Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
FASEB J ; 35(4): e21402, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33724567

RESUMEN

Leukemias are challenging diseases to treat due, in part, to interactions between leukemia cells and the bone marrow microenvironment (BMME) that contribute significantly to disease progression. Studies have shown that leukemic cells secrete C-chemokine (C-C motif) ligand 3 (CCL3), to disrupt the BMME resulting in loss of hematopoiesis and support of leukemic cell survival and proliferation. In this study, a murine model of blast crisis chronic myelogenous leukemia (bcCML) that expresses the translocation products BCR/ABL and Nup98/HoxA9 was used to determine the role of CCL3 in BMME regulation. Leukemic cells derived from CCL3-/- mice were shown to minimally engraft in a normal BMME, thereby demonstrating that CCL3 signaling was necessary to recapitulate bcCML disease. Further analysis showed disruption in hematopoiesis within the BMME in the bcCML model. To rescue the altered BMME, therapeutic inhibition of CCL3 signaling was investigated using bone-targeted nanoparticles (NP) to deliver Maraviroc, an inhibitor of C-C chemokine receptor type 5 (CCR5), a CCL3 receptor. NP-mediated Maraviroc delivery partially restored the BMME, significantly reduced leukemic burden, and improved survival. Overall, our results demonstrate that inhibiting CCL3 via CCR5 antagonism is a potential therapeutic approach to restore normal hematopoiesis as well as reduce leukemic burden within the BMME.


Asunto(s)
Leucemia/tratamiento farmacológico , Animales , Proteínas Bacterianas , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Proteínas Fluorescentes Verdes , Leucemia/etiología , Leucemia Mieloide Aguda , Proteínas Luminiscentes , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Dosis de Radiación
2.
Blood ; 127(5): 616-25, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26637787

RESUMEN

In vitro evidence suggests that the bone marrow microenvironment (BMME) is altered in myelodysplastic syndromes (MDSs). Here, we study the BMME in MDS in vivo using a transgenic murine model of MDS with hematopoietic expression of the translocation product NUP98-HOXD13 (NHD13). This model exhibits a prolonged period of cytopenias prior to transformation to leukemia and is therefore ideal to interrogate the role of the BMME in MDS. In this model, hematopoietic stem and progenitor cells (HSPCs) were decreased in NHD13 mice by flow cytometric analysis. The reduction in the total phenotypic HSPC pool in NHD13 mice was confirmed functionally with transplantation assays. Marrow microenvironmental cellular components of the NHD13 BMME were found to be abnormal, including increases in endothelial cells and in dysfunctional mesenchymal and osteoblastic populations, whereas megakaryocytes were decreased. Both CC chemokine ligand 3 and vascular endothelial growth factor, previously shown to be increased in human MDS, were increased in NHD13 mice. To assess whether the BMME contributes to disease progression in NHD13 mice, we performed transplantation of NHD13 marrow into NHD13 mice or their wild-type (WT) littermates. WT recipients as compared with NHD13 recipients of NHD13 marrow had a lower rate of the combined outcome of progression to leukemia and death. Moreover, hematopoietic function was superior in a WT BMME as compared with an NHD13 BMME. Our data therefore demonstrate a contributory role of the BMME to disease progression in MDS and support a therapeutic strategy whereby manipulation of the MDS microenvironment may improve hematopoietic function and overall survival.


Asunto(s)
Médula Ósea/patología , Microambiente Celular , Células Madre Hematopoyéticas/patología , Síndromes Mielodisplásicos/patología , Animales , Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Síndromes Mielodisplásicos/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Transgenes
3.
Bioorg Med Chem ; 26(7): 1365-1373, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28826596

RESUMEN

The plant-derived sesquiterpene lactone parthenolide (PTL) was recently found to possess promising anticancer activity but elaboration of this natural product scaffold for optimization of its pharmacological properties has proven challenging via available chemical methods. In this work, P450-catalyzed C-H hydroxylation of positions C9 and C14 in PTL was coupled to carbamoylation chemistry to yield a panel of novel carbamate-based PTL analogs ('parthenologs'). These compounds, along with a series of other C9- and C14-functionalized parthenologs obtained via O-H acylation, alkylation, and metal-catalyzed carbene insertion, were profiled for their cytotoxicity against a diverse panel of human cancer cell lines. These studies led to the discovery of several parthenologs with significantly improved anticancer activity (2-14-fold) compared to the parent molecule. Most interestingly, two PTL analogs with high cytotoxicity (LC50∼1-3µM) against T cell leukemia (Jurkat), mantle cell lymphoma (JeKo-1), and adenocarcinoma (HeLa) cells as well as a carbamate derivative with potent activity (LC50=0.6µM) against neuroblastoma cells (SK-N-MC) were obtained. In addition, these analyses resulted in the identification of parthenologs featuring both a broad spectrum and tumor cell-specific anticancer activity profile, thus providing valuable probes for the future investigation of biomolecular targets that can affect cell viability across multiple as well as specific types of human cancers. Altogether, these results highlight the potential of P450-mediated chemoenzymatic C-H functionalization toward tuning and improving the anticancer activity of the natural product parthenolide.


Asunto(s)
Antineoplásicos/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Sesquiterpenos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
Blood ; 120(2): 303-13, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-22596259

RESUMEN

Hematopoietic stem cell (HSC) regulation is highly dependent on interactions with the marrow microenvironment. Controversy exists on N-cadherin's role in support of HSCs. Specifically, it is unknown whether microenvironmental N-cadherin is required for normal marrow microarchitecture and for hematopoiesis. To determine whether osteoblastic N-cadherin is required for HSC regulation, we used a genetic murine model in which deletion of Cdh2, the gene encoding N-cadherin, has been targeted to cells of the osteoblastic lineage. Targeted deletion of N-cadherin resulted in an age-dependent bone phenotype, ultimately characterized by decreased mineralized bone, but no difference in steady-state HSC numbers or function at any time tested, and normal recovery from myeloablative injury. Intermittent parathyroid hormone (PTH) treatment is well established as anabolic to bone and to increase marrow HSCs through microenvironmental interactions. Lack of osteoblastic N-cadherin did not block the bone anabolic or the HSC effects of PTH treatment. This report demonstrates that osteoblastic N-cadherin is not required for regulation of steady-state hematopoiesis, HSC response to myeloablation, or for rapid expansion of HSCs through intermittent treatment with PTH.


Asunto(s)
Cadherinas/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Envejecimiento/genética , Envejecimiento/patología , Animales , Secuencia de Bases , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Densidad Ósea/fisiología , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/genética , Remodelación Ósea/fisiología , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Huesos/fisiología , Cadherinas/deficiencia , Cadherinas/genética , Microambiente Celular/fisiología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Blood ; 119(2): 540-50, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21957195

RESUMEN

Pancytopenia is a major cause of morbidity in acute myeloid leukemia (AML), yet its cause is unclear. Normal osteoblastic cells have been shown to support hematopoiesis. To define the effects of leukemia on osteoblastic cells, we used an immunocompetent murine model of AML. Leukemic mice had inhibition of osteoblastic cells, with decreased serum levels of the bone formation marker osteocalcin. Osteoprogenitor cells and endosteal-lining osteopontin(+) cells were reduced, and osteocalcin mRNA in CD45(-) marrow cells was diminished. This resulted in severe loss of mineralized bone. Osteoclasts were only transiently increased without significant increases in bone resorption, and their inhibition only partially rescued leukemia-induced bone loss. In vitro data suggested that a leukemia-derived secreted factor inhibited osteoblastic cells. Because the chemokine CCL-3 was recently reported to inhibit osteoblastic function in myeloma, we tested its expression in our model and in AML patients. Consistent with its potential novel role in leukemic-dependent bone loss, CCL-3 mRNA was significantly increased in malignant marrow cells from leukemic mice and from samples from AML patients. Based on these results, we propose that therapeutic mitigation of leukemia-induced uncoupling of osteoblastic and osteoclastic cells may represent a novel approach to promote normal hematopoiesis in patients with myeloid neoplasms.


Asunto(s)
Quimiocina CCL3/metabolismo , Modelos Animales de Enfermedad , Hematopoyesis , Leucemia Mieloide/patología , Osteoblastos/patología , Osteoclastos/patología , Animales , Western Blotting , Conservadores de la Densidad Ósea/farmacología , Médula Ósea/metabolismo , Médula Ósea/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Quimiocina CCL3/genética , Difosfonatos/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Imidazoles/farmacología , Inmunocompetencia , Técnicas para Inmunoenzimas , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/patología , Ácido Zoledrónico
6.
Blood ; 119(11): 2489-99, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22262765

RESUMEN

Microenvironmental expansion of hematopoietic stem cells (HSCs) is induced by treatment with parathyroid hormone (PTH) or activation of the PTH receptor (PTH1R) in osteoblastic cells; however, the osteoblastic subset mediating this action of PTH is unknown. Osteocytes are terminally differentiated osteoblasts embedded in mineralized bone matrix but are connected with the BM. Activation of PTH1R in osteocytes increases osteoblastic number and bone mass. To establish whether osteocyte-mediated PTH1R signaling expands HSCs, we studied mice expressing a constitutively active PTH1R in osteocytes (TG mice). Osteoblasts, osteoclasts, and trabecular bone were increased in TG mice without changes in BM phenotypic HSCs or HSC function. TG mice had progressively increased trabecular bone but decreased HSC function. In severely affected TG mice, phenotypic HSCs were decreased in the BM but increased in the spleen. TG osteocytes had no increase in signals associated with microenvironmental HSC support, and the spindle-shaped osteoblastic cells that increased with PTH treatment were not present in TG bones. These findings demonstrate that activation of PTH1R signaling in osteocytes does not expand BM HSCs, which are instead decreased in TG mice. Therefore, osteocytes do not mediate the HSC expansion induced by PTH1R signaling. Further, osteoblastic expansion is not sufficient to increase HSCs.


Asunto(s)
Remodelación Ósea , Células Madre Hematopoyéticas/citología , Osteoblastos/citología , Osteocitos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/fisiología , Animales , Citometría de Flujo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Transgénicos , Mutación/genética , Osteoblastos/metabolismo , Osteocitos/citología , Hormona Paratiroidea/metabolismo , Ratas , Transducción de Señal
7.
Stem Cells ; 31(2): 372-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169593

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs), which continuously maintain all mature blood cells, are regulated within the marrow microenvironment. We previously reported that pharmacologic treatment of naïve mice with prostaglandin E2 (PGE2) expands HSPCs. However, the cellular mechanisms mediating this expansion remain unknown. Here, we demonstrate that PGE2 treatment in naïve mice inhibits apoptosis of HSPCs without changing their proliferation rate. In a murine model of sublethal total body irradiation (TBI), in which HSPCs are rapidly lost, treatment with a long-acting PGE2 analog (dmPGE2) reversed the apoptotic program initiated by TBI. dmPGE2 treatment in vivo decreased the loss of functional HSPCs following radiation injury, as demonstrated both phenotypically and by their increased reconstitution capacity. The antiapoptotic effect of dmPGE2 on HSPCs did not impair their ability to differentiate in vivo, resulting instead in improved hematopoietic recovery after TBI. dmPGE2 also increased microenvironmental cyclooxygenase-2 expression and expanded the α-smooth muscle actin-expressing subset of marrow macrophages, thus enhancing the bone marrow microenvironmental response to TBI. Therefore, in vivo treatment with PGE2 analogs may be particularly beneficial to HSPCs in the setting of injury by targeting them both directly and also through their niche. The current data provide rationale for in vivo manipulation of the HSPC pool as a strategy to improve recovery after myelosuppression.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Dinoprostona/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Actinas/genética , Actinas/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Células de la Médula Ósea/patología , Células de la Médula Ósea/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Microambiente Celular/efectos de los fármacos , Microambiente Celular/efectos de la radiación , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Dinoprostona/análogos & derivados , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Macrófagos/patología , Macrófagos/efectos de la radiación , Masculino , Ratones , Ratones Transgénicos , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/patología , Irradiación Corporal Total
8.
bioRxiv ; 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39185193

RESUMEN

Interactions between acute myeloid leukemia (AML) and the bone marrow microenvironment (BMME) are critical to leukemia progression and chemoresistance. Altered metabolite levels in the tumor microenvironment contribute to immunosuppression in solid tumors, while this has not been studied yet in the leukemic BMME. Metabolomics of AML patient bone marrow serum detected elevated metabolites, including lactate, compared to age- and sex-matched controls. Excess lactate has been implicated in solid tumors for inducing suppressive tumor-associated macrophages (TAMs) and correlates with poor prognosis. We describe the role of lactate in the polarization of leukemia-associated macrophages (LAMs) using a murine model of blast crisis chronic myelogenous leukemia (bcCML) and mice genetically lacking the lactate receptor GPR81. LAMs were CD206hi and suppressive in transcriptomics and cytokine profiling. Yet, LAMs had a largely unique expression profile from other types of TAMs. We demonstrate GPR81 signaling as a mechanism of both LAM polarization and the direct support of leukemia cell growth and self-repopulation. Furthermore, LAMs and elevated lactate diminished the function of hematopoietic progenitors and stromal support, while knockout of GPR81 had modest protective effects on the hematopoietic system. We report microenvironmental lactate as a critical driver of AML-induced immunosuppression and leukemic progression, thus identifying GPR81 signaling as an exciting and novel therapeutic target for treating this devastating disease.

9.
bioRxiv ; 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37066307

RESUMEN

Mesenchymal stem/stromal cells (MSCs) within the bone marrow microenvironment (BMME) support normal hematopoietic stem and progenitor cells (HSPCs). However, the heterogeneity of human MSCs has limited the understanding of their contribution to clonal dynamics and evolution to myelodysplastic syndromes (MDS). We combined three MSC cell surface markers, CD271, VCAM-1 (Vascular Cell Adhesion Molecule-1) and CD146, to isolate distinct subsets of human MSCs from bone marrow aspirates of healthy controls (Control BM). Based on transcriptional and functional analysis, CD271+CD106+CD146+ (NGFR+/VCAM1+/MCAM+/Lin-; NVML) cells display stem cell characteristics, are compatible with murine BM-derived Leptin receptor positive MSCs and provide superior support for normal HSPCs. MSC subsets from 17 patients with MDS demonstrated shared transcriptional changes in spite of mutational heterogeneity in the MDS clones, with loss of preferential support of normal HSPCs by MDS-derived NVML cells. Our data provide a new approach to dissect microenvironment-dependent mechanisms regulating clonal dynamics and progression of MDS.

10.
Front Bioeng Biotechnol ; 10: 855777, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795163

RESUMEN

Hematopoiesis takes place in the bone marrow and is supported by a complex cellular and molecular network in the bone marrow microenvironment. Commonly used models of the human bone marrow microenvironment include murine models and two-dimensional and three-dimensional tissue cultures. While these model systems have led to critical advances in the field, they fail to recapitulate many aspects of the human bone marrow. This has limited our understanding of human bone marrow pathophysiology and has led to deficiencies in therapy for many bone marrow pathologies such as bone marrow failure syndromes and leukemias. Therefore, we have developed a modular murine bone marrow microenvironment-on-chip using a commercially available microfluidic platform. This model includes a vascular channel separated from the bone marrow channel by a semi-porous membrane and incorporates critical components of the bone marrow microenvironment, including osteoblasts, endothelial cells, mesenchymal stem cells, and hematopoietic stem and progenitor cells. This system is capable of maintaining functional hematopoietic stem cells in vitro for at least 14 days at frequencies similar to what is found in the primary bone marrow. The modular nature of this system and its accessibility will allow for acceleration of our understanding of the bone marrow.

11.
Adv Ther (Weinh) ; 5(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35097186

RESUMEN

Micheliolide (MCL) is a naturally occurring sesquiterpene lactone that selectively targets leukemic stem cells (LSCs), which persist after conventional chemotherapy for myeloid leukemias, leading to disease relapse. To overcome modest MCL cytotoxicity, analogs with ≈two-threefold greater cytotoxicity against LSCs are synthesized via late-stage chemoenzymatic C-H functionalization. To enhance bone marrow delivery, MCL analogs are entrapped within bone-targeted polymeric nanoparticles (NPs). Robust drug loading capacities of up to 20% (mg drug mg-1 NP) are obtained, with release dominated by analog hydrophobicity. NPs loaded with a hydrolytically stable analog are tested in a leukemic mouse model. Median survival improved by 13% and bone marrow LSCs are decreased 34-fold following NPMCL treatments versus controls. Additionally, selective leukemic cell and LSC cytotoxicity of the treatment versus normal hematopoietic cells is observed. Overall, these studies demonstrate that MCL-based antileukemic agents combined with bone-targeted NPs offer a promising strategy for eradicating LSCs.

12.
Blood ; 114(19): 4054-63, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19726721

RESUMEN

Microenvironmental signals can determine hematopoietic stem cell (HSC) fate choices both directly and through stimulation of niche cells. In the bone marrow, prostaglandin E(2) (PGE(2)) is known to affect both osteoblasts and osteoclasts, whereas in vitro it expands HSCs and affects differentiation of hematopoietic progenitors. We hypothesized that in vivo PGE(2) treatment could expand HSCs through effects on both HSCs and their microenvironment. PGE(2)-treated mice had significantly decreased number of bone trabeculae, suggesting disruption of their microarchitecture. In addition, in vivo PGE(2) increased lineage(-) Sca-1(+) c-kit(+) bone marrow cells without inhibiting their differentiation. However, detailed immunophenotyping demonstrated a PGE(2)-dependent increase in short-term HSCs/multipotent progenitors (ST-HSCs/MPPs) only. Bone marrow cells transplanted from PGE(2) versus vehicle-treated donors had superior lymphomyeloid reconstitution, which ceased by 16 weeks, also suggesting that ST-HSCs were preferentially expanded. This was confirmed by serial transplantation studies. Thus in vivo PGE(2) treatment, probably through a combination of direct and microenvironmental actions, preferentially expands ST-HSCs in the absence of marrow injury, with no negative impact on hematopoietic progenitors or long-term HSCs. These novel effects of PGE(2) could be exploited clinically to increase donor ST-HSCs, which are highly proliferative and could accelerate hematopoietic recovery after stem cell transplantation.


Asunto(s)
Dinoprostona/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Supervivencia de Injerto/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos
13.
Methods Mol Biol ; 2230: 467-477, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33197034

RESUMEN

The adult hematopoietic system is repopulated in its entirety from a rare cell type known as hematopoietic stem cells (HSCs) that reside in the marrow space throughout the skeletal system. Here we describe the isolation and identification of HSCs both phenotypically and functionally.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Adulto , Animales , Huesos/citología , Humanos , Ratones
14.
JBMR Plus ; 5(10): e10516, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34693187

RESUMEN

The bone marrow microenvironment (BMME) regulates hematopoiesis through a complex network of cellular and molecular components. Hematologic malignancies reside within, and extensively interact with, the same BMME. These interactions consequently alter both malignant and benign hematopoiesis in multiple ways, and can encompass initiation of malignancy, support of malignant progression, resistance to chemotherapy, and loss of normal hematopoiesis. Herein, we will review supporting studies for interactions of the BMME with hematologic malignancies and discuss challenges still facing this exciting field of research. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

15.
Bone ; 119: 8-12, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29778716

RESUMEN

Hematopoietic stem cells (HSCs) require a supportive microenvironment to regulate their function and produce sufficient hematopoietic cells over the lifetime of an individual. With the exception of fish, all vertebrates, including mammals, maintain HSCs in a complex niche within the bone marrow. Several bone specific cellular populations have been implicated as components of the HSC niche and are part of a complex network that regulates HSC functions. However, the full extent of interactions within the HSC niche, and the role of individual cell populations remain to be fully elucidated. Further, it is not clear why fish are the exception, and what advantage is gained by housing HSCs in the bone marrow. To gain a better understanding of hematopoiesis and the mechanisms that drive hematopoietic disease processes a clearer picture of the complex HSC regulatory interactions in the bone marrow microenvironment is required.


Asunto(s)
Huesos/citología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre , Animales , Células de la Médula Ósea/citología , Humanos , Modelos Biológicos , Osteoblastos/citología
16.
Int J Radiat Biol ; 95(11): 1447-1461, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31329495

RESUMEN

Purpose: Incidents, such as nuclear facility accidents and the release of a 'dirty bomb', might result in not only external irradiation of personnel, but additional internal exposures through concomitant inhalation and/or ingestion of radioactive particulates. The purpose of this study was to define the impact of such a combination of radiation injuries on the hematopoietic niche.Material and methods: To assess changes in the murine hematopoietic system, we used a combined exposure of total body irradiation (TBI, 6 Gy) followed immediately by an internal (intraperitoneal) administration of 100 µCi of soluble 137Cs. We then evaluated acute survival in combined versus single modality exposure groups, as well as assessing hematopoietic function at 12 and 26 week time points.Results: Acutely, the combination of external and internal exposures led to an unexpected delay in excretion of 137Cs, increasing the absorbed dose in the combined exposure group and leading to mortality from an acute hematopoietic syndrome. At 12 weeks, all exposure paradigms resulted in decreased numbers of phenotypic hematopoietic stem cells (HSCs), particularly the short-term HSCs (ST-HSC); long-term HSCs (LT-HSC) were depleted only in the internal and combined exposure groups. At 26 weeks, there was significant anemia in both the TBI alone and combined exposure groups. There were decreased numbers in both the LT- and ST-HSCs and decreased functionality, as measured by competitive repopulation, was seen in all radiation groups, with the greatest effects seen in the internal and combined exposure groups.Conclusions: Our data indicate that a combined injury of sublethal external irradiation with internal contamination induces significant and persistent changes in the hematopoietic system, as may have been predicted from the literature and our own group's findings. However, a novel observation was that the combined exposure led to an alteration in the excretion kinetics of the internal contamination, increasing the acute effects beyond those anticipated. As a result, we believe that a combined exposure poses a unique challenge to the medical community during both the acute and, possibly, delayed recovery stages.


Asunto(s)
Médula Ósea/efectos de la radiación , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Irradiación Corporal Total , Animales , Células Cultivadas , Radioisótopos de Cesio , Femenino , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Fenotipo , Traumatismos Experimentales por Radiación/fisiopatología , Especies Reactivas de Oxígeno/metabolismo
17.
JCI Insight ; 52019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30998506

RESUMEN

The bone marrow microenvironment (BMME) contributes to the regulation of hematopoietic stem cell (HSC) function, though its role in age-associated lineage skewing is poorly understood. Here we show that dysfunction of aged marrow macrophages (Mφs) directs HSC platelet-bias. Mφs from the marrow of aged mice and humans exhibited an activated phenotype, with increased expression of inflammatory signals. Aged marrow Mφs also displayed decreased phagocytic function. Senescent neutrophils, typically cleared by marrow Mφs, were markedly increased in aged mice, consistent with functional defects in Mφ phagocytosis and efferocytosis. In aged mice, Interleukin 1B (IL1B) was elevated in the bone marrow and caspase 1 activity, which can process pro-IL1B, was increased in marrow Mφs and neutrophils. Mechanistically, IL1B signaling was necessary and sufficient to induce a platelet bias in HSCs. In young mice, depletion of phagocytic cell populations or loss of the efferocytic receptor Axl expanded platelet-biased HSCs. Our data support a model wherein increased inflammatory signals and decreased phagocytic function of aged marrow Mφs induce the acquisition of platelet bias in aged HSCs. This work highlights the instructive role of Mφs and IL1B in the age-associated lineage-skewing of HSCs, and reveals the therapeutic potential of their manipulation as antigeronic targets.


Asunto(s)
Envejecimiento/fisiología , Plaquetas/metabolismo , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Animales , Médula Ósea/patología , Caspasa 1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos , Fagocitosis , Fenotipo , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Tirosina Quinasa del Receptor Axl
18.
Nat Commun ; 9(1): 4239, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315161

RESUMEN

Inv(3q26) and t(3:3)(q21;q26) are specific to poor-prognosis myeloid malignancies, and result in marked overexpression of EVI1, a zinc-finger transcription factor and myeloid-specific oncoprotein. Despite extensive study, the mechanism by which EVI1 contributes to myeloid malignancy remains unclear. Here we describe a new mouse model that mimics the transcriptional effects of 3q26 rearrangement. We show that EVI1 overexpression causes global distortion of hematopoiesis, with suppression of erythropoiesis and lymphopoiesis, and marked premalignant expansion of myelopoiesis that eventually results in leukemic transformation. We show that myeloid skewing is dependent on DNA binding by EVI1, which upregulates Spi1, encoding master myeloid regulator PU.1. We show that EVI1 binds to the -14 kb upstream regulatory element (-14kbURE) at Spi1; knockdown of Spi1 dampens the myeloid skewing. Furthermore, deletion of the -14kbURE at Spi1 abrogates the effects of EVI1 on hematopoietic stem cells. These findings support a novel mechanism of leukemogenesis through EVI1 overexpression.


Asunto(s)
Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Alelos , Animales , Apoptosis/genética , Apoptosis/fisiología , Línea Celular , Proliferación Celular/genética , Proliferación Celular/fisiología , Citometría de Flujo , Hematopoyesis/genética , Hematopoyesis/fisiología , Proteína del Locus del Complejo MDS1 y EV11/genética , Ratones , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética
19.
Sci Rep ; 8(1): 14691, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30279500

RESUMEN

The chemokine CCL3 is frequently overexpressed in malignancies and overexpression leads to microenvironmental dysfunction. In murine models of chronic myelogenous leukemia (CML), CCL3 is critical for the maintenance of a leukemia stem cell population, and leukemia progression. With CCL3 implicated as a potentially viable therapeutic target, it is important to carefully characterize its role in normal hematopoietic homeostasis. CCL3-/- mice were used to evaluate the role of CCL3 in regulating hematopoietic stem and progenitor cell (HSPC) populations. CCL3-/- mice had loss of mature myeloid populations, while myeloid progenitors and HSPCs were increased, and microenvironmental populations were unchanged. These data show that CCL3 promotes myeloid lineage differentiation and the size of the HSPC pool independent of the supportive bone marrow microenvironment. Our results demonstrate a previously unrecognized role of CCL3 in the maintenance of homeostatic hematopoiesis that should be evaluated when targeting CCL3 signaling for the treatment of hematologic malignancy.


Asunto(s)
Diferenciación Celular , Quimiocina CCL3/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Células Mieloides/efectos de los fármacos , Células Mieloides/fisiología , Animales , Recuento de Células , Quimiocina CCL3/deficiencia , Ratones , Ratones Noqueados
20.
J Bone Miner Res ; 32(6): 1320-1331, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28277610

RESUMEN

Notch signaling is critical for osteoblastic differentiation; however, the specific contribution of individual Notch ligands is unknown. Parathyroid hormone (PTH) regulates the Notch ligand Jagged1 in osteoblastic cells. To determine if osteolineage Jagged1 contributes to bone homeostasis, selective deletion of Jagged1 in osteolineage cells was achieved through the presence of Prx1 promoter-driven Cre recombinase expression, targeting mesenchymal stem cells (MSCs) and their progeny (PJag1 mice). PJag1 mice were viable and fertile and did not exhibit any skeletal abnormalities at 2 weeks of age. At 2 months of age, however, PJag1 mice had increased trabecular bone mass compared to wild-type (WT) littermates. Dynamic histomorphometric analysis showed increased osteoblastic activity and increased mineral apposition rate. Immunohistochemical analysis showed increased numbers of osteocalcin-positive mature osteoblasts in PJag1 mice. Also increased phenotypically defined Lin- /CD45- /CD31- /Sca1- /CD51+ osteoblastic cells were measured by flow cytometric analysis. Surprisingly, phenotypically defined Lin- /CD45- /CD31- /Sca1+ /CD51+ MSCs were unchanged in PJag1 mice as measured by flow cytometric analysis. However, functional osteoprogenitor (OP) cell frequency, measured by Von Kossa+ colony formation, was decreased, suggesting that osteolineage Jagged1 contributes to maintenance of the OP pool. The trabecular bone increases were not due to osteoclastic defects, because PJag1 mice had increased bone resorption. Because PTH increases osteoblastic Jagged1, we sought to understand if osteolineage Jagged1 modulates PTH-mediated bone anabolism. Intermittent PTH treatment resulted in a significantly greater increase in BV/TV in PJag1 hind limbs compared to WT. These findings demonstrate a critical role of osteolineage Jagged1 in bone homeostasis, where Jagged1 maintains the transition of OP to maturing osteoblasts. This novel role of Jagged1 not only identifies a regulatory loop maintaining appropriate populations of osteolineage cells, but also provides a novel approach to increase trabecular bone mass, particularly in combination with PTH, through modulation of Jagged1. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Linaje de la Célula , Proteína Jagged-1/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Resorción Ósea/metabolismo , Resorción Ósea/patología , Hueso Esponjoso/citología , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/metabolismo , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Placa de Crecimiento/citología , Placa de Crecimiento/diagnóstico por imagen , Placa de Crecimiento/metabolismo , Ligandos , Ratones , Modelos Biológicos , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/farmacología , Células Madre/efectos de los fármacos , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA