-239, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36609840

RESUMEN

Differential diagnosis of pancytopenia with bone marrow (BM) hypoplasia represented by aplastic anemia (AA) is often challenging for physicians, because no laboratory tests have been established, until recently, to distinguish immune-mediated BM failure, which includes acquired AA (aAA) and a subset of low-risk myelodysplastic syndrome (MDS), from non-immune BM failure, which is primarily caused by genetic abnormalities in hematopoietic stem cells (HSCs). HSCs of healthy individuals often undergo somatic mutations, and some acquire phenotypic changes that allow them to escape immune attack against themselves. Once an immune attack against HSCs occurs, HSCs that undergo somatic mutations survive the immune attack and continue to produce their progenies with the same genetic or phenotypic changes. The presence of mature blood cells derived from mutated HSCs in the peripheral blood serves as evidence of the immune-mediated destruction of HSCs. Glycosylphosphatidylinositol-anchored protein-deficient (GPI[-]) blood cells and HLA class I allele-lacking (HLA[-]) leukocytes are two major aberrant cell types that represent the immune mechanism underlying BM failure. This review focuses on the importance of identifying immune mechanisms using laboratory markers, including GPI(-) cells and HLA(-) leukocytes, in the management of BM failure.


Asunto(s)
Anemia Aplásica , Pancitopenia , Humanos , Pancitopenia/metabolismo , Anemia Aplásica/genética , Trastornos de Fallo de la Médula Ósea , Células Madre Hematopoyéticas/metabolismo , Leucocitos
2.
Biomed Pharmacother ; 169: 115863, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37952356

RESUMEN

Acquired aplastic anemia (AA) is a bone marrow failure (BMF) disease, characterized by fatty bone marrow (BM) and BM hypocellularity resulted from auto-immune dysregulated T cells-mediated destruction of BM haemopoietic stem cells (HPSC). The objective of this study was to investigate potential therapeutic effect of irisin, a molecule involved in adipose tissue transition, on AA mouse model. Our results showed that the concentration of irisin in serum was lower in AA patients than in healthy controls, suggesting a role of irisin in the pathogenesis of AA. In the AA mice, irisin administration prolonged the survival rate, prevented or attenuated peripheral pancytopenia, and preserved HPSC in the BM. Moreover, irisin also markedly reduced BM adipogenesis. In vitro results showed that irisin increased both cell proliferation and colony numbers of HPSC. Furthermore, our results demonstrated that irisin upregulated the expression of mitochondrial ATPase Inhibitory Factor 1 (IF1) in HPSC, inhibited the activation of mitochondrial fission protein (DRP1) and enhanced aerobic glycolysis. Taken together, our findings indicate novel roles of irisin in the pathogenesis of AA, and in the protection of HPSC through stimulation of proliferation and regulation of mitochondria function, which provides a proof-of-concept for the application of irisin in AA therapy.


Asunto(s)
Anemia Aplásica , Células Madre Hematopoyéticas , Pancitopenia , Animales , Humanos , Ratones , Anemia Aplásica/patología , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Fibronectinas/metabolismo , Fibronectinas/farmacología , Pancitopenia/metabolismo , Pancitopenia/patología , Células Madre Hematopoyéticas/efectos de los fármacos
3.
Elife ; 122023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37272618

RESUMEN

Ribosomal protein (Rp) gene haploinsufficiency can result in Diamond-Blackfan Anemia (DBA), characterized by defective erythropoiesis and skeletal defects. Some mouse Rp mutations recapitulate DBA phenotypes, although others lack erythropoietic or skeletal defects. We generated a conditional knockout mouse to partially delete Rps12. Homozygous Rps12 deletion resulted in embryonic lethality. Mice inheriting the Rps12KO/+ genotype had growth and morphological defects, pancytopenia, and impaired erythropoiesis. A striking reduction in hematopoietic stem cells (HSCs) and progenitors in the bone marrow (BM) was associated with decreased ability to repopulate the blood system after competitive and non-competitive BM transplantation. Rps12KO/+ lost HSC quiescence, experienced ERK and MTOR activation, and increased global translation in HSC and progenitors. Post-natal heterozygous deletion of Rps12 in hematopoietic cells using Tal1-Cre-ERT also resulted in pancytopenia with decreased HSC numbers. However, post-natal Cre-ERT induction led to reduced translation in HSCs and progenitors, suggesting that this is the most direct consequence of Rps12 haploinsufficiency in hematopoietic cells. Thus, RpS12 has a strong requirement in HSC function, in addition to erythropoiesis.


Asunto(s)
Anemia de Diamond-Blackfan , Pancitopenia , Animales , Ratones , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Eritropoyesis/genética , Genes Esenciales , Haploinsuficiencia , Células Madre Hematopoyéticas/metabolismo , Ratones Noqueados , Pancitopenia/genética , Pancitopenia/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
4.
Haematologica ; 108(10): 2652-2663, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37021532

RESUMEN

Clinical trials have shown that lentiviral-mediated gene therapy can ameliorate bone marrow failure (BMF) in nonconditioned Fanconi anemia (FA) patients resulting from the proliferative advantage of corrected FA hematopoietic stem and progenitor cells (HSPC). However, it is not yet known if gene therapy can revert affected molecular pathways in diseased HSPC. Single-cell RNA sequencing was performed in chimeric populations of corrected and uncorrected HSPC co-existing in the BM of gene therapy-treated FA patients. Our study demonstrates that gene therapy reverts the transcriptional signature of FA HSPC, which then resemble the transcriptional program of healthy donor HSPC. This includes a down-regulated expression of TGF-ß and p21, typically up-regulated in FA HSPC, and upregulation of DNA damage response and telomere maintenance pathways. Our results show for the first time the potential of gene therapy to rescue defects in the HSPC transcriptional program from patients with inherited diseases; in this case, in FA characterized by BMF and cancer predisposition.


Asunto(s)
Anemia de Fanconi , Pancitopenia , Humanos , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Terapia Genética/métodos , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Pancitopenia/metabolismo , Trastornos de Fallo de la Médula Ósea/metabolismo
5.
Blood ; 138(3): 221-233, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34292326

RESUMEN

The Chd8 gene encodes a member of the chromodomain helicase DNA-binding (CHD) family of SNF2H-like adenosine triphosphate (ATP)-dependent chromatin remodeler, the mutations of which define a subtype of autism spectrum disorders. Increasing evidence from recent studies indicates that ATP-dependent chromatin-remodeling genes are involved in the control of crucial gene-expression programs in hematopoietic stem/progenitor cell (HSPC) regulation. In this study, we identified CHD8 as a specific and essential regulator of normal hematopoiesis. Loss of Chd8 leads to severe anemia, pancytopenia, bone marrow failure, and engraftment failure related to a drastic depletion of HSPCs. CHD8 forms a complex with ATM and its deficiency increases chromatin accessibility and drives genomic instability in HSPCs causing an activation of ATM kinase that further stabilizes P53 protein by phosphorylation and leads to increased HSPC apoptosis. Deletion of P53 rescues the apoptotic defects of HSPCs and restores overall hematopoiesis in Chd8-/- mice. Our findings demonstrate that chromatin organization by CHD8 is uniquely necessary for the maintenance of hematopoiesis by integrating the ATM-P53-mediated survival of HSPCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/citología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Supervivencia Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Ratones , Pancitopenia/genética , Pancitopenia/metabolismo , Estabilidad Proteica
6.
Blood ; 137(18): 2450-2462, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33512449

RESUMEN

Inborn errors of immunity (IEI) are a genetically heterogeneous group of disorders with a broad clinical spectrum. Identification of molecular and functional bases of these disorders is important for diagnosis, treatment, and an understanding of the human immune response. We identified 6 unrelated males with neutropenia, infections, lymphoproliferation, humoral immune defects, and in some cases bone marrow failure associated with 3 different variants in the X-linked gene TLR8, encoding the endosomal Toll-like receptor 8 (TLR8). Interestingly, 5 patients had somatic variants in TLR8 with <30% mosaicism, suggesting a dominant mechanism responsible for the clinical phenotype. Mosaicism was also detected in skin-derived fibroblasts in 3 patients, demonstrating that mutations were not limited to the hematopoietic compartment. All patients had refractory chronic neutropenia, and 3 patients underwent allogeneic hematopoietic cell transplantation. All variants conferred gain of function to TLR8 protein, and immune phenotyping demonstrated a proinflammatory phenotype with activated T cells and elevated serum cytokines associated with impaired B-cell maturation. Differentiation of myeloid cells from patient-derived induced pluripotent stem cells demonstrated increased responsiveness to TLR8. Together, these findings demonstrate that gain-of-function variants in TLR8 lead to a novel childhood-onset IEI with lymphoproliferation, neutropenia, infectious susceptibility, B- and T-cell defects, and in some cases, bone marrow failure. Somatic mosaicism is a prominent molecular mechanism of this new disease.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/patología , Mutación con Ganancia de Función , Síndromes de Inmunodeficiencia/patología , Inflamación/patología , Mosaicismo , Pancitopenia/patología , Receptor Toll-Like 8/genética , Adolescente , Adulto , Linfocitos B/patología , Trastornos de Fallo de la Médula Ósea/etiología , Trastornos de Fallo de la Médula Ósea/metabolismo , Diferenciación Celular , Niño , Preescolar , Citocinas/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Síndromes de Inmunodeficiencia/etiología , Síndromes de Inmunodeficiencia/metabolismo , Lactante , Inflamación/etiología , Inflamación/metabolismo , Activación de Linfocitos , Masculino , Pancitopenia/etiología , Pancitopenia/metabolismo , Linaje , Pronóstico , Linfocitos T/inmunología , Adulto Joven
7.
Sci Rep ; 10(1): 22211, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335275

RESUMEN

The only available option to treat radiation-induced hematopoietic syndrome is allogeneic hematopoietic cell transplantation, a therapy unavailable to many patients undergoing treatment for malignancy, which would also be infeasible in a radiological disaster. Stromal cells serve as critical components of the hematopoietic stem cell niche and are thought to protect hematopoietic cells under stress. Prior studies that have transplanted mesenchymal stromal cells (MSCs) without co-administration of a hematopoietic graft have shown underwhelming rescue of endogenous hematopoiesis and have delivered the cells within 24 h of radiation exposure. Herein, we examine the efficacy of a human bone marrow-derived MSC therapy delivered at 3 h or 30 h in ameliorating radiation-induced hematopoietic syndrome and show that pancytopenia persists despite MSC therapy. Animals exposed to radiation had poorer survival and experienced loss of leukocytes, platelets, and red blood cells. Importantly, mice that received a therapeutic dose of MSCs were significantly less likely to die but experienced equivalent collapse of the hematopoietic system. The cause of the improved survival was unclear, as complete blood counts, splenic and marrow cellularity, numbers and function of hematopoietic stem and progenitor cells, and frequency of niche cells were not significantly improved by MSC therapy. Moreover, human MSCs were not detected in the bone marrow. MSC therapy reduced crypt dropout in the small intestine and promoted elevated expression of growth factors with established roles in gut development and regeneration, including PDGF-A, IGFBP-3, IGFBP-2, and IGF-1. We conclude that MSC therapy improves survival not through overt hematopoietic rescue but by positive impact on other radiosensitive tissues, such as the intestinal mucosa. Collectively, these data reveal that MSCs could be an effective countermeasure in cancer patients and victims of nuclear accidents but that MSCs alone do not significantly accelerate or contribute to recovery of the blood system.


Asunto(s)
Hematopoyesis/efectos de la radiación , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Traumatismos por Radiación/mortalidad , Traumatismos por Radiación/terapia , Animales , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Inmunofenotipificación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Células Madre Mesenquimatosas/citología , Pancitopenia/etiología , Pancitopenia/metabolismo , Pancitopenia/patología , Pronóstico , Traumatismos por Radiación/patología , Radioterapia/efectos adversos , Resultado del Tratamiento
8.
J Immunol Res ; 2020: 4098235, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32149157

RESUMEN

OBJECTIVES: To explore effects of Epstein-Barr virus (EBV) infection on CD19+ B lymphocytes in patients with immunorelated pancytopenia (IRP). METHODS: An enzyme-linked immunosorbent assay (ELISA) in vitro diagnostic kit was used to detect EBV capsid antigen- (CA-) IgG and VCA-IgM antibodies in the serum. We analyzed the EBV-DNA copies of CD19+ B lymphocyte by using real-time quantitative polymerase chain reaction (RT-qPCR). CD21, CD23, CD5, CD80, and CD86 receptors on the surfaces of CD19+ B cells were detected by flow cytometry (FCM). The correlation between these receptors and EBV-DNA copies were evaluated. RESULTS: The results revealed that the positive rate of EBVCA-IgM and CD19+ B lymphocyte EBV-DNA copy in the IRP group were significantly higher than those in the control group (P < 0.05). CD19+ B lymphocyte EBV-DNA copies were also more abundant in IRP patients than in control subjects (P < 0.05). CD19+ B lymphocyte EBV-DNA copies were also more abundant in IRP patients than in control subjects (P < 0.05). CD19+ B lymphocyte EBV-DNA copies were also more abundant in IRP patients than in control subjects (. CONCLUSIONS: EBV infection may activate CD19+ B lymphocytes and further disrupt bone marrow hematopoiesis in IRP patients.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4 , Pancitopenia/etiología , Adolescente , Adulto , Anciano , Antígenos CD19/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/diagnóstico , Infecciones por Virus de Epstein-Barr/virología , Femenino , Humanos , Inmunoglobulina M/sangre , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Pancitopenia/sangre , Pancitopenia/metabolismo , Pronóstico , Adulto Joven
9.
EBioMedicine ; 53: 102677, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32114389

RESUMEN

BACKGROUND: Endothelial cells (ECs) function as an instructive platform to support haematopoietic stem cell (HSC) homeostasis. Our recent studies found that impaired bone marrow (BM) ECs are responsible for the defective haematopoiesis in patients with poor graft function (PGF), which is characterised by pancytopenia post-allotransplant. Although activated autophagy was reported to benefit ECs, whether EC autophagy plays a critical role in supporting HSCs and its effect on PGF patients post-allotransplant remain unclear. METHODS: To evaluate whether the autophagy status of ECs modulates their ability to support haematopoiesis, human umbilical vein endothelial cells (HUVECs) and primary BM ECs derived from healthy donors were subjected to knockdown or overexpression of Beclin-1 (an autophagy-related protein). Moreover, BM ECs derived from PGF patients were studied. FINDINGS: Beclin-1 knockdown significantly reduced the haematopoiesis-supporting ability of ECs by suppressing autophagy, which could be restored by activating autophagy via Beclin-1 upregulation. Moreover, autophagy positively regulated haematopoiesis-related genes in HUVECs. Subsequently, a prospective case-control study demonstrated that defective autophagy reduced Beclin-1 expression and the colony-forming unit (CFU) plating efficiency in BM ECs from PGF patients compared to matched patients with good graft function. Rapamycin, an autophagy activator, quantitatively and functionally improved BM ECs from PGF patients in vitro and enhanced their ability to support HSCs by activating the Beclin-1 pathway. INTERPRETATION: Our results suggest that the autophagy status of ECs modulates their ability to support haematopoiesis by regulating the Beclin-1 pathway. Defective autophagy in BM ECs may be involved in the pathogenesis of PGF post-allotransplant. Rapamycin provides a promising therapeutic approach for PGF patients. FUNDING: Please see funding sources.


Asunto(s)
Autofagia , Endotelio Vascular/metabolismo , Hematopoyesis , Pancitopenia/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Transfusión de Sangre Autóloga/efectos adversos , Células Cultivadas , Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Monocitos/citología , Monocitos/metabolismo , Pancitopenia/etiología
10.
J Pediatr Hematol Oncol ; 42(7): e670-e672, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31593008

RESUMEN

Alterations in copper homeostasis is an uncommon cause for hematologic alterations frequently presenting with dysplastic features in the bone marrow. Most of these alterations have been documented in adult patients with copper deficiency. Rare cases show hematogone hyperplasia in these patients. Effects of mild copper excess have not been documented in literature. We are describing a pediatric patient who presented with pancytopenia associated with hypercupraemia (excess of copper). Bone marrow examination showed hematogone hyperplasia. Interestingly, correction of serum copper levels with zinc therapy lead to complete improvement in pancytopenia. Hematogones had also reduced in subsequent marrow biopsy after therapy.


Asunto(s)
Cobre/metabolismo , Enfermedades Hematológicas/diagnóstico , Hiperplasia/diagnóstico , Pancitopenia/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Niño , Enfermedades Hematológicas/complicaciones , Enfermedades Hematológicas/metabolismo , Humanos , Hiperplasia/complicaciones , Hiperplasia/metabolismo , Masculino , Pancitopenia/complicaciones , Pancitopenia/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Pronóstico
11.
Clin Liver Dis ; 23(4): 781-807, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31563222

RESUMEN

Idiopathic portal hypertension (IPH) and extrahepatic portal venous obstruction (EHPVO) are prototype noncirrhotic causes of portal hypertension (PHT), characterized by normal hepatic venous pressure gradient, variceal bleeds, and moderate to massive splenomegaly with preserved liver synthetic functions. Infections, toxins, and immunologic, prothrombotic and genetic disorders are possible causes in IPH, whereas prothrombotic and local factors around the portal vein lead to EHPVO. Growth failure, portal biliopathy, and minimal hepatic encephalopathy are long-term concerns in EHPVO. Surgical shunts and transjugular intrahepatic portosystemic shunt resolve the complications secondary to PHT. Meso-Rex shunt is now the standard-of-care surgery in children with EHPVO.


Asunto(s)
Hipertensión Portal/fisiopatología , Cirrosis Hepática/fisiopatología , Pancitopenia/fisiopatología , Vena Porta , Esplenomegalia/fisiopatología , Trombosis de la Vena/fisiopatología , Animales , Enfermedades de las Vías Biliares/etiología , Enfermedades de las Vías Biliares/terapia , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Várices Esofágicas y Gástricas , Hemorragia Gastrointestinal/etiología , Hemorragia Gastrointestinal/prevención & control , Hemorragia Gastrointestinal/terapia , Trastornos del Crecimiento/etiología , Encefalopatía Hepática/etiología , Encefalopatía Hepática/terapia , Humanos , Hipertensión Portal/complicaciones , Hipertensión Portal/diagnóstico , Hipertensión Portal/etiología , Hipertensión Portal/metabolismo , Hipertensión Portal/terapia , Cirrosis Hepática/complicaciones , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/metabolismo , Trasplante de Hígado , Metabolómica , Pancitopenia/complicaciones , Pancitopenia/diagnóstico , Pancitopenia/metabolismo , Derivación Portosistémica Quirúrgica , Derivación Portosistémica Intrahepática Transyugular , Calidad de Vida , Esplenomegalia/complicaciones , Esplenomegalia/diagnóstico , Esplenomegalia/metabolismo , Transcriptoma , Trombosis de la Vena/complicaciones , Trombosis de la Vena/diagnóstico , Trombosis de la Vena/metabolismo , Hipertensión Portal Idiopática no Cirrótica
12.
Mol Med Rep ; 20(5): 3979-3990, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31545490

RESUMEN

In the past decade, a group of cases with persisting haemocytopenia were separated from those with idiopathic cytopenia of undetermined significance due to the optimal response of these patients to immunosuppression therapy and due to the detection of autoantibodies in the bone marrow of haemopoietic cells. This condition was termed immune­related haemocytopenia (IRH). However, the quantity of T lymphocytes remained unknown. In the present study, the percentage of CD4+ T­cell subsets and related cytokines was measured using flow cytometry and an enzyme­linked immunosorbent assay. An abnormal number of CD4+ T cell subsets was found, including increased percentages of T helper (Th)2, Th9 and Th17 cells and a decreased number of regulatory T (Treg) cells. In addition, the results showed downregulation in the levels of interleukin (IL)­2, transforming growth factor­ß and IL­35, and upregulation in the levels of IL­4, IL­6, IL­17, IL­23 and interferon­Î³ in patients who did not receive therapy (untreated patients). These levels were significantly associated with the number of peripheral blood cells and were recovered following treatment. In conclusion, an abnormal number of CD4+ T cell subsets and corresponding abnormal levels of regulatory cytokines resulted in the stimulation of B1 lymphocytes to produce autoantibodies in IRH, which may be considered as markers to evaluate disease prognosis and treatment strategies.


Asunto(s)
Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/genética , Regulación de la Expresión Génica , Pancitopenia/etiología , Pancitopenia/metabolismo , Adolescente , Adulto , Anciano , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Niño , Citocinas/biosíntesis , Manejo de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pancitopenia/diagnóstico , Pancitopenia/terapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
14.
Haematologica ; 104(11): 2164-2177, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30923091

RESUMEN

Loss-of-function studies have determined that Notch signaling is essential for hematopoietic and endothelial development. By deleting a single allele of the Notch1 transcriptional activation domain we generated viable, post-natal mice exhibiting hypomorphic Notch signaling. These heterozygous mice, which lack only one copy of the transcriptional activation domain, appear normal and have no endothelial or hematopoietic phenotype, apart from an inherent, cell-autonomous defect in T-cell lineage development. Following chemotherapy, these hypomorphs exhibited severe pancytopenia, weight loss and morbidity. This phenotype was confirmed in an endothelial-specific, loss-of-function Notch1 model system. Ang1, secreted by hematopoietic progenitors after damage, activated endothelial Tie2 signaling, which in turn enhanced expression of Notch ligands and potentiated Notch1 receptor activation. In our heterozygous, hypomorphic model system, the mutant protein that lacks the Notch1 transcriptional activation domain accumulated in endothelial cells and interfered with optimal activity of the wildtype Notch1 transcriptional complex. Failure of the hypomorphic mutant to efficiently drive transcription of key gene targets such as Hes1 and Myc prolonged apoptosis and limited regeneration of the bone marrow niche. Thus, basal Notch1 signaling is sufficient for niche development, but robust Notch activity is required for regeneration of the bone marrow endothelial niche and hematopoietic recovery.


Asunto(s)
Microambiente Celular , Células Endoteliales/fisiología , Receptor Notch1/metabolismo , Receptor TIE-2/metabolismo , Regeneración , Transducción de Señal , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Microambiente Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fluorouracilo/farmacología , Rayos gamma/efectos adversos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Noqueados , Pancitopenia/etiología , Pancitopenia/metabolismo , Pancitopenia/patología , Transducción de Señal/efectos de los fármacos
17.
Sci Rep ; 8(1): 14133, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237523

RESUMEN

Kidney proximal tubular cells (PTCs) are highly specialized for ultrafiltrate reabsorption and serve as paradigm of apical epithelial differentiation. Vps34/PI3-kinase type III (PI3KC3) regulates endosomal dynamics, macroautophagy and lysosomal function. However, its in vivo role in PTCs has not been evaluated. Conditional deletion of Vps34/PI3KC3 in PTCs by Pax8-Cre resulted in early (P7) PTC dysfunction, manifested by Fanconi-like syndrome, followed by kidney failure (P14) and death. By confocal microscopy, Vps34∆/∆ PTCs showed preserved apico-basal specification (brush border, NHERF-1 versus Na+/K+-ATPase, ankyrin-G) but basal redistribution of late-endosomes/lysosomes (LAMP-1) and mis-localization to lysosomes of apical recycling endocytic receptors (megalin, cubilin) and apical non-recycling solute carriers (NaPi-IIa, SGLT-2). Defective endocytosis was confirmed by Texas-red-ovalbumin tracing and reduced albumin content. Disruption of Rab-11 and perinuclear galectin-3 compartments suggested mechanistic clues for defective receptor recycling and apical biosynthetic trafficking. p62-dependent autophagy was triggered yet abortive (p62 co-localization with LC3 but not LAMP-1) and PTCs became vacuolated. Impaired lysosomal positioning and blocked autophagy are known causes of cell stress. Thus, early trafficking defects show that Vps34 is a key in vivo component of molecular machineries governing apical vesicular trafficking, thus absorptive function in PTCs. Functional defects underline the essential role of Vps34 for PTC homeostasis and kidney survival.


Asunto(s)
Autofagia/genética , Fosfatidilinositol 3-Quinasas Clase III/genética , Hipersensibilidad Tardía/genética , Síndromes de Inmunodeficiencia/genética , Túbulos Renales Proximales/metabolismo , Pancitopenia/genética , Insuficiencia Renal/genética , Neoplasias Cutáneas/genética , Animales , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endocitosis/fisiología , Endosomas/metabolismo , Hipersensibilidad Tardía/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Ratones , Ratones Noqueados , Pancitopenia/metabolismo , Transporte de Proteínas , Insuficiencia Renal/metabolismo , Neoplasias Cutáneas/metabolismo
18.
Clin Lab ; 64(6): 1041-1048, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29945307

RESUMEN

BACKGROUND: Immuno-related hemocytopenia (IRH) is defined as idiopathic cytopenia of undetermined significance (ICUS) patients with autoantibodies. In our previous studies, we found that IgG1 levels were increased in IRH patients and might cause the destruction of hematopoietic cells. METHODS: In this study, we analyzed IgG subclasses in 30 IRH patients (male:female = 13:17, median age 32 years, range 18 - 56), 15 IRH remission patients (IRH-R) (male:female = 6:9, median age 34, range 20 - 52) and 20 normal controls (male:female = 8:12, median age 27, range 24 - 36) by Cytometric Bead Array, Flow Cytometry and Immunohistochemical staining. RESULTS: Levels of IgG1/IgG3 in the bone marrow supernatant of IRH patents, as well as the proportion of CD5+ B lymphocytes and Th2 cells (CD3+CD8-IL-4+) were higher than those of IRH-R patients and normal controls, and IgG1 levels had a positive correlation with the proportion of Th2 cells. In IRH patients, IgG1 and IgG3 were positive on nucleated erythrocytes and granulocytes, which were negative in IRH-R patients and healthy controls and had inverse correlations with hematopoietic function. Using immunohistochemical staining, IgG1 were also detected on bone marrow biopsies of IRH patients. CONCLUSIONS: The results indicated that IgG1 and IgG3 autoantibodies in IRH patients might play a key role in the IRH pathogenesis and in the abnormal immune function of IRH patients.


Asunto(s)
Autoanticuerpos/sangre , Inmunoglobulina G/sangre , Pancitopenia/sangre , Adolescente , Adulto , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Niño , Eritrocitos/inmunología , Eritrocitos/metabolismo , Femenino , Citometría de Flujo , Granulocitos/inmunología , Granulocitos/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Pancitopenia/inmunología , Pancitopenia/metabolismo , Adulto Joven
19.
Am J Pathol ; 188(6): 1457-1468, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29574182

RESUMEN

The fundamental structure of eukaryotic cell plasma membrane is the phospholipid bilayer, which contains four major phospholipids. These phospholipids are asymmetrically distributed between the outer and inner leaflets. P4-ATPase flippase complexes play essential roles in ensuring this asymmetry. We found that conditional deletion of Tmem30a, the ß subunit of P4-ATPase flippase complex, caused pancytopenia in mice. Tmem30a deficiency resulted in depletion of lineage-committed blood cells in the peripheral blood, spleen, and bone marrow. Ablation of Tmem30a also caused the depletion of hematopoietic stem cells (HSCs). HSC RNA sequencing results revealed that multiple biological processes and signal pathways were involved in the event, including mammalian target of rapamycin signaling, genes for HSC stemness, and genes responding to interferons. Our results also revealed that targeting Tmem30a signaling had therapeutic utility in BCR/ABL1-induced chronic myeloid leukemia.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de la Membrana/fisiología , Pancitopenia/patología , Proteínas Proto-Oncogénicas c-bcr/metabolismo , Animales , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Ratones Noqueados , Pancitopenia/etiología , Pancitopenia/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...