Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(4): 114049, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573853

RESUMEN

Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.


Asunto(s)
Linfangiogénesis , Células Madre Mesenquimatosas , Osificación Heterotópica , Factor C de Crecimiento Endotelial Vascular , Animales , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Osificación Heterotópica/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Ratones , Células Madre Mesenquimatosas/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Diferenciación Celular , Tenocitos/metabolismo , Osteogénesis , Haploinsuficiencia , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino
2.
Cytotherapy ; 26(5): 498-505, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38372680

RESUMEN

BACKGROUND AIMS: Mesenchymal stem/stromal cells (MSCs) are defined as culture-expanded populations, and although these cells recapitulate many properties of bone marrow (BM) resident skeletal stem/progenitor cells, few studies have directly compared these populations to evaluate how culture adaptation and expansion impact critical quality attributes. METHODS: We analyzed by RNA sequencing Lin-SCA1+ MSCs enriched from BM by immunodepletion (ID) and after subsequent culture expansion (Ex) and Lin-LEPR+ MSCs sorted (S) directly from BM. Pairwise comparisons were used to identify differentially expressed genes (DEGs) between populations, and gene set enrichment analysis was employed to identify biological pathways/processes unique to each population. K-means cluster analysis resolved isolation status-dependent changes in transcription in pseudotime. RESULTS: Hierarchical clustering segregated populations by isolation process, and principal component analysis identified transcripts related to vasculature development, ossification and inflammatory/cytokine signaling as key drivers of population variance. Pairwise comparisons identified 3849 DEGs in ID versus S BM-MSCs mapping to Gene Ontology (GO) terms related to immune and metabolic processes and 334 DEGs in Ex versus ID BM-MSCs mapping to GO terms related to tissue development, cell growth and replication and organelle organization. K-means cluster analysis revealed significant differences in transcripts encoding stemness and differentiation markers, extracellular matrix structural constituents and remodeling enzymes and paracrine-acting factors between populations. CONCLUSIONS: These comparative analyses reveal significant differences in gene expression signatures between BM resident and culture-expanded MSCs, thereby providing new insight into how culture adaptation/expansion endows the latter with unique quality attributes.


Asunto(s)
Células de la Médula Ósea , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas , Transcriptoma , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Ratones , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Transcriptoma/genética , Diferenciación Celular/genética , Células Cultivadas , Proliferación Celular/genética , Ratones Endogámicos C57BL
3.
Front Cell Dev Biol ; 11: 1294438, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37965574

RESUMEN

Introduction: In the rapidly aging U.S. population, age-induced bone loss (senile osteoporosis) represents a major public health concern that is associated with a significant increased risk for low trauma fragility fractures, which are debilitating to patients, cause significant morbidity and mortality, and are costly to treat and manage. While various treatments exist to slow bone loss in osteoporosis patients, these suffer from poor tolerability and label restrictions that limit their overall effectiveness. Over the past decade, skeletal stem/progenitor cells (SSPCs), which are the main precursor of osteoblasts and adipocytes in adult bone marrow (BM), have emerged as important players in osteoporosis. Methods: Age-induced skeletal pathology was quantified in elderly (24-month-old) vs. mature (3-month-old) mice by micro-CT and changes in SSPC abundance in the BM of these mice was quantified by fluorescence-activated cell sorting (FACS). SSPCs from elderly vs. mature mice were also analyzed by RNA-Seq to identify differentially expressed genes (DEGs), and gain and loss-of-function studies were performed in human BM-derived mesenchymal stromal cells (BM-MSCs) to assess A2M function. Results: Elderly mice were shown to exhibit significant age-induced skeletal pathology, which correlated with a significant increase in SSPC abundance in BM. RNA-seq analysis identified alpha-2-macroglobulin (A2M), a pan-protease inhibitor that also binds inflammatory cytokines, as one of the most downregulated transcripts in SSPCs isolated from the BM of elderly vs. mature mice, and silencing of A2M expression in human BM-MSCs induced their proliferation and skewed their lineage bifurcation toward adipogenesis at the expense of osteogenesis thereby recapitulating critical aspects of age-induced stem cell dysfunction. Conclusion: These findings identify A2M as a novel disease modifying protein in osteoporosis, downregulation of which in bone marrow promotes SSPC dysfunction and imbalances in skeletal homeostasis.

4.
Stem Cells ; 41(12): 1185-1200, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-37665974

RESUMEN

Despite extensive clinical testing, mesenchymal stem/stromal cell (MSC)-based therapies continue to underperform with respect to efficacy, which reflects the paucity of biomarkers that predict potency prior to patient administration. Previously, we reported that TWIST1 predicts inter-donor differences in MSC quality attributes that confer potency. To define the full spectrum of TWIST1 activity in MSCs, the present work employed integrated omics-based profiling to identify a high-confidence set of TWIST1 targets, which mapped to cellular processes related to ECM structure/organization, skeletal and circulatory system development, interferon gamma signaling, and inflammation. These targets are implicated in contributing to both stem/progenitor and paracrine activities of MSCs indicating these processes are linked mechanistically in a TWIST1-dependent manner. Targets implicated in extracellular matrix dynamics further implicate TWIST1 in modulating cellular responses to niche remodeling. Novel TWIST1-regulated genes identified herein may be prioritized for future mechanistic and functional studies.


Asunto(s)
Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Biomarcadores/metabolismo , Matriz Extracelular/metabolismo , Unión Proteica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
5.
Cytotherapy ; 25(4): 362-368, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36481320

RESUMEN

BACKGROUND AIMS: Fanconi anemia (FA) is an inherited bone marrow failure syndrome caused by defects in the repair of DNA inter-strand crosslinks and manifests as aplastic anemia, myelodysplastic syndrome and acute myeloid leukemia. FA also causes defects in mesenchymal stromal cell (MSC) function, but how different FA gene mutations alter function remains understudied. METHODS: We compared the growth, differentiation and transcript profile of a single MSC isolate from an asymptomatic patient with FA with a FANCG nonsense mutation who underwent hematopoietic stem cell transplantation 10 years prior to that from a representative healthy donor (HD). RESULTS: We show that FANCG-/- MSCs exhibit rapid onset of growth cessation, skewed bi-lineage differentiation in favor of adipogenesis and increased cellular oxidate stress consistent with an aging-like phenotype. Transcript profiling identified pathways related to cell growth, senescence, cellular stress responses and DNA replication/repair as over-represented in FANCG-/- MSC, and real-time polymerase chain reaction confirmed these MSCs expressed reduced levels of transcripts implicated in cell growth (TWIST1, FGFR2v7-8) and osteogenesis (TWIST1, RUNX2) and increased levels of transcripts regulating adipogenesis (GPR116) and insulin signaling. They also expressed reduced levels of mRNAs implicated in HSC self-maintenance and homing (KITLG, HGF, GDNF, PGF, CFB, IL-1B and CSF1) and elevated levels of those implicated in myelodysplasia (IL-6, GDF15). CONCLUSIONS: Together, these findings demonstrate how inactivation of FANCG impacts MSC behavior, which parallels observed defects in osteogenesis, HSC depletion and leukemic blast formation seen in patients with FA.


Asunto(s)
Anemia de Fanconi , Células Madre Mesenquimatosas , Síndromes Mielodisplásicos , Humanos , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Anemia de Fanconi/metabolismo , Síndromes Mielodisplásicos/genética , Hematopoyesis/genética , Fenotipo , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo
6.
Biology (Basel) ; 11(9)2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36138736

RESUMEN

Obesity and type II diabetes mellitus (T2DM) are prominent risk factors for secondary osteoporosis due to the negative impacts of hyperglycemia and excessive body fat on bone metabolism. While the armamentarium of anti-diabetic drugs is expanding, their negative or unknown impacts on bone metabolism limits effectiveness. The inactivation of inositol hexakisphosphate kinase 1 (IP6K1) protects mice from high-fat-diet (HFD)-induced obesity (DIO) and insulin resistance by enhancing thermogenic energy expenditure, but the role of this kinase and the consequences of its inhibition on bone metabolism are unknown. To determine if IP6K1 inhibition in obese mice affords protection against obesity-induced metabolic derangements and bone loss, we maintained 2-month-old mice on a normal chow control diet or HFD under thermal neutral conditions for 100 d. Beginning on day 40, HFD-fed mice were divided into two groups and administered daily injections of vehicle or the pan-IP6K inhibitor TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl) purine]. HFD-fed mice developed obesity, hyperglycemia, hyperlipidemia, and secondary osteoporosis, while TNP administration protected mice against HFD-induced metabolic and lipid derangements and preserved bone mass, mineral density, and trabecular microarchitecture, which correlated with reduced serum leptin levels, reduced marrow adiposity, and preservation of marrow resident skeletal stem/progenitor cells (SSPCs). TNP also exhibited hypotensive activity, an unrealized benefit of the drug, and its prolonged administration had no adverse impacts on spermatogenesis. Together, these data indicate that the inhibition of IP6K1 using selective inhibitors, such as TNP, may provide an effective strategy to manage obesity and T2DM due to its bone sparing effects.

8.
NPJ Microgravity ; 7(1): 49, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34836964

RESUMEN

Disuse osteoporosis (DO) results from mechanical unloading of weight-bearing bones and causes structural changes that compromise skeletal integrity, leading to increased fracture risk. Although bone loss in DO results from imbalances in osteoblast vs. osteoclast activity, its effects on skeletal stem/progenitor cells (SSCs) is indeterminate. We modeled DO in mice by 8 and 14 weeks of hindlimb unloading (HU) or 8 weeks of unloading followed by 8 weeks of recovery (HUR) and monitored impacts on animal physiology and behavior, metabolism, marrow adipose tissue (MAT) volume, bone density and micro-architecture, and bone marrow (BM) leptin and tyrosine hydroxylase (TH) protein expression, and correlated multi-systems impacts of HU and HUR with the transcript profiles of Lin-LEPR+ SSCs and mesenchymal stem cells (MSCs) purified from BM. Using this integrative approach, we demonstrate that prolonged HU induces muscle atrophy, progressive bone loss, and MAT accumulation that paralleled increases in BM but not systemic leptin levels, which remained low in lipodystrophic HU mice. HU also induced SSC quiescence and downregulated bone anabolic and neurogenic pathways, which paralleled increases in BM TH expression, but had minimal impacts on MSCs, indicating a lack of HU memory in culture-expanded populations. Although most impacts of HU were reversed by HUR, trabecular micro-architecture remained compromised and time-resolved changes in the SSC transcriptome identified various signaling pathways implicated in bone formation that were unresponsive to HUR. These findings indicate that HU-induced alterations to the SSC transcriptome that persist after reloading may contribute to poor bone recovery.

9.
Cell Death Differ ; 25(4): 679-692, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29311623

RESUMEN

Marrow-resident mesenchymal stem cells (MSCs) serve as a functional component of the perivascular niche that regulates hematopoiesis. They also represent the main source of bone formed in adult bone marrow, and their bifurcation to osteoblast and adipocyte lineages plays a key role in skeletal homeostasis and aging. Although the tumor suppressor p53 also functions in bone organogenesis, homeostasis, and neoplasia, its role in MSCs remains poorly described. Herein, we examined the normal physiological role of p53 in primary MSCs cultured under physiologic oxygen levels. Using knockout mice and gene silencing we show that p53 inactivation downregulates expression of TWIST2, which normally restrains cellular differentiation to maintain wild-type MSCs in a multipotent state, depletes mitochondrial reactive oxygen species (ROS) levels, and suppresses ROS generation and PPARG gene and protein induction in response to adipogenic stimuli. Mechanistically, this loss of adipogenic potential skews MSCs toward an osteogenic fate, which is further potentiated by TWIST2 downregulation, resulting in highly augmented osteogenic differentiation. We also show that p53-/- MSCs are defective in supporting hematopoiesis as measured in standard colony assays because of decreased secretion of various cytokines including CXCL12 and CSF1. Lastly, we show that transient exposure of wild-type MSCs to 21% oxygen upregulates p53 protein expression, resulting in increased mitochondrial ROS production and enhanced adipogenic differentiation at the expense of osteogenesis, and that treatment of cells with FGF2 mitigates these effects by inducing TWIST2. Together, these findings indicate that basal p53 levels are necessary to maintain MSC bi-potency, and oxygen-induced increases in p53 expression modulate cell fate and survival decisions. Because of the critical function of basal p53 in MSCs, our findings question the use of p53 null cell lines as MSC surrogates, and also implicate dysfunctional MSC responses in the pathophysiology of p53-related skeletal disorders.


Asunto(s)
Adipogénesis , Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Proteína p53 Supresora de Tumor/metabolismo , Animales , Supervivencia Celular , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
10.
Stem Cells ; 36(1): 7-10, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28960677

RESUMEN

Mesenchymal stem cells (MSCs) have gained widespread use in regenerative medicine due to their demonstrated efficacy in a broad range of experimental animal models of disease and their excellent safety profile in human clinical trials. Outcomes from these studies suggest that MSCs achieve therapeutic effects in vivo in nonhomologous applications predominantly by paracrine action. This paracrine-centric viewpoint has become widely entrenched in the field, and has spurred a campaign to rename MSCs as "medicinal signaling cells" to better reflect this mode of action. In this Commentary, we argue that the paracrine-centric viewpoint and proposed name change ignores a wealth of old and new data that unequivocally demonstrate the stem cell nature of MSCs, and also overlooks a large effort to exploit homologous applications of MSCs in human clinical trials. Furthermore, we offer evidence that a stem cell-centric viewpoint of MSCs provides a comprehensive understanding of MSC biology that encompasses their paracrine activity, and provides a better foundation to develop metrics that quantify the biological potency of MSC batches for both homologous and nonhomologous clinical applications. Stem Cells 2018;36:7-10.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas , Medicina Regenerativa/métodos , Diferenciación Celular , Humanos
11.
Stem Cells ; 35(8): 1973-1983, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28577302

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are the predominant source of bone and adipose tissue in adult bone marrow and play a critical role in skeletal homeostasis. Age-induced changes in bone marrow favor adipogenesis over osteogenesis leading to skeletal involution and increased marrow adiposity so pathways that prevent MSC aging are potential therapeutic targets for treating age-related bone diseases. Here, we show that inositol hexakisphosphate kinase 1 (Ip6k1) deletion in mice increases MSC yields from marrow and enhances cell growth and survival ex vivo. In response to the appropriate stimuli, Ip6k1-/- versus Ip6k1+/+ MSCs also exhibit enhanced osteogenesis and hematopoiesis-supporting activity and reduced adipogenic differentiation. Mechanistic-based studies revealed that Ip6k1-/- MSCs express higher MDM2 and lower p53 protein levels resulting in lower intrinsic mitochondrial reactive oxygen species (ROS) levels as compared to Ip6k1+/+ MSCs, but both populations upregulate mitochondrial ROS to similar extents in response to oxygen-induced stress. Finally, we show that mice fed a high fat diet exhibit reduced trabecular bone volume, and that pharmacological inhibition of IP6K1 using a pan-IP6K inhibitor largely reversed this phenotype while increasing MSC yields from bone marrow. Together, these findings reveal an important role for IP6K1 in regulating MSC fitness and differentiation fate. Unlike therapeutic interventions that target peroxisome proliferator-activated receptor gamma and leptin receptor activity, which yield detrimental side effects including increased fracture risk and altered feeding behavior, respectively, inhibition of IP6K1 maintains insulin sensitivity and prevents obesity while preserving bone integrity. Therefore, IP6K1 inhibitors may represent more effective insulin sensitizers due to their bone sparing properties. Stem Cells 2017;35:1973-1983.


Asunto(s)
Dieta Alta en Grasa , Células Madre Mesenquimatosas/enzimología , Músculo Esquelético/patología , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Adipogénesis , Animales , Médula Ósea/metabolismo , Proliferación Celular , Supervivencia Celular , Eliminación de Gen , Hematopoyesis , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis , Estrés Oxidativo , Fosfotransferasas (Aceptor del Grupo Fosfato)/antagonistas & inhibidores , Fosfotransferasas (Aceptor del Grupo Fosfato)/deficiencia
12.
J Clin Invest ; 126(4): 1173-80, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27035808

RESUMEN

Almost all cell types release extracellular vesicles (EVs), which are derived either from multivesicular bodies or from the plasma membrane. EVs contain a subset of proteins, lipids, and nucleic acids from the cell from which they are derived. EV factors, particularly small RNAs such as miRNAs, likely play important roles in cell-to-cell communication both locally and systemically. Most of the functions associated with EVs are in the regulation of immune responses to pathogens and cancer, as well as in regulating autoimmunity. This Review will focus on the different modes of immune regulation, both direct and indirect, by EVs. The therapeutic utility of EVs for the regulation of immune responses will also be discussed.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Micropartículas Derivadas de Células/inmunología , Exosomas/inmunología , MicroARNs/inmunología , Neoplasias/inmunología , ARN Neoplásico/inmunología , Animales , Enfermedades Autoinmunes/patología , Micropartículas Derivadas de Células/patología , Enfermedad Crónica , Exosomas/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...