Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood Cells Mol Dis ; 105: 102824, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38262104

RESUMEN

In preparation for hematopoietic stem cell mobilization and collection, current ex vivo gene therapy protocols for sickle cell disease require patients to undergo several months of chronic red cell transfusion. For health care equity, alternatives to red cell transfusion should be available. We examined whether treatment with GBT1118, the murine analog of voxelotor, could be a safe and feasible alternative to red cell transfusion. We found that 3 weeks of treatment with GBT1118 increased the percentage of bone marrow hematopoietic stem cells and upon plerixafor mobilization, the percentage of peripheral blood hematopoietic stem cells. Our data suggest that voxelotor should be further explored for its potential safety and utility as preparation for hematopoietic stem cell mobilization and collection.


Asunto(s)
Anemia de Células Falciformes , Benzaldehídos , Trasplante de Células Madre Hematopoyéticas , Compuestos Heterocíclicos , Niacinamida/análogos & derivados , Pirazinas , Humanos , Ratones , Animales , Movilización de Célula Madre Hematopoyética/métodos , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/uso terapéutico , Compuestos Heterocíclicos/farmacología , Pirazoles , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/metabolismo , Terapia Genética/efectos adversos , Factor Estimulante de Colonias de Granulocitos/farmacología
2.
Blood ; 143(11): 1018-1031, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38127913

RESUMEN

ABSTRACT: Disordered erythropoiesis is a feature of many hematologic diseases, including sickle cell disease (SCD). However, very little is known about erythropoiesis in SCD. Here, we show that although bone marrow (BM) erythroid progenitors and erythroblasts in Hbbth3/+ thalassemia mice were increased more than twofold, they were expanded by only ∼40% in Townes sickle mice (SS). We further show that the colony-forming ability of SS erythroid progenitors was decreased and erythropoietin (EPO)/EPO receptor (EPOR) signaling was impaired in SS erythroid cells. Furthermore, SS mice exhibited reduced responses to EPO. Injection of mice with red cell lysates or hemin, mimicking hemolysis in SCD, led to suppression of erythropoiesis and reduced EPO/EPOR signaling, indicating hemolysis, a hallmark of SCD, and could contribute to the impaired erythropoiesis in SCD. In vitro hemin treatment did not affect Stat5 phosphorylation, suggesting that hemin-induced erythropoiesis suppression in vivo is via an indirect mechanism. Treatment with interferon α (IFNα), which is upregulated by hemolysis and elevated in SCD, led to suppression of mouse BM erythropoiesis in vivo and human erythropoiesis in vitro, along with inhibition of Stat5 phosphorylation. Notably, in sickle erythroid cells, IFN-1 signaling was activated and the expression of cytokine inducible SH2-containing protein (CISH), a negative regulator of EPO/EPOR signaling, was increased. CISH deletion in human erythroblasts partially rescued IFNα-mediated impairment of cell growth and EPOR signaling. Knocking out Ifnar1 in SS mice rescued the defective BM erythropoiesis and improved EPO/EPOR signaling. Our findings identify an unexpected role of hemolysis on the impaired erythropoiesis in SCD through inhibition of EPO/EPOR signaling via a heme-IFNα-CISH axis.


Asunto(s)
Anemia de Células Falciformes , Eritropoyesis , Ratones , Animales , Humanos , Eritropoyesis/fisiología , Factor de Transcripción STAT5/metabolismo , Hemólisis , Hemina/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Anemia de Células Falciformes/complicaciones
3.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37490346

RESUMEN

Sickle cell disease (SCD) is a hereditary hemoglobinopathy characterized by painful vaso-occlusive crises (VOC) and chronic hemolysis. The mononuclear phagocyte system is pivotal to SCD pathophysiology, but the mechanisms governing monocyte/macrophage differentiation remain unknown. This study examined the influence of hemolysis on circulating monocyte trajectories in SCD. We discovered that hemolysis stimulated CSF-1 production, partly by endothelial cells via Nrf2, promoting classical monocyte (CMo) differentiation into blood patrolling monocytes (PMo) in SCD mice. However, hemolysis also upregulated CCL-2 through IFN-I, inducing CMo transmigration and differentiation into tissue monocyte-derived macrophages. Blocking CMo transmigration by anti-P selectin antibody in SCD mice increased circulating PMo, corroborating that CMo-to-tissue macrophage differentiation occurs at the expense of CMo-to-blood PMo differentiation. We observed a positive correlation between plasma CSF-1/CCL-2 ratios and blood PMo levels in patients with SCD, underscoring the clinical significance of these two opposing factors in monocyte differentiation. Combined treatment with CSF-1 and anti-P selectin antibody more effectively increased PMo numbers and reduced stasis compared with single-agent therapies in SCD mice. Altogether, these data indicate that monocyte fates are regulated by the balance between two heme pathways, Nrf2/CSF-1 and IFN-I/CCL-2, and suggest that the CSF-1/CCL-2 ratio may present a diagnostic and therapeutic target in SCD.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Ratones , Animales , Hemólisis , Monocitos/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Células Endoteliales/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/tratamiento farmacológico , Enfermedades Vasculares/metabolismo , Diferenciación Celular , Selectinas/metabolismo , Selectinas/uso terapéutico
4.
Blood Adv ; 7(4): 649-663, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35977077

RESUMEN

Sickle red blood cells (RBCs) represent a naturally existing host-cell resistance mechanism to hemoparasite infections. We investigate the basis of this resistance using Babesia divergens grown in sickle (SS) and sickle trait (AS) cells. We found that oxygenation and its corresponding effect on RBC sickling, frequency of fetal hemoglobin positive (HbF+) cells, cellular redox environment, and parasite proliferation dynamics, all played a role in supporting or inhibiting Babesia proliferation. To identify cellular determinants that supported infection, an image flow cytometric tool was developed that could identify sickled cells and constituent Hb. We showed that hypoxic conditions impaired parasite growth in both SS and AS cells. Furthermore, cell sickling was alleviated by oxygenation (hyperoxic conditions), which decreased inhibition of parasite proliferation in SS cells. Interestingly, our tool identified HbF+-SS as host-cells of choice under both hypoxic and hyperoxic conditions, which was confirmed using cord RBCs containing high amounts of HbF+ cells. Uninfected SS cells showed a higher reactive oxygen species-containing environment, than AA or AS cells, which was further perturbed on infection. In hostile SS cells we found that Babesia alters its subpopulation structure, with 1N dominance under hypoxic conditions yielding to equivalent ratios of all parasite forms at hyperoxic conditions, favorable for growth. Multiple factors, including oxygenation and its impact on cell shape, HbF positivity, redox status, and parasite pleiotropy allow Babesia propagation in sickle RBCs. Our studies provide a cellular and molecular basis of natural resistance to Babesia, which will aid in defining novel therapies against human babesiosis.


Asunto(s)
Anemia de Células Falciformes , Babesia , Babesiosis , Parásitos , Animales , Humanos , Babesiosis/parasitología , Eritrocitos/parasitología , Eritrocitos Anormales , Babesia/fisiología , Hipoxia
5.
Curr Opin Hematol ; 28(6): 424-430, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34232141

RESUMEN

PURPOSE OF REVIEW: Platelet transfusion can be life-saving but carries a risk of infection or alloimmunization and is limited by insufficient donor sources and restricted unit shelf life. Generating sufficient platelets in vitro to replace a unit of collected blood remains a challenge. Here, we examine the latest advances in the regulation of megakaryocyte maturation and expansion along with platelet formation and survival. We also discuss alternative therapies investigated to induce platelet production. RECENT FINDINGS: Recent studies examined candidate niche cells in the bone marrow microenvironment for promoting platelet formation and developed an explant-based bioreactor to enhance platelet production ex vivo. Chemical inhibitors were examined for their ability to promote megakaryocyte maturation and expansion. Microparticles from megakaryocytes or platelets were found to improve megakaryocyte maturation and platelet formation. Membrane budding was identified as a novel mode of platelet formation. Lastly, a chemical inhibitor to improve cold-stored platelets was identified. SUMMARY: Recent advances in the regulation of megakaryocyte expansion and platelet production provide exciting promise for the development of improved approaches to generate platelets in vitro. These findings bring the field one step closer to achieving the ultimate goal of creating a unit of platelets without the need for donation.


Asunto(s)
Plaquetas , Megacariocitos , Trombopoyesis , Médula Ósea , Humanos , Transfusión de Plaquetas
6.
Blood ; 138(24): 2570-2582, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34329381

RESUMEN

Sickle cell disease (SCD) is characterized by hemolytic anemia, which can trigger oxidative stress, inflammation, and tissue injury that contribute to disease complications. Bone marrow mesenchymal stromal cells (MSCs) tightly regulate hematopoietic stem cell (HSC) homeostasis in health and disease, but their functionality in SCD remains unclear. We identified for the first time that murine SCD MSCs have altered gene signatures, reduced stem cell properties, and increased oxidative stress, due in part to hemolysis. Murine SCD MSCs had lower HSC maintenance ability in vitro and in vivo, as manifested by increased HSC mobilization and decreased HSC engraftment after transplant. Activation of Toll-like receptor-4 through p65 in MSCs further contributed to MSC dysfunction. Transfusions led to an improved MSC and HSC oxidative state in SCD mice. Improving the regulation between MSCs and HSCs has vital implications for enhancing clinical HSC transplantation and gene therapy outcomes and for identification of new molecular targets for alleviating SCD complications.


Asunto(s)
Anemia de Células Falciformes/patología , Células Madre Hematopoyéticas/patología , Células Madre Mesenquimatosas/patología , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/terapia , Animales , Transfusión Sanguínea , Femenino , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hemólisis , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Estrés Oxidativo , Transcriptoma
7.
Blood ; 138(20): 1986-1997, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34098576

RESUMEN

The erythropoietin receptor (EpoR) has traditionally been thought of as an erythroid-specific gene. Notably, accumulating evidence suggests that EpoR is expressed well beyond erythroid cells. However, the expression of EpoR in non-erythroid cells has been controversial. In this study, we generated EpoR-tdTomato-Cre mice and used them to examine the expression of EpoR in tissue macrophages and hematopoietic cells. We show that in marked contrast to the previously available EpoR-eGFPcre mice, in which a very weak eGFP signal was detected in erythroid cells, tdTomato was readily detectable in both fetal liver (FL) and bone marrow (BM) erythroid cells at all developmental stages and exhibited dynamic changes during erythropoiesis. Consistent with our recent finding that erythroblastic island (EBI) macrophages are characterized by the expression of EpoR, tdTomato was readily detected in both FL and BM EBI macrophages. Moreover, tdTomato was also detected in subsets of hematopoietic stem cells, progenitors, megakaryocytes, and B cells in BM as well as in spleen red pulp macrophages and liver Kupffer cells. The expression of EpoR was further shown by the EpoR-tdTomato-Cre-mediated excision of the floxed STOP sequence. Importantly, EPO injection selectively promoted proliferation of the EpoR-expressing cells and induced erythroid lineage bias during hematopoiesis. Our findings imply broad roles for EPO/EpoR in hematopoiesis that warrant further investigation. The EpoR-tdTomato-Cre mouse line provides a powerful tool to facilitate future studies on EpoR expression and regulation in various non-hematopoietic cells and to conditionally manipulate gene expression in EpoR-expressing cells for functional studies.


Asunto(s)
Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Macrófagos/metabolismo , Receptores de Eritropoyetina/genética , Animales , Células Madre Hematopoyéticas/citología , Humanos , Integrasas/análisis , Integrasas/genética , Sustancias Luminiscentes/análisis , Sustancias Luminiscentes/metabolismo , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Macrófagos/citología , Ratones , Receptores de Eritropoyetina/análisis , Proteína Fluorescente Roja
8.
JCI Insight ; 4(16)2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31434805

RESUMEN

The complex process of platelet formation originates with the hematopoietic stem cell, which differentiates through the myeloid lineage, matures, and releases proplatelets into the BM sinusoids. How formed platelets maintain a low basal activation state in the circulation remains unknown. We identify Lepr+ stromal cells lining the BM sinusoids as important contributors to sustaining low platelet activation. Ablation of murine Lepr+ cells led to a decreased number of platelets in the circulation with an increased activation state. We developed a potentially novel culture system for supporting platelet formation in vitro using a unique population of CD51+PDGFRα+ perivascular cells, derived from human umbilical cord tissue, which display numerous mesenchymal stem cell (MSC) properties. Megakaryocytes cocultured with MSCs had altered LAT and Rap1b gene expression, yielding platelets that are functional with low basal activation levels, a critical consideration for developing a transfusion product. Identification of a regulatory cell that maintains low baseline platelet activation during thrombopoiesis opens up new avenues for improving blood product production ex vivo.


Asunto(s)
Plaquetas/fisiología , Células Madre Mesenquimatosas/fisiología , Activación Plaquetaria , Trombopoyesis/fisiología , Animales , Antígenos CD34 , Plaquetas/inmunología , Plaquetas/metabolismo , Técnicas de Cocultivo , Sangre Fetal , Humanos , Ratones , Ratones Transgénicos , Receptores de Leptina/genética , Receptores de Leptina/fisiología
9.
Blood ; 134(5): 480-491, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31101625

RESUMEN

The erythroblastic island (EBI), composed of a central macrophage and surrounding erythroid cells, was the first hematopoietic niche discovered. The identity of EBI macrophages has thus far remained elusive. Given that Epo is essential for erythropoiesis and that Epor is expressed in numerous nonerythroid cells, we hypothesized that EBI macrophages express Epor so that Epo can act on both erythroid cells and EBI macrophages simultaneously to ensure efficient erythropoiesis. To test this notion, we used Epor-eGFPcre knockin mouse model. We show that in bone marrow (BM) and fetal liver, a subset of macrophages express Epor-eGFP. Imaging flow cytometry analyses revealed that >90% of native EBIs comprised F4/80+Epor-eGFP+ macrophages. Human fetal liver EBIs also comprised EPOR+ macrophages. Gene expression profiles of BM F4/80+Epor-eGFP+ macrophages suggest a specialized function in supporting erythropoiesis. Molecules known to be important for EBI macrophage function such as Vcam1, CD169, Mertk, and Dnase2α were highly expressed in F4/80+Epor-eGFP+ macrophages compared with F4/80+Epor-eGFP- macrophages. Key molecules involved in iron recycling were also highly expressed in BM F4/80+Epor-eGFP+ macrophages, suggesting that EBI macrophages may provide an iron source for erythropoiesis within this niche. Thus, we have characterized EBI macrophages in mouse and man. Our findings provide important resources for future studies of EBI macrophage function during normal as well as disordered erythropoiesis in hematologic diseases such as thalassemia, polycythemia vera, and myelodysplastic syndromes.


Asunto(s)
Eritroblastos/metabolismo , Perfilación de la Expresión Génica , Macrófagos/metabolismo , Transcriptoma , Animales , Biomarcadores , Biología Computacional/métodos , Eritropoyesis/genética , Expresión Génica , Humanos , Inmunofenotipificación , Ratones , Monocitos/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Nicho de Células Madre/genética , Estrés Fisiológico
10.
Blood ; 134(7): 579-590, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31076443

RESUMEN

Painful vaso-occlusive crisis (VOC) is the most common complication of sickle cell disease (SCD). Increasing evidence suggests that vaso-occlusion is initiated by increased adherence of sickle red blood cells (RBCs) to the vascular endothelium. Thus, the mechanisms that remove endothelial-attached sickle RBCs from the microvasculature are expected to be critical for optimal blood flow and prevention of VOC in SCD. We hypothesized that patrolling monocytes (PMos), which protect against vascular damage by scavenging cellular debris, could remove endothelial-adherent sickle RBCs and ameliorate VOC in SCD. We detected RBC (GPA+)-engulfed material in circulating PMos of patients with SCD, and their frequency was further increased during acute crisis. RBC uptake by PMos was specific to endothelial-attached sickle, but not control, RBCs and occurred mostly through ICAM-1, CD11a, and CD18. Heme oxygenase 1 induction, by counteracting the cytotoxic effects of engulfed RBC breakdown products, increased PMo viability. In addition, transfusions, by lowering sickle RBC uptake, improved PMo survival. Selective depletion of PMos in Townes sickle mice exacerbated vascular stasis and tissue damage, whereas treatment with muramyl dipeptide (NOD2 ligand), which increases PMo mass, reduced stasis and SCD associated organ damage. Altogether, these data demonstrate a novel mechanism for removal of endothelial attached sickle RBCs mediated by PMos that can protect against VOC pathogenesis, further supporting PMos as a promising therapeutic target in SCD VOC.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Endotelio Vascular/patología , Eritrocitos/patología , Monocitos/citología , Enfermedades Vasculares/etiología , Anemia de Células Falciformes/patología , Animales , Adhesión Celular , Línea Celular , Humanos , Ratones , Monocitos/patología , Enfermedades Vasculares/patología
12.
Blood Adv ; 2(23): 3462-3478, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30518538

RESUMEN

The intraerythrocytic parasite Babesia microti is the number 1 cause of transfusion-transmitted infection and can induce serious, often life-threatening complications in immunocompromised individuals including transfusion-dependent patients with sickle cell disease (SCD). Despite the existence of strong long-lasting immunological protection against a second infection in mouse models, little is known about the cell types or the kinetics of protective adaptive immunity mounted following Babesia infection, especially in infection-prone SCD that are thought to have an impaired immune system. Here, we show, using a mouse B microti infection model, that infected wild-type (WT) mice mount a very strong adaptive immune response, characterized by (1) coordinated induction of a robust germinal center (GC) reaction; (2) development of follicular helper T (TFH) cells that comprise ∼30% of splenic CD4+ T cells at peak expansion by 10 days postinfection; and (3) high levels of effector T-cell cytokines, including interleukin 21 and interferon γ, with an increase in the secretion of antigen (Ag)-specific antibodies (Abs). Strikingly, the Townes SCD mouse model had significantly lower levels of parasitemia. Despite a highly disorganized splenic architecture before infection, these mice elicited a surprisingly robust adaptive immune response (including comparable levels of GC B cells, TFH cells, and effector cytokines as control and sickle trait mice), but higher immunoglobulin G responses against 2 Babesia-specific proteins, which may contain potential immunogenic epitopes. Together, these studies establish the robust emergence of adaptive immunity to Babesia even in immunologically compromised SCD mice. Identification of potentially immunogenic epitopes has implications to identify long-term carriers, and aid Ag-specific vaccine development.


Asunto(s)
Inmunidad Adaptativa , Anemia de Células Falciformes/patología , Babesia microti/inmunología , Babesiosis/patología , Parasitemia/diagnóstico , Anemia de Células Falciformes/parasitología , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Babesia microti/aislamiento & purificación , Babesiosis/inmunología , Babesiosis/parasitología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Epítopos/inmunología , Eritrocitos/citología , Inmunoglobulina G/sangre , Interferón gamma/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
Blood ; 131(14): 1600-1610, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29437594

RESUMEN

Patients with sickle cell disease (SCD) suffer from intravascular hemolysis associated with vascular injury and dysfunction in mouse models, and painful vaso-occlusive crisis (VOC) involving increased attachment of sickle erythrocytes and activated leukocytes to damaged vascular endothelium. Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme. Here, we show that HO-1-expressing patrolling monocytes protect SCD vasculature from ongoing hemolytic insult and vaso-occlusion. We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. SCD patients with a recent VOC episode had lower numbers of HO-1hi patrolling monocytes. Heme-mediated vaso-occlusion by mouse SCD red blood cells was exacerbated in mice lacking patrolling monocytes, and reversed following transfer of patrolling monocytes. Altogether, these data indicate that SCD patrolling monocytes remove hemolysis-damaged endothelial cells, resulting in HO-1 upregulation and dampening of VOC, and that perturbation in patrolling monocyte numbers resulting in lower numbers of HO-1hi patrolling monocyte may predispose SCD patients to VOC. These data suggest that HO-1hi patrolling monocytes are key players in VOC pathophysiology and have potential as therapeutic targets for VOC.


Asunto(s)
Anemia de Células Falciformes/enzimología , Hemo-Oxigenasa 1/metabolismo , Hemólisis , Monocitos/enzimología , Enfermedades Vasculares/prevención & control , Adolescente , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/patología , Niño , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/patología , Enfermedades Vasculares/enzimología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
14.
Science ; 351(6269): 176-80, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26634440

RESUMEN

Whereas the cellular basis of the hematopoietic stem cell (HSC) niche in the bone marrow has been characterized, the nature of the fetal liver niche is not yet elucidated. We show that Nestin(+)NG2(+) pericytes associate with portal vessels, forming a niche promoting HSC expansion. Nestin(+)NG2(+) cells and HSCs scale during development with the fractal branching patterns of portal vessels, tributaries of the umbilical vein. After closure of the umbilical inlet at birth, portal vessels undergo a transition from Neuropilin-1(+)Ephrin-B2(+) artery to EphB4(+) vein phenotype, associated with a loss of periportal Nestin(+)NG2(+) cells and emigration of HSCs away from portal vessels. These data support a model in which HSCs are titrated against a periportal vascular niche with a fractal-like organization enabled by placental circulation.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Hígado/embriología , Sistema Porta/embriología , Nicho de Células Madre/fisiología , Animales , Antígenos/análisis , Efrina-B2/análisis , Femenino , Hígado/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Nestina/análisis , Neuropilina-1/análisis , Circulación Placentaria , Sistema Porta/química , Embarazo , Proteoglicanos/análisis , Receptor EphB4/análisis
15.
Nat Med ; 20(8): 833-46, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25100529

RESUMEN

The bone marrow niche has mystified scientists for many years, leading to widespread investigation to shed light into its molecular and cellular composition. Considerable efforts have been devoted toward uncovering the regulatory mechanisms of hematopoietic stem cell (HSC) niche maintenance. Recent advances in imaging and genetic manipulation of mouse models have allowed the identification of distinct vascular niches that have been shown to orchestrate the balance between quiescence, proliferation and regeneration of the bone marrow after injury. Here we highlight the recently discovered intrinsic mechanisms, microenvironmental interactions and communication with surrounding cells involved in HSC regulation, during homeostasis and in regeneration after injury and discuss their implications for regenerative therapy.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Regeneración/fisiología , Nicho de Células Madre/fisiología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Endoteliales/citología , Hematopoyesis , Homeostasis , Humanos , Ratones , Modelos Animales
16.
Dev Cell ; 29(3): 340-9, 2014 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-24823377

RESUMEN

Mesenchymal stem and progenitor cells (MSPCs) contribute to bone marrow (BM) homeostasis by generating multiple types of stromal cells. MSPCs can be labeled in the adult BM by Nestin-GFP, whereas committed osteoblast progenitors are marked by Osterix expression. However, the developmental origin and hierarchical relationship of stromal cells remain largely unknown. Here, by using a lineage-tracing system, we describe three distinct waves of contributions of Osterix(+) cells in the BM. First, Osterix(+) progenitors in the fetal BM contribute to nascent bone tissues and transient stromal cells that are replaced in the adult marrow. Second, Osterix-expressing cells perinatally contribute to osteolineages and long-lived BM stroma, which have characteristics of Nestin-GFP(+) MSPCs. Third, Osterix labeling in the adult marrow is osteolineage-restricted, devoid of stromal contribution. These results uncover a broad expression profile of Osterix and raise the intriguing possibility that distinct waves of stromal cells, primitive and definitive, may organize the developing BM.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/embriología , Huesos/embriología , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción/metabolismo , Adipocitos/citología , Animales , Regeneración Ósea/fisiología , Regeneración Ósea/efectos de la radiación , Linaje de la Célula , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nestina/metabolismo , Osteoblastos/metabolismo , Factor de Transcripción Sp7
17.
Plast Reconstr Surg ; 133(6): 1344-1353, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24867716

RESUMEN

BACKGROUND: Current augmentative and reconstructive rhinoplasties use auto logous tissue grafts or synthetic bioinert materials to repair nasal trauma or attain an aesthetic shape. Autologous grafts are associated with donor-site trauma and morbidity. Synthetic materials are widely used but often yield an unnatural appearance and are prone to infection or dislocation. There is an acute clinical need for the generation of native tissues to serve as rhinoplasty grafts without the undesirable features that are associated with autologous grafts or current synthetic materials. METHODS: Bioactive scaffolds were developed that not only recruited cells in the nasal dorsum in vivo, but also induced chondrogenesis of the recruited cells. Bilayered scaffolds were fabricated with alginate-containing gelatin microspheres encapsulating cytokines atop a porous poly(lactic-co-glycolic acid) base. Microspheres were fabricated to contain recombinant human transforming growth factor-ß3 at doses of 200, 500, or 1000 ng, with phosphate-buffered saline-loaded microspheres used as a control. A rat model of augmentation rhinoplasty was created by implanting scaffolds atop the native nasal cartilage surface that was scored to induce cell migration. Tissue formation and chondrogenesis in the scaffolds were evaluated by image analysis and histologic staining with hematoxylin and eosin, toluidine blue, Verhoeff elastic-van Geison, and aggrecan immunohistochemistry. RESULTS: Sustained release of increasing doses of transforming growth factor-ß3 for up to the tested 10 weeks promoted orthotopic cartilage-like tissue formation in a dose-dependent manner. CONCLUSIONS: These findings represent the first attempt to engineer cartilage tissue by cell homing for rhinoplasty, and could potentially serve as an alternative material for augmentative and reconstructive rhinoplasty.


Asunto(s)
Rinoplastia/métodos , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Materiales Biocompatibles/uso terapéutico , Movimiento Celular , Condrogénesis/fisiología , Materiales Biocompatibles Revestidos/química , Ácido Láctico/uso terapéutico , Células Madre Mesenquimatosas/citología , Microesferas , Modelos Animales , Cartílagos Nasales/citología , Ácido Poliglicólico/uso terapéutico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Sprague-Dawley , Procedimientos de Cirugía Plástica , Factor de Crecimiento Transformador beta3/administración & dosificación
18.
Lab Chip ; 13(6): 1156-64, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23364311

RESUMEN

A multitude of cytotactic cues direct cell migration in development, cancer metastasis and wound healing. However, our understanding of cell motility remains fragmented partially because current migration devices only allow the study of independent factors. We developed a cell motility assay that allows competitive recruitment of a given cell population simultaneously by gradients of multiple cytotactic cues, observable under real-time imaging. Well-defined uniform gradients of cytotactic cues can be independently generated and sustained in each channel. As a case study, bone marrow mesenchymal stem/stromal cells (MSCs) were exposed to 15 cytokines that are commonly present in arthritis. Cytokines that induced robust recruitment of MSCs in multiple groups were selected to 'compete' in a final round to yield the most chemotactic factor(s) based on cell migration numbers, distances, migration indices and motility over time. The potency of a given cytokine in competition frequently differed from its individual action, substantiating the need to test multiple cytokines concurrently due to synergistic or antagonistic effects. This new device has the rare capacity to screen molecules that induce cell migration in cancer therapy, drug development and tissue regeneration.


Asunto(s)
Citocinas/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células de la Médula Ósea/citología , Movimiento Celular/efectos de los fármacos , Factores Quimiotácticos , Colorantes Fluorescentes/química , Humanos , Células Madre Mesenquimatosas/citología , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Sefarosa/química
19.
Cell Tissue Res ; 347(3): 665-76, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22382390

RESUMEN

From its inception, tissue engineering has had three tenets: cells, biomaterial scaffolds and signaling molecules. Among the triad, cells are the center piece, because cells are the building blocks of tissues. For decades, cell therapies have focused on the procurement, manipulation and delivery of healthy cells for the treatment of diseases or trauma. Given the complexity and potential high cost of cell delivery, there is recent and surging interest to orchestrate endogenous cells for tissue regeneration. Biomaterial scaffolds are vital for many but not all, tissue-engineering applications and serve to accommodate or promote multiple cellular functions. Signaling molecules can be produced by transplanted cells or endogenous cells, or delivered specifically to regulate cell functions. This review highlights recent work in tissue engineering and cell therapies, with a focus on harnessing the capacity of endogenous cells as an alternative or adjunctive approach for tissue regeneration.


Asunto(s)
Fenómenos Fisiológicos Musculoesqueléticos , Células Madre/citología , Ingeniería de Tejidos/métodos , Animales , Humanos , Trasplante de Células Madre
20.
FASEB J ; 25(10): 3496-504, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21746864

RESUMEN

Cell transplantation has been well explored for cartilage regeneration. We recently showed that the entire articular surface of a synovial joint can regenerate by endogenous cell homing and without cell transplantation. However, the sources of endogenous cells that regenerate articular cartilage remain elusive. Here, we studied whether cytokines not only chemotactically recruit adipose stem cells (ASCs), mesenchymal stem cells (MSCs), and synovium stem cells (SSCs) but also induce chondrogenesis of the recruited cells. Recombinant human transforming growth factor-ß3 (TGF-ß3; 100 ng) and/or recombinant human stromal derived factor-1ß (SDF-1ß; 100 ng) was control released into an acellular collagen sponge cube with underlying ASCs, MSCs, or SSCs in monolayer culture. Although all cell types randomly migrated into the acellular collagen sponge cube, TGF-ß3 and/or SDF-1ß recruited significantly more cells than the cytokine-free control group. In 6 wk, TGF-ß3 alone recruited substantial numbers of ASCs (558±65) and MSCs (302±52), whereas codelivery of TGF-ß3 and SDF-1ß was particularly chemotactic to SSCs (400±120). Proliferation of the recruited cells accounted for some, but far from all, of the observed cellularity. TGF-ß3 and SDF-1ß codelivery induced significantly higher aggrecan gene expression than the cytokine-free group for ASCs, MSCs, and SSCs. Type II collagen gene expression was also significantly higher for ASCs and SSCs by SDF-1 and TGF-ß3 codelivery. Remarkably, the expression of aggrecan and type II collagen was detected among all cell types. Thus, homing of multiple stem/progenitor cell populations may potentially serve as an alternative or adjunctive approach to cell transplantation for cartilage regeneration.


Asunto(s)
Adipocitos/fisiología , Células de la Médula Ósea/fisiología , Quimiotaxis/fisiología , Condrogénesis/fisiología , Células Madre/fisiología , Membrana Sinovial/citología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Citocinas , Humanos , Células Madre/efectos de los fármacos , Membrana Sinovial/fisiología , Ingeniería de Tejidos , Andamios del Tejido , Factor de Crecimiento Transformador beta3/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...