Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Stem Cells Dev ; 27(10): 671-682, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29631496

RESUMEN

Emerging evidence demonstrates that megakaryocytes (MK) play key roles in regulating skeletal homeostasis and hematopoiesis. To test if the loss of MK negatively impacts osteoblastogenesis and hematopoiesis, we generated conditional knockout mice where Mpl, the receptor for the main MK growth factor, thrombopoietin, was deleted specifically in MK (Mplf/f;PF4cre). Unexpectedly, at 12 weeks of age, these mice exhibited a 10-fold increase in platelets, a significant expansion of hematopoietic/mesenchymal precursors, and a remarkable 20-fold increase in femoral midshaft bone volume. We then investigated whether MK support hematopoietic stem cell (HSC) function through the interaction of MK with osteoblasts (OB). LSK cells (Lin-Sca1+CD117+, enriched HSC population) were co-cultured with OB+MK for 1 week (1wk OB+MK+LSK) or OB alone (1wk OB+LSK). A significant increase in colony-forming units was observed with cells from 1wk OB+MK cultures. Competitive repopulation studies demonstrated significantly higher engraftment in mice transplanted with cells from 1wk OB+MK+LSK cultures compared to 1wk OB+LSK or LSK cultured alone for 1 week. Furthermore, single-cell expression analysis of OB cultured±MK revealed adiponectin as the most significantly upregulated MK-induced gene, which is required for optimal long-term hematopoietic reconstitution. Understanding the interactions between MK, OB, and HSC can inform the development of novel treatments to enhance both HSC recovery following myelosuppressive injuries, as well as bone loss diseases, such as osteoporosis.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Megacariocitos/citología , Osteoblastos/citología , Animales , Células Cultivadas , Técnicas de Cocultivo/métodos , Femenino , Células Madre Hematopoyéticas/metabolismo , Masculino , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/metabolismo , Trombopoyetina/metabolismo
2.
J Cell Biochem ; 118(8): 2231-2240, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28067429

RESUMEN

The Lnk adapter protein negatively regulates the signaling of thrombopoietin (TPO), the main megakaryocyte (MK) growth factor. Lnk-deficient (-/-) mice have increased TPO signaling and increased MK number. Interestingly, several mouse models exist in which increased MK number leads to a high bone mass phenotype. Here we report the bone phenotype of these mice. MicroCT and static histomorphometric analyses at 20 weeks showed the distal femur of Lnk-/- mice to have significantly higher bone volume fraction and trabecular number compared to wild-type (WT) mice. Notably, despite a significant increase in the number of osteoclasts (OC), and decreased bone formation rate in Lnk-/- mice compared to WT mice, Lnk-/- mice demonstrated a 2.5-fold greater BV/TV suggesting impaired OC function in vivo. Additionally, Lnk-/- mouse femurs exhibited non-significant increases in mid-shaft cross-sectional area, yet increased periosteal BFR compared to WT femurs was observed. Lnk-/- femurs also had non-significant increases in polar moment of inertia and decreased cortical bone area and thickness, resulting in reduced bone stiffness, modulus, and strength compared to WT femurs. Of note, Lnk is expressed by OC lineage cells and when Lnk-/- OC progenitors are cultured in the presence of TPO, significantly more OC are observed than in WT cultures. Lnk is also expressed in osteoblast (OB) cells and in vitro reduced alkaline phosphatase activity was observed in Lnk-/- cultures. These data suggest that both direct effects on OB and OC as well as indirect effects of MK in regulating OB contributes to the observed high bone mass. J. Cell. Biochem. 118: 2231-2240, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Trombopoyetina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Células de la Médula Ósea/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Femenino , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Megacariocitos/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Células RAW 264.7 , Trombopoyetina/genética , Microtomografía por Rayos X
3.
J Cell Biochem ; 117(4): 959-69, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26375403

RESUMEN

C-Mpl is the receptor for thrombopoietin (TPO), the main megakaryocyte (MK) growth factor, and c-Mpl is believed to be expressed on cells of the hematopoietic lineage. As MKs have been shown to enhance bone formation, it may be expected that mice in which c-Mpl was globally knocked out (c-Mpl(-/-) mice) would have decreased bone mass because they have fewer MKs. Instead, c-Mpl(-/-) mice have a higher bone mass than WT controls. Using c-Mpl(-/-) mice we investigated the basis for this discrepancy and discovered that c-Mpl is expressed on both osteoblasts (OBs) and osteoclasts (OCs), an unexpected finding that prompted us to examine further how c-Mpl regulates bone. Static and dynamic bone histomorphometry parameters suggest that c-Mpl deficiency results in a net gain in bone volume with increases in OBs and OCs. In vitro, a higher percentage of c-Mpl(-/-) OBs were in active phases of the cell cycle, leading to an increased number of OBs. No difference in OB differentiation was observed in vitro as examined by real-time PCR and functional assays. In co-culture systems, which allow for the interaction between OBs and OC progenitors, c-Mpl(-/-) OBs enhanced osteoclastogenesis. Two of the major signaling pathways by which OBs regulate osteoclastogenesis, MCSF/OPG/RANKL and EphrinB2-EphB2/B4, were unaffected in c-Mpl(-/-) OBs. These data provide new findings for the role of MKs and c-Mpl expression in bone and may provide insight into the homeostatic regulation of bone mass as well as bone loss diseases such as osteoporosis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Receptores de Trombopoyetina/genética , Trombopoyetina/genética , Animales , Animales Recién Nacidos , Densidad Ósea , Recuento de Células , Diferenciación Celular , División Celular , Efrina-B2/genética , Efrina-B2/metabolismo , Homeostasis/genética , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Receptor EphB2/genética , Receptor EphB2/metabolismo , Receptor EphB4/genética , Receptor EphB4/metabolismo , Receptores de Trombopoyetina/deficiencia , Transducción de Señal , Cráneo/citología , Cráneo/metabolismo , Trombopoyetina/metabolismo
4.
Cell ; 161(7): 1553-65, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26073944

RESUMEN

Hematopoietic stem cells (HSCs) reside in hypoxic niches within bone marrow and cord blood. Yet, essentially all HSC studies have been performed with cells isolated and processed in non-physiologic ambient air. By collecting and manipulating bone marrow and cord blood in native conditions of hypoxia, we demonstrate that brief exposure to ambient oxygen decreases recovery of long-term repopulating HSCs and increases progenitor cells, a phenomenon we term extraphysiologic oxygen shock/stress (EPHOSS). Thus, true numbers of HSCs in the bone marrow and cord blood are routinely underestimated. We linked ROS production and induction of the mitochondrial permeability transition pore (MPTP) via cyclophilin D and p53 as mechanisms of EPHOSS. The MPTP inhibitor cyclosporin A protects mouse bone marrow and human cord blood HSCs from EPHOSS during collection in air, resulting in increased recovery of transplantable HSCs. Mitigating EPHOSS during cell collection and processing by pharmacological means may be clinically advantageous for transplantation.


Asunto(s)
Médula Ósea , Sangre Fetal/citología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Peptidil-Prolil Isomerasa F , Ciclofilinas/metabolismo , Femenino , Trasplante de Células Madre Hematopoyéticas/instrumentación , Células Madre Hematopoyéticas/citología , Humanos , Hipoxia , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
Am J Physiol Renal Physiol ; 308(12): F1421-30, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25925259

RESUMEN

Prostatic inflammation is a nearly ubiquitous pathological feature observed in specimens from benign prostate hyperplasia and prostate cancer patients. The microenvironment of the inflamed prostate is highly reactive, and epithelial hyperplasia is a hallmark feature of inflamed prostates. How inflammation orchestrates epithelial proliferation as part of its repair and recovery action is not well understood. Here, we report that a novel epithelial progenitor cell population is induced to expand during inflammation. We used sphere culture assays, immunofluorescence, and flow cytometry to show that this population is increased in bacterially induced inflamed mouse prostates relative to naïve control prostates. We confirmed from previous reports that this population exclusively possesses the ability to regrow entire prostatic structures from single cell culture using renal grafts. In addition, putative progenitor cells harvested from inflamed animals have greater aggregation capacity than those isolated from naïve control prostates. Expansion of this critical cell population requires IL-1 signaling, as IL-1 receptor 1-null mice exhibit inflammation similar to wild-type inflamed animals but exhibit significantly reduced progenitor cell proliferation and hyperplasia. These data demonstrate that inflammation promotes hyperplasia in the mouse prostatic epithelium by inducing the expansion of a selected epithelial progenitor cell population in an IL-1 receptor-dependent manner. These findings may have significant impact on our understanding of how inflammation promotes proliferative diseases such as benign prostatic hyperplasia and prostate cancer, both of which depend on expansion of cells that exhibit a progenitor-like nature.


Asunto(s)
Proliferación Celular/fisiología , Hiperplasia Prostática/patología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Inflamación/patología , Interleucina-1/metabolismo , Masculino , Ratones , Ratones Noqueados
6.
Blood Cells Mol Dis ; 55(1): 15-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25976461

RESUMEN

Ames hypopituitary dwarf mice are deficient in growth hormone, thyroid-stimulating hormone, and prolactin. The phenotype of these mice demonstrates irregularities in the immune system with skewing of the normal cytokine milieu towards a more anti-inflammatory environment. However, the hematopoietic stem and progenitor cell composition of the bone marrow (BM) and spleen in Ames dwarf mice has not been well characterized. We found that there was a significant decrease in overall cell count when comparing the BM and spleen of 4-5 month old dwarf mice to their littermate controls. Upon adjusting counts to differences in body weight between the dwarf and control mice, the number of granulocyte-macrophage progenitors, confirmed by immunophenotyping and colony-formation assay was increased in the BM. In contrast, the numbers of all myeloid progenitor populations in the spleen were greatly reduced, as confirmed by colony-formation assays. This suggests that there is a shift of myelopoiesis from the spleen to the BM of Ames dwarf mice; however, this shift does not appear to involve erythropoiesis. The reasons for this unusual shift in spleen to marrow hematopoiesis in Ames dwarf mice are yet to be determined but may relate to the decreased hormone levels in these mice.


Asunto(s)
Médula Ósea/patología , Enanismo/patología , Hipopituitarismo/patología , Células Mieloides/patología , Mielopoyesis/inmunología , Bazo/patología , Animales , Médula Ósea/inmunología , Recuento de Células , Cruzamientos Genéticos , Enanismo/genética , Enanismo/inmunología , Femenino , Fémur/inmunología , Fémur/patología , Expresión Génica , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/genética , Hormona del Crecimiento/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Hipopituitarismo/genética , Hipopituitarismo/inmunología , Inmunofenotipificación , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Mieloides/inmunología , Mielopoyesis/genética , Prolactina/deficiencia , Prolactina/genética , Prolactina/inmunología , Bazo/inmunología , Tirotropina/deficiencia , Tirotropina/genética , Tirotropina/inmunología
7.
J Cell Physiol ; 230(9): 2142-51, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25656774

RESUMEN

Emerging data suggest that megakaryocytes (MKs) play a significant role in skeletal homeostasis. Indeed, osteosclerosis observed in several MK-related disorders may be a result of increased numbers of MKs. In support of this idea, we have previously demonstrated that MKs increase osteoblast (OB) proliferation by a direct cell-cell contact mechanism and that MKs also inhibit osteoclast (OC) formation. As MKs and OCs are derived from the same hematopoietic precursor, in these osteoclastogenesis studies we examined the role of the main MK growth factor, thrombopoietin (TPO) on OC formation and bone resorption. Here we show that TPO directly increases OC formation and differentiation in vitro. Specifically, we demonstrate the TPO receptor (c-mpl or CD110) is expressed on cells of the OC lineage, c-mpl is required for TPO to enhance OC formation in vitro, and TPO activates the mitogen-activated protein kinases, Janus kinase/signal transducer and activator of transcription, and nuclear factor-kappaB signaling pathways, but does not activate the PI3K/AKT pathway. Further, we found TPO enhances OC resorption in CD14+CD110+ human OC progenitors derived from peripheral blood mononuclear cells, and further separating OC progenitors based on CD110 expression enriches for mature OC development. The regulation of OCs by TPO highlights a novel therapeutic target for bone loss diseases and may be important to consider in the numerous hematologic disorders associated with alterations in TPO/c-mpl signaling as well as in patients suffering from bone disorders.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Osteoclastos/metabolismo , Proteínas Recombinantes/administración & dosificación , Trombopoyetina/administración & dosificación , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Hematopoyesis/genética , Humanos , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Ratones Noqueados , Osteoclastos/patología , Receptores de Trombopoyetina/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Células Madre/efectos de los fármacos , Trombopoyetina/metabolismo
8.
J Am Soc Nephrol ; 26(9): 2172-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25556169

RESUMEN

Tamm-Horsfall protein (THP) is a glycoprotein uniquely expressed in the kidney. We recently showed an important role for THP in mediating tubular cross-talk in the outer medulla and in suppressing neutrophil infiltration after kidney injury. However, it remains unclear whether THP has a broader role in neutrophil homeostasis. In this study, we show that THP deficiency in mice increases the number of neutrophils, not only in the kidney but also in the circulation and in the liver, through enhanced granulopoiesis in the bone marrow. Using multiplex ELISA, we identified IL-17 as a key granulopoietic cytokine specifically upregulated in the kidneys but not in the liver of THP(-/-) mice. Indeed, neutralization of IL-17 in THP(-/-) mice completely reversed the systemic neutrophilia. Furthermore, IL-23 was also elevated in THP(-/-) kidneys. We performed real-time PCR on laser microdissected tubular segments and FACS-sorted renal immune cells and identified the S3 proximal segments, but not renal macrophages, as a major source of increased IL-23 synthesis. In conclusion, we show that THP deficiency stimulates proximal epithelial activation of the IL-23/IL-17 axis and systemic neutrophilia. Our findings provide evidence that the kidney epithelium in the outer medulla can regulate granulopoiesis. When this novel function is added to its known role in erythropoiesis, the kidney emerges as an important regulator of the hematopoietic system.


Asunto(s)
Granulocitos , Hematopoyesis , Homeostasis , Neutrófilos , Uromodulina/deficiencia , Animales , Médula Ósea/fisiología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Túbulos Renales Proximales/metabolismo , Ratones , Ratones Noqueados , Uromodulina/genética
9.
J Cell Physiol ; 230(3): 578-86, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25160801

RESUMEN

Recent studies suggest that megakaryocytes (MKs) may play a significant role in skeletal homeostasis, as evident by the occurrence of osteosclerosis in multiple MK related diseases (Lennert et al., 1975; Thiele et al., 1999; Chagraoui et al., 2006). We previously reported a novel interaction whereby MKs enhanced proliferation of osteoblast lineage/osteoprogenitor cells (OBs) by a mechanism requiring direct cell-cell contact. However, the signal transduction pathways and the downstream effector molecules involved in this process have not been characterized. Here we show that MKs contact with OBs, via beta1 integrin, activate the p38/MAPKAPK2/p90RSK kinase cascade in the bone cells, which causes Mdm2 to neutralizes p53/Rb-mediated check point and allows progression through the G1/S. Interestingly, activation of MAPK (ERK1/2) and AKT, collateral pathways that regulate the cell cycle, remained unchanged with MK stimulation of OBs. The MK-to-OB signaling ultimately results in significant increases in the expression of c-fos and cyclin A, necessary for sustaining the OB proliferation. Overall, our findings show that OBs respond to the presence of MKs, in part, via an integrin-mediated signaling mechanism, activating a novel response axis that de-represses cell cycle activity. Understanding the mechanisms by which MKs enhance OB proliferation will facilitate the development of novel anabolic therapies to treat bone loss associated with osteoporosis and other bone-related diseases.


Asunto(s)
Diferenciación Celular/genética , Megacariocitos/citología , Osteoblastos/citología , Transducción de Señal/genética , Ciclo Celular/genética , Linaje de la Célula , Proliferación Celular/genética , Células Cultivadas , Humanos , Sistema de Señalización de MAP Quinasas/genética , Megacariocitos/metabolismo , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
10.
Stem Cells ; 33(2): 468-78, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25329668

RESUMEN

OBJECTIVE: Bone marrow-derived hematopoietic stem and progenitor cells (HSC/HPC) are critical to homeostasis and tissue repair. The aims of this study were to delineate the myelotoxicity of cigarette smoking (CS) in a murine model, to explore human adipose-derived stem cells (hASC) as a novel approach to mitigate this toxicity, and to identify key mediating factors for ASC activities. METHODS: C57BL/6 mice were exposed to CS with or without i.v. injection of regular or siRNA-transfected hASC. For in vitro experiments, cigarette smoke extract was used to mimic the toxicity of CS exposure. Analysis of bone marrow HPC was performed both by flow cytometry and colony-forming unit assays. RESULTS: In this study, we demonstrate that as few as 3 days of CS exposure results in marked cycling arrest and diminished clonogenic capacity of HPC, followed by depletion of phenotypically defined HSC/HPC. Intravenous injection of hASC substantially ameliorated both acute and chronic CS-induced myelosuppression. This effect was specifically dependent on the anti-inflammatory factor TSG-6, which is induced from xenografted hASC, primarily located in the lung and capable of responding to host inflammatory signals. Gene expression analysis within bone marrow HSC/HPC revealed several specific signaling molecules altered by CS and normalized by hASC. CONCLUSION: Our results suggest that systemic administration of hASC or TSG-6 may be novel approaches to reverse CS-induced myelosuppression.


Asunto(s)
Tejido Adiposo/metabolismo , Moléculas de Adhesión Celular/metabolismo , Mielopoyesis , Fumar/efectos adversos , Trasplante de Células Madre , Células Madre/metabolismo , Tejido Adiposo/patología , Animales , Moléculas de Adhesión Celular/farmacología , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fumar/patología , Células Madre/patología
11.
Cell Stem Cell ; 15(1): 51-65, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24996169

RESUMEN

The microRNA miR-155 has been implicated in regulating inflammatory responses and tumorigenesis, but its precise role in linking inflammation and cancer has remained elusive. Here, we identify a connection between miR-155 and Notch signaling in this context. Loss of Notch signaling in the bone marrow (BM) niche alters hematopoietic homeostasis and leads to lethal myeloproliferative-like disease. Mechanistically, Notch signaling represses miR-155 expression by promoting binding of RBPJ to the miR-155 promoter. Loss of Notch/RBPJ signaling upregulates miR-155 in BM endothelial cells, leading to miR-155-mediated targeting of the nuclear factor κB (NF-κB) inhibitor κB-Ras1, NF-κB activation, and increased proinflammatory cytokine production. Deletion of miR-155 in the stroma of RBPJ(-/-) mice prevented the development of myeloproliferative-like disease and cytokine induction. Analysis of BM from patients carrying myeloproliferative neoplasia also revealed elevated expression of miR-155. Thus, the Notch/miR-155/κB-Ras1/NF-κB axis regulates the inflammatory state of the BM niche and affects the development of myeloproliferative disorders.


Asunto(s)
Médula Ósea/fisiología , Neoplasias Hematológicas/genética , MicroARNs/metabolismo , Trastornos Mieloproliferativos/genética , Receptores Notch/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Represión Epigenética , Regulación Neoplásica de la Expresión Génica , Hematopoyesis/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , MicroARNs/genética , FN-kappa B/metabolismo , Transducción de Señal/genética , Nicho de Células Madre , Regulación hacia Arriba
12.
Stem Cells Dev ; 23(21): 2661-71, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24914588

RESUMEN

A hyaluronic-acid-rich node and duct system (HAR-NDS) was found on the surface of internal organs of mice, and inside their blood and lymph vessels. The nodes (HAR-Ns) were filled with immune cells of the innate system and were especially enriched with mast cells and histiocytes. They also contained hematopoietic progenitor cells (HPCs), such as granulocyte-macrophage, erythroid, multipotential progenitors, and mast cell progenitors (MCPs). MCPs were the most abundant among the HPCs in HAR-Ns. Their frequency was fivefold higher than that of the MCPs in bone marrow. In addition, the system contained pluripotent stem cells (PSCs) capable of producing CD45(-)Flk1(+) hemangioblast-like cells, which subsequently generated various types of HPCs and differentiated blood cells. Although HAR-Ns did not appear to harbor enough number of cells capable of long-term reconstitution or short-term radioprotection of lethally irradiated recipients, bone marrow cells were able to engraft in the HAR-NDS and reconstitute hematopoietic potentials of the system. PSCs and HPCs were consistently found in intravenous, intralymphatic, and intestinal HAR-ND. We infer that PSCs and HPCs reside in the HAR-ND and that this novel system may serve as an alternative means to traffic immature and mature blood cells throughout the body.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ácido Hialurónico/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Citometría de Flujo , Hemangioblastos/citología , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/ultraestructura , Histiocitos/citología , Histiocitos/metabolismo , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Mastocitos/citología , Mastocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/ultraestructura , Bazo/citología , Bazo/metabolismo , Trasplante de Células Madre/métodos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Curr Opin Hematol ; 20(4): 273-80, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23615053

RESUMEN

PURPOSE OF REVIEW: Many surface antigens have been previously used to identify hematopoietic stem cells or cellular elements of the hematopoietic niche. However, to date, not a single surface marker has been identified as a common marker expressed on murine and human hematopoietic stem cells and on cells of the hematopoietic niche. Recently, a few laboratories, including ours, recognized the importance of CD166 as a functional marker on both stem cells and osteoblasts and have begun to characterize the role of CD166 in hematopoiesis. RECENT FINDINGS: Expression of CD166 on hematopoietic cells and cells in the marrow microenvironment was first reported more than a decade ago. Lately, however, a more prominent role for CD166 in normal hematopoiesis and in cancer biology including metastasis began to emerge. This review will cover the significance of CD166 in identifying normal hematopoietic stem cells and cells of the hematopoietic niche and highlight how CD166-mediated homophilic interactions between both cell types may be critical for stem cell function. SUMMARY: The conserved homology between murine and human CD166 and its involvement in metastasis provides an excellent bridge for translational investigations aimed at enhancing stem cell engraftment and clinical utility of stem cells and at using CD166 as a therapeutic target in cancer.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Células Madre Hematopoyéticas/metabolismo , Animales , Biomarcadores/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Neoplasias/metabolismo
14.
Nature ; 495(7441): 365-9, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23485965

RESUMEN

To maintain lifelong production of blood cells, haematopoietic stem cells (HSCs) are tightly regulated by inherent programs and extrinsic regulatory signals received from their microenvironmental niche. Long-term repopulating HSCs reside in several, perhaps overlapping, niches that produce regulatory molecules and signals necessary for homeostasis and for increased output after stress or injury. Despite considerable advances in the specific cellular or molecular mechanisms governing HSC-niche interactions, little is known about the regulatory function in the intact mammalian haematopoietic niche. Recently, we and others described a positive regulatory role for prostaglandin E2 (PGE2) on HSC function ex vivo. Here we show that inhibition of endogenous PGE2 by non-steroidal anti-inflammatory drug (NSAID) treatment in mice results in modest HSC egress from the bone marrow. Surprisingly, this was independent of the SDF-1-CXCR4 axis implicated in stem-cell migration. Stem and progenitor cells were found to have differing mechanisms of egress, with HSC transit to the periphery dependent on niche attenuation and reduction in the retentive molecule osteopontin. Haematopoietic grafts mobilized with NSAIDs had superior repopulating ability and long-term engraftment. Treatment of non-human primates and healthy human volunteers confirmed NSAID-mediated egress in other species. PGE2 receptor knockout mice demonstrated that progenitor expansion and stem/progenitor egress resulted from reduced E-prostanoid 4 (EP4) receptor signalling. These results not only uncover unique regulatory roles for EP4 signalling in HSC retention in the niche, but also define a rapidly translatable strategy to enhance transplantation therapeutically.


Asunto(s)
Dinoprostona/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre/citología , Animales , Antiinflamatorios no Esteroideos/farmacología , Bencilaminas , Recuento de Células , Movimiento Celular/fisiología , Células Cultivadas , Ciclamas , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Meloxicam , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteopontina/genética , Papio , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Madre/efectos de los fármacos , Tiazinas/farmacología , Tiazoles/farmacología
15.
Bone ; 54(1): 58-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23369988

RESUMEN

The role of osteoblasts (OB) in maintaining hematopoietic stem cells (HSC) in their niche is well elucidated, but the exact definition, both phenotypically and hierarchically of OB responsible for these functions is not clearly known. We previously demonstrated that OB maturational status influences HSC function whereby immature OB with high Runx2 expression promote hematopoietic expansion. Here, we show that Activated Leukocyte Cell Adhesion Molecule (ALCAM) or CD166 expression on OB is directly correlated with Runx2 expression and high hematopoiesis enhancing activity (HEA). Fractionation of OB with lineage markers: Sca1, osteopontin (OPN), CD166, CD44, and CD90 revealed that Lin-Sca1-OPN+CD166+ cells (CD166+) and their subpopulations fractionated with CD44 and CD90 expressed high levels of Runx2 and low levels of osteocalcin (OC) demonstrating the relatively immature status of these cells. Conversely, the majority of the Lin-Sca1-OPN+CD166- cells (CD166-) expressed high OC levels suggesting that CD166- OB are more mature. In vitro hematopoietic potential of LSK cells co-cultured for 7days with fresh OB or OB pre-cultured for 1, 2, or 3 weeks declined precipitously with increasing culture duration concomitant with loss of CD166 expression. Importantly, LSK cells co-cultured with CD166+CD44+CD90+ OB maintained their in vivo repopulating potential through primary and secondary transplantation, suggesting that robust HEA activity is best mediated by immature CD166+ OB with high Runx2 and low OC expression. These studies begin to define the hierarchical organization of osteoblastic cells and provide a more refined definition of OB that can mediate HEA.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Separación Celular , Técnicas de Cocultivo , Citometría de Flujo , Hematopoyesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo
16.
Curr Osteoporos Rep ; 11(2): 99-106, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23423562

RESUMEN

Osteoblasts (OBs) and adipocytes (APs) share a common mesenchymal ancestor. It is now clear that mesenchymal stem cell (MSC) maturation along the OB lineage comes at the expense of adipogenesis and vice versa. During aging, this balance increasingly favors the formation of APs. Hematopoiesis also slowly declines during the aging process. The role of OB lineage cells in hematopoiesis has been studied, but less is known about how APs regulate hematopoiesis. A few studies have demonstrated a negative relationship between APs and hematopoiesis; however, there is also evidence that brown adipose tissue (BAT) may promote hematopoiesis. This review will examine the current knowledge of how adipogenesis and osteogenesis change with aging and the implications of this changing environment on hematopoeisis.


Asunto(s)
Adipocitos/citología , Adipogénesis/fisiología , Envejecimiento/fisiología , Hematopoyesis/fisiología , Células Madre Mesenquimatosas/fisiología , Osteoblastos/citología , Osteogénesis/fisiología , Diferenciación Celular , Linaje de la Célula , Humanos
17.
J Bone Miner Res ; 28(6): 1434-45, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23362087

RESUMEN

Preclinical and clinical evidence from megakaryocyte (MK)-related diseases suggests that MKs play a significant role in maintaining bone homeostasis. Findings from our laboratories reveal that MKs significantly increase osteoblast (OB) number through direct MK-OB contact and the activation of integrins. We, therefore, examined the role of Pyk2, a tyrosine kinase known to be regulated downstream of integrins, in the MK-mediated enhancement of OBs. When OBs were co-cultured with MKs, total Pyk2 levels in OBs were significantly enhanced primarily because of increased Pyk2 gene transcription. Additionally, p53 and Mdm2 were both decreased in OBs upon MK stimulation, which would be permissive of cell cycle entry. We then demonstrated that OB number was markedly reduced when Pyk2-/- OBs, as opposed to wild-type (WT) OBs, were co-cultured with MKs. We also determined that MKs inhibit OB differentiation in the presence and absence of Pyk2 expression. Finally, given that MK-replete spleen cells from GATA-1-deficient mice can robustly stimulate OB proliferation and bone formation in WT mice, we adoptively transferred spleen cells from these mice into Pyk2-/- recipient mice. Importantly, GATA-1-deficient spleen cells failed to stimulate an increase in bone formation in Pyk2-/- mice, suggesting in vivo the important role of Pyk2 in the MK-induced increase in bone volume. Further understanding of the signaling pathways involved in the MK-mediated enhancement of OB number and bone formation will facilitate the development of novel anabolic therapies to treat bone loss diseases.


Asunto(s)
Diferenciación Celular/fisiología , Quinasa 2 de Adhesión Focal/metabolismo , Megacariocitos/enzimología , Osteoblastos/enzimología , Osteogénesis/fisiología , Animales , Células Cultivadas , Técnicas de Cocultivo , Quinasa 2 de Adhesión Focal/genética , Megacariocitos/citología , Ratones , Ratones Noqueados , Osteoblastos/citología , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Nat Med ; 18(12): 1786-96, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23160239

RESUMEN

Enhancement of hematopoietic recovery after radiation, chemotherapy, or hematopoietic stem cell (HSC) transplantation is clinically relevant. Dipeptidylpeptidase (DPP4) cleaves a wide variety of substrates, including the chemokine stromal cell-derived factor-1 (SDF-1). In the course of experiments showing that inhibition of DPP4 enhances SDF-1-mediated progenitor cell survival, ex vivo cytokine expansion and replating frequency, we unexpectedly found that DPP4 has a more general role in regulating colony-stimulating factor (CSF) activity. DPP4 cleaved within the N-termini of the CSFs granulocyte-macrophage (GM)-CSF, G-CSF, interleukin-3 (IL-3) and erythropoietin and decreased their activity. Dpp4 knockout or DPP4 inhibition enhanced CSF activities both in vitro and in vivo. The reduced activity of DPP4-truncated versus full-length human GM-CSF was mechanistically linked to effects on receptor-binding affinity, induction of GM-CSF receptor oligomerization and signaling capacity. Hematopoiesis in mice after radiation or chemotherapy was enhanced in Dpp4(-/-) mice or mice receiving an orally active DPP4 inhibitor. DPP4 inhibition enhanced engraftment in mice without compromising HSC function, suggesting the potential clinical utility of this approach.


Asunto(s)
Quimiocina CXCL12/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hematopoyesis/fisiología , Radioterapia/efectos adversos , Transducción de Señal/fisiología , Animales , Línea Celular , Cartilla de ADN/genética , Dipeptidil Peptidasa 4/genética , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Humanos , Inmunofenotipificación , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética
19.
J Bone Miner Res ; 26(5): 1111-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542011

RESUMEN

Osteoblasts (OBs) exert a prominent regulatory effect on hematopoietic stem cells (HSCs). We evaluated the difference in hematopoietic expansion and function in response to co-culture with OBs at various stages of development. Murine calvarial OBs were seeded directly (fresh) or cultured for 1, 2, or 3 weeks prior to seeding with 1000 Lin-Sca1 + cKit+ (LSK) cells for 1 week. Significant increases in the following hematopoietic parameters were detected when comparing co-cultures of fresh OBs to co-cultures containing OBs cultured for 1, 2, or 3 weeks: total hematopoietic cell number (up to a 3.4-fold increase), total colony forming unit (CFU) number in LSK progeny (up to an 18.1-fold increase), and percentage of Lin-Sca1+ cells (up to a 31.8-fold increase). Importantly, these studies were corroborated by in vivo reconstitution studies in which LSK cells maintained in fresh OB co-cultures supported a significantly higher level of chimerism than cells maintained in co-cultures containing 3-week OBs. Characterization of OBs cultured for 1, 2, or 3 weeks with real-time PCR and functional mineralization assays showed that OB maturation increased with culture duration but was not affected by the presence of LSK cells in culture. Linear regression analyses of multiple parameters measured in these studies show that fresh, most likely more immature OBs better promote hematopoietic expansion and function than cultured, presumably more mature OBs and suggest that the hematopoiesis-enhancing activity is mediated by cells present in fresh OB cultures de novo.


Asunto(s)
Diferenciación Celular , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Apoptosis , Calcio/metabolismo , Recuento de Células , Ciclo Celular , Linaje de la Célula , Proliferación Celular , Separación Celular , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
J Cell Biochem ; 111(2): 284-94, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20506198

RESUMEN

Although osteoblasts (OB) play a key role in the hematopoietic stem cell (HSC) niche, little is known as to which specific OB lineage cells are critical for the enhancement of stem and progenitor cell function. Unlike hematopoietic cells, OB cell surface phenotypic definitions are not well developed. Therefore, to determine which OB lineage cells are most important for hematopoietic progenitor cell (HPC) function, we characterized OB differentiation by gene expression and OB function, and determined whether associations existed between OB and HPC properties. OB were harvested from murine calvariae, used immediately (fresh OB) or cultured for 1, 2, or 3 weeks prior to their co-culture with Lin(-)Sca1(+)c-kit(+) (LSK) cells for 1 week. OB gene expression, alkaline phosphatase activity, calcium deposition, hematopoietic cell number fold increase, CFU fold increase, and fold increase of Lin(-)Sca1(+) cells were determined. As expected, HPC properties were enhanced when LSK cells were cultured with OB compared to being cultured alone. Initial alkaline phosphatase and calcium deposition levels were significantly and inversely associated with an increase in the number of LSK progeny. Final calcium deposition levels and OB culture duration were inversely associated with all HPC parameters, while Runx2 levels were positively associated with all HPC properties. Since calcium deposition is associated with OB maturation and high levels of Runx2 are associated with less mature OB lineage cells, these results suggest that less mature OB better promote HPC proliferation and function than do more mature OB.


Asunto(s)
Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Células Madre Hematopoyéticas/fisiología , Osteoblastos/fisiología , Animales , Linaje de la Célula , Técnicas de Cocultivo , Células Madre Hematopoyéticas/citología , Ratones , Osteoblastos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA