Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 186(10): 2127-2143.e22, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37098344

RESUMEN

Pathogen infection and tissue injury are universal insults that disrupt homeostasis. Innate immunity senses microbial infections and induces cytokines/chemokines to activate resistance mechanisms. Here, we show that, in contrast to most pathogen-induced cytokines, interleukin-24 (IL-24) is predominately induced by barrier epithelial progenitors after tissue injury and is independent of microbiome or adaptive immunity. Moreover, Il24 ablation in mice impedes not only epidermal proliferation and re-epithelialization but also capillary and fibroblast regeneration within the dermal wound bed. Conversely, ectopic IL-24 induction in the homeostatic epidermis triggers global epithelial-mesenchymal tissue repair responses. Mechanistically, Il24 expression depends upon both epithelial IL24-receptor/STAT3 signaling and hypoxia-stabilized HIF1α, which converge following injury to trigger autocrine and paracrine signaling involving IL-24-mediated receptor signaling and metabolic regulation. Thus, parallel to innate immune sensing of pathogens to resolve infections, epithelial stem cells sense injury signals to orchestrate IL-24-mediated tissue repair.


Asunto(s)
Citocinas , Heridas y Lesiones , Animales , Ratones , Inmunidad Adaptativa , Quimiocinas , Epidermis , Inmunidad Innata , Heridas y Lesiones/inmunología
2.
Annu Rev Cell Dev Biol ; 32: 649-675, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27576121

RESUMEN

In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.


Asunto(s)
Vasos Sanguíneos/metabolismo , Hematopoyesis , Osteogénesis , Transducción de Señal , Animales , Médula Ósea/irrigación sanguínea , Células Endoteliales/metabolismo , Humanos
3.
Nat Immunol ; 13(11): 1072-82, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22983360

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are regulated by various bone marrow stromal cell types. Here we identified rare activated bone marrow monocytes and macrophages with high expression of α-smooth muscle actin (α-SMA) and the cyclooxygenase COX-2 that were adjacent to primitive HSPCs. These myeloid cells resisted radiation-induced cell death and further upregulated COX-2 expression under stress conditions. COX-2-derived prostaglandin E(2) (PGE(2)) prevented HSPC exhaustion by limiting the production of reactive oxygen species (ROS) via inhibition of the kinase Akt and higher stromal-cell expression of the chemokine CXCL12, which is essential for stem-cell quiescence. Our study identifies a previously unknown subset of α-SMA(+) activated monocytes and macrophages that maintain HSPCs and protect them from exhaustion during alarm situations.


Asunto(s)
Actinas/inmunología , Médula Ósea/inmunología , Células Madre Hematopoyéticas/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Actinas/genética , Animales , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Comunicación Celular/genética , Comunicación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Supervivencia Celular/efectos de la radiación , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Dinoprostona/biosíntesis , Dinoprostona/inmunología , Rayos gamma , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/efectos de la radiación , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Macrófagos/citología , Macrófagos/efectos de la radiación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/efectos de la radiación , Ratones , Monocitos/citología , Monocitos/efectos de la radiación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación
4.
Proc Natl Acad Sci U S A ; 117(10): 5339-5350, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094197

RESUMEN

Aging manifests with architectural alteration and functional decline of multiple organs throughout an organism. In mammals, aged skin is accompanied by a marked reduction in hair cycling and appearance of bald patches, leading researchers to propose that hair follicle stem cells (HFSCs) are either lost, differentiate, or change to an epidermal fate during aging. Here, we employed single-cell RNA-sequencing to interrogate aging-related changes in the HFSCs. Surprisingly, although numbers declined, aging HFSCs were present, maintained their identity, and showed no overt signs of shifting to an epidermal fate. However, they did exhibit prevalent transcriptional changes particularly in extracellular matrix genes, and this was accompanied by profound structural perturbations in the aging SC niche. Moreover, marked age-related changes occurred in many nonepithelial cell types, including resident immune cells, sensory neurons, and arrector pili muscles. Each of these SC niche components has been shown to influence HF regeneration. When we performed skin injuries that are known to mobilize young HFSCs to exit their niche and regenerate HFs, we discovered that aged skin is defective at doing so. Interestingly, however, in transplantation assays in vivo, aged HFSCs regenerated HFs when supported with young dermis, while young HFSCs failed to regenerate HFs when combined with aged dermis. Together, our findings highlight the importance of SC:niche interactions and favor a model where youthfulness of the niche microenvironment plays a dominant role in dictating the properties of its SCs and tissue health and fitness.


Asunto(s)
Folículo Piloso/fisiología , Regeneración/fisiología , Envejecimiento de la Piel/fisiología , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Dermis/fisiología , Células Epidérmicas/fisiología , Epidermis/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculos/fisiología , Repitelización , Regeneración/genética , Células Receptoras Sensoriales/fisiología , Envejecimiento de la Piel/genética , Nicho de Células Madre/genética , Trasplante de Células Madre , Transcriptoma , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
5.
Nature ; 532(7599): 323-8, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27074509

RESUMEN

Bone marrow endothelial cells (BMECs) form a network of blood vessels that regulate both leukocyte trafficking and haematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles, and whether these events occur at the same vascular site. We found that mammalian bone marrow stem cell maintenance and leukocyte trafficking are regulated by distinct blood vessel types with different permeability properties. Less permeable arterial blood vessels maintain haematopoietic stem cells in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the bone marrow. A functional consequence of high permeability of blood vessels is that exposure to blood plasma increases bone marrow HSPC ROS levels, augmenting their migration and differentiation, while compromising their long-term repopulation and survival. These findings may have relevance for clinical haematopoietic stem cell transplantation and mobilization protocols.


Asunto(s)
Vasos Sanguíneos/citología , Vasos Sanguíneos/fisiología , Médula Ósea/irrigación sanguínea , Hematopoyesis , Animales , Antígenos Ly/metabolismo , Arterias/citología , Arterias/fisiología , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Autorrenovación de las Células , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Células Endoteliales/fisiología , Femenino , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Leucocitos/citología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Nestina/metabolismo , Pericitos/fisiología , Permeabilidad , Plasma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR4/metabolismo
8.
Blood ; 120(9): 1843-55, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22645180

RESUMEN

Cytokine-induced expansion of hematopoietic stem and progenitor cells (HSPCs) is not fully understood. In the present study, we show that whereas steady-state hematopoiesis is normal in basic fibroblast growth factor (FGF-2)-knockout mice, parathyroid hormone stimulation and myeloablative treatments failed to induce normal HSPC proliferation and recovery. In vivo FGF-2 treatment expanded stromal cells, including perivascular Nestin(+) supportive stromal cells, which may facilitate HSPC expansion by increasing SCF and reducing CXCL12 via mir-31 up-regulation. FGF-2 predominantly expanded a heterogeneous population of undifferentiated HSPCs, preserving and increasing durable short- and long-term repopulation potential. Mechanistically, these effects were mediated by c-Kit receptor activation, STAT5 phosphorylation, and reduction of reactive oxygen species levels. Mice harboring defective c-Kit signaling exhibited abrogated HSPC expansion in response to FGF-2 treatment, which was accompanied by elevated reactive oxygen species levels. The results of the present study reveal a novel mechanism underlying FGF-2-mediated in vivo expansion of both HSPCs and their supportive stromal cells, which may be used to improve stem cell engraftment after clinical transplantation.


Asunto(s)
Proliferación Celular , Quimiocina CXCL12/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células del Estroma/metabolismo , Animales , Secuencia de Bases , Trasplante de Médula Ósea , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/genética , Regulación hacia Abajo/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Hormona Paratiroidea/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/metabolismo , Células del Estroma/efectos de los fármacos
9.
Curr Opin Hematol ; 20(3): 237-44, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23567340

RESUMEN

PURPOSE OF REVIEW: Fibroblast growth factor (FGF) signaling activates many bone marrow cell types, including various stem cells, osteoblasts, and osteoclasts. However, the role of FGF signaling in regulation of normal and leukemic stem cells is poorly understood. This review highlights the physiological roles of FGF signaling in regulating bone marrow mesenchymal and hematopoietic stem and progenitor cells (MSPCs and HSPCs) and their dynamic microenvironment. In addition, this review summarizes the recent studies which provide an overview of FGF-activated mechanisms regulating physiological stem cell maintenance, self-renewal, and motility. RECENT FINDINGS: Current results indicate that partial deficiencies in FGF signaling lead to mild defects in hematopoiesis and bone remodeling. However, FGF signaling was shown to be crucial for stem cell self-renewal and for proper hematopoietic poststress recovery. FGF signaling activation was shown to be important also for rapid AMD3100 or post 5-fluorouracil-induced HSPC mobilization. In vivo, FGF-2 administration successfully expanded both MSPCs and HSPCs. FGF-induced expansion was characterized by enhanced HSPC cycling without further exhaustion of the stem cell pool. In addition, FGF signaling expands and remodels the supportive MSPC niche cells. Finally, FGF signaling is constitutively activated in many leukemias, suggesting that malignant HSPCs exploit this pathway for their constant expansion and for remodeling a malignant-supportive microenvironment. SUMMARY: The summarized studies, concerning regulation of stem cells and their microenvironment, suggest that FGF signaling manipulation can serve to improve current clinical stem cell mobilization and transplantation protocols. In addition, it may help to develop therapies specifically targeting leukemic stem cells and their supportive microenvironment.


Asunto(s)
Remodelación Ósea/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Células Madre Hematopoyéticas/fisiología , Células Madre Mesenquimatosas/fisiología , Transducción de Señal/fisiología , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Mesenquimatosas/citología , Microambiente Tumoral/fisiología
10.
Cell Stem Cell ; 29(7): 1067-1082.e18, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35728595

RESUMEN

Barrier epithelia depend upon resident stem cells for homeostasis, defense, and repair. Epithelial stem cells of small and large intestines (ISCs) respond to their local microenvironments (niches) to fulfill a continuous demand for tissue turnover. The complexity of these niches and underlying communication pathways are not fully known. Here, we report a lymphatic network at the intestinal crypt base that intimately associates with ISCs. Employing in vivo loss of function and lymphatic:organoid cocultures, we show that crypt lymphatics maintain ISCs and inhibit their precocious differentiation. Pairing single-cell and spatial transcriptomics, we apply BayesPrism to deconvolve expression within spatial features and develop SpaceFold to robustly map the niche at high resolution, exposing lymphatics as a central signaling hub for the crypt in general and ISCs in particular. We identify WNT-signaling factors (WNT2, R-SPONDIN-3) and a hitherto unappreciated extracellular matrix protein, REELIN, as crypt lymphatic signals that directly govern the regenerative potential of ISCs.


Asunto(s)
Intestinos , Células Madre , Proliferación Celular , Mucosa Intestinal/metabolismo , Organoides , Transducción de Señal , Proteínas Wnt/metabolismo
11.
Nat Cell Biol ; 22(11): 1396, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33046885

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Nat Cell Biol ; 22(7): 779-790, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32451440

RESUMEN

Tissue stem cells are the cell of origin for many malignancies. Metabolites regulate the balance between self-renewal and differentiation, but whether endogenous metabolic pathways or nutrient availability predispose stem cells towards transformation remains unknown. Here, we address this question in epidermal stem cells (EpdSCs), which are a cell of origin for squamous cell carcinoma. We find that oncogenic EpdSCs are serine auxotrophs whose growth and self-renewal require abundant exogenous serine. When extracellular serine is limited, EpdSCs activate de novo serine synthesis, which in turn stimulates α-ketoglutarate-dependent dioxygenases that remove the repressive histone modification H3K27me3 and activate differentiation programmes. Accordingly, serine starvation or enforced α-ketoglutarate production antagonizes squamous cell carcinoma growth. Conversely, blocking serine synthesis or repressing α-ketoglutarate-driven demethylation facilitates malignant progression. Together, these findings reveal that extracellular serine is a critical determinant of EpdSC fate and provide insight into how nutrient availability is integrated with stem cell fate decisions during tumour initiation.


Asunto(s)
Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/patología , Células Epidérmicas/patología , Ácidos Cetoglutáricos/metabolismo , Serina/metabolismo , Células Madre/patología , Animales , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Células Epidérmicas/metabolismo , Femenino , Humanos , Masculino , Ratones , Células Madre/metabolismo
13.
Nat Cell Biol ; 22(6): 640-650, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32393888

RESUMEN

Tissue homeostasis and regeneration rely on resident stem cells (SCs), whose behaviour is regulated through niche-dependent crosstalk. The mechanisms underlying SC identity are still unfolding. Here, using spatiotemporal gene ablation in murine hair follicles, we uncover a critical role for the transcription factors (TFs) nuclear factor IB (NFIB) and IX (NFIX) in maintaining SC identity. Without NFI TFs, SCs lose their hair-regenerating capability, and produce skin bearing striking resemblance to irreversible human alopecia, which also displays reduced NFIs. Through single-cell transcriptomics, ATAC-Seq and ChIP-Seq profiling, we expose a key role for NFIB and NFIX in governing super-enhancer maintenance of the key hair follicle SC-specific TF genes. When NFIB and NFIX are genetically removed, the stemness epigenetic landscape is lost. Super-enhancers driving SC identity are decommissioned, while unwanted lineages are de-repressed ectopically. Together, our findings expose NFIB and NFIX as crucial rheostats of tissue homeostasis, functioning to safeguard the SC epigenome from a breach in lineage confinement that otherwise triggers irreversible tissue degeneration.


Asunto(s)
Alopecia/patología , Diferenciación Celular , Cromatina/metabolismo , Folículo Piloso/citología , Factores de Transcripción NFI/fisiología , Células Madre/citología , Alopecia/genética , Alopecia/metabolismo , Animales , Células Cultivadas , Cromatina/genética , Femenino , Folículo Piloso/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración , Células Madre/metabolismo
14.
Nat Commun ; 11(1): 3547, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669546

RESUMEN

Neutrophils provide first line of host defense against bacterial infections utilizing glycolysis for their effector functions. How glycolysis and its major byproduct lactate are triggered in bone marrow (BM) neutrophils and their contribution to neutrophil mobilization in acute inflammation is not clear. Here we report that bacterial lipopolysaccharides (LPS) or Salmonella Typhimurium triggers lactate release by increasing glycolysis, NADPH-oxidase-mediated reactive oxygen species and HIF-1α levels in BM neutrophils. Increased release of BM lactate preferentially promotes neutrophil mobilization by reducing endothelial VE-Cadherin expression, increasing BM vascular permeability via endothelial lactate-receptor GPR81 signaling. GPR81-/- mice mobilize reduced levels of neutrophils in response to LPS, unless rescued by VE-Cadherin disrupting antibodies. Lactate administration also induces release of the BM neutrophil mobilizers G-CSF, CXCL1 and CXCL2, indicating that this metabolite drives neutrophil mobilization via multiple pathways. Our study reveals a metabolic crosstalk between lactate-producing neutrophils and BM endothelium, which controls neutrophil mobilization under bacterial infection.


Asunto(s)
Células de la Médula Ósea/inmunología , Ácido Láctico/metabolismo , Neutrófilos/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Salmonella/inmunología , Animales , Médula Ósea/irrigación sanguínea , Células de la Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Femenino , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Receptores Acoplados a Proteínas G/genética , Infecciones por Salmonella/microbiología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
15.
Science ; 366(6470): 1218-1225, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31672914

RESUMEN

Tissues rely on stem cells (SCs) for homeostasis and wound repair. SCs reside in specialized microenvironments (niches) whose complexities and roles in orchestrating tissue growth are still unfolding. Here, we identify lymphatic capillaries as critical SC-niche components. In skin, lymphatics form intimate networks around hair follicle (HF) SCs. When HFs regenerate, lymphatic-SC connections become dynamic. Using a mouse model, we unravel a secretome switch in SCs that controls lymphatic behavior. Resting SCs express angiopoietin-like protein 7 (Angptl7), promoting lymphatic drainage. Activated SCs switch to Angptl4, triggering transient lymphatic dissociation and reduced drainage. When lymphatics are perturbed or the secretome switch is disrupted, HFs cycle precociously and tissue regeneration becomes asynchronous. In unearthing lymphatic capillaries as a critical SC-niche element, we have learned how SCs coordinate their activity across a tissue.


Asunto(s)
Folículo Piloso/fisiología , Vasos Linfáticos/fisiología , Regeneración , Nicho de Células Madre/fisiología , Células Madre/fisiología , Proteína 4 Similar a la Angiopoyetina/metabolismo , Proteína 7 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/metabolismo , Animales , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Madre/metabolismo , Proteínas Supresoras de Tumor/genética
16.
Elife ; 72018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30520726

RESUMEN

Adult stem cells are responsible for life-long tissue maintenance. They reside in and interact with specialized tissue microenvironments (niches). Using murine hair follicle as a model, we show that when junctional perturbations in the niche disrupt barrier function, adjacent stem cells dramatically change their transcriptome independent of bacterial invasion and become capable of directly signaling to and recruiting immune cells. Additionally, these stem cells elevate cell cycle transcripts which reduce their quiescence threshold, enabling them to selectively proliferate within this microenvironment of immune distress cues. However, rather than mobilizing to fuel new tissue regeneration, these ectopically proliferative stem cells remain within their niche to contain the breach. Together, our findings expose a potential communication relay system that operates from the niche to the stem cells to the immune system and back. The repurposing of proliferation by these stem cells patch the breached barrier, stoke the immune response and restore niche integrity.


Asunto(s)
Proliferación Celular/genética , Perfilación de la Expresión Génica/métodos , Folículo Piloso/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Animales , Comunicación Celular/genética , Ciclo Celular/genética , Células Cultivadas , Folículo Piloso/citología , Folículo Piloso/ultraestructura , Homeostasis/genética , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Células Madre/citología , Células Madre/ultraestructura
17.
Cell Stem Cell ; 23(4): 572-585.e7, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30174297

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) tightly couple maintenance of the bone marrow (BM) reservoir, including undifferentiated long-term repopulating hematopoietic stem cells (LT-HSCs), with intensive daily production of mature leukocytes and blood replenishment. We found two daily peaks of BM HSPC activity that are initiated by onset of light and darkness providing this coupling. Both peaks follow transient elevation of BM norepinephrine and TNF secretion, which temporarily increase HSPC reactive oxygen species (ROS) levels. Light-induced norepinephrine and TNF secretion augments HSPC differentiation and increases vascular permeability to replenish the blood. In contrast, darkness-induced TNF increases melatonin secretion to drive renewal of HSPCs and LT-HSC potential through modulating surface CD150 and c-Kit expression, increasing COX-2/αSMA+ macrophages, diminishing vascular permeability, and reducing HSPC ROS levels. These findings reveal that light- and darkness-induced daily bursts of norepinephrine, TNF, and melatonin within the BM are essential for synchronized mature blood cell production and HSPC pool repopulation.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Oscuridad , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Luz , Animales , Células Cultivadas , Epigénesis Genética/genética , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Cell Rep ; 23(7): 1962-1976, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768197

RESUMEN

Nitric oxide (NO) plays an established role in numerous physiological and pathological processes, but the specific cellular sources of NO in disease pathogenesis remain unclear, preventing the implementation of NO-related therapy. Argininosuccinate lyase (ASL) is the only enzyme able to produce arginine, the substrate for NO generation by nitric oxide synthase (NOS) isoforms. Here, we generated cell-specific conditional ASL knockout mice in combination with genetic and chemical colitis models. We demonstrate that NO derived from enterocytes alleviates colitis by decreasing macrophage infiltration and tissue damage, whereas immune cell-derived NO is associated with macrophage activation, resulting in increased severity of inflammation. We find that induction of endogenous NO production by enterocytes with supplements that upregulate ASL expression and complement its substrates results in improved epithelial integrity and alleviation of colitis and of inflammation-associated colon cancer.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Enterocitos/metabolismo , Enterocitos/patología , Inflamación/patología , Óxido Nítrico/metabolismo , Animales , Arginina/biosíntesis , Argininosuccinatoliasa/metabolismo , Células Epiteliales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Bio Protoc ; 7(4): e2134, 2017 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-34458455

RESUMEN

Hematopoietic stem cells (HSCs) are defined by their functional ability to self-renew and to differentiate into all blood cell lineages. The majority of HSC reside in specific anatomical locations in the bone marrow (BM) microenvironment, in a quiescent non motile mode. Adhesion interactions between HSCs and their supporting BM microenvironment cells are critical for maintaining stem cell quiescence and protection from DNA damaging agents to prevent hematology failure and death. Multiple signaling proteins play a role in controlling retention and migration of bone marrow HSCs. Adhesion molecules are involved in both processes regulating hematopoiesis and stem- and progenitor-cell BM retention, migration and development. The mechanisms underlying the movement of stem cells from and to the marrow have not been completely elucidated and are still an object of intense study. One important aspect is the modification of expression and affinity of adhesion molecules by stem and progenitor cells which are required both for stem cell retention, migration and development. Adhesion is regulated by expression of the adhesion molecules, their affinity and avidity. Affinity regulation is related to the molecular binding recognition and bond strength. Here, we describe the in vitro FACS assay used in our research to explore the expression, affinity and function of the integrin α4ß1 (also termed VLA-4) for murine bone marrow retained EPCR+ long term repopulation HSC (LT-HSC) (Gur- Cohen et al., 2015 ).

20.
Bio Protoc ; 7(4): e2135, 2017 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-34458456

RESUMEN

Hematopoietic stem cells (HSCs) are defined by their functional abilities to self-renew and to give rise to all mature blood and immune cell types throughout life. Most HSCs are retained in a non-motile quiescent state within a specialized protective microenvironment in the bone marrow (BM) termed the niche. HSCs are typically distinguished from other adult stem cells by their motility capacity. Movement of HSCs across the physical barrier of the marrow extracellular matrix and blood vessel endothelial cells is facilitated by suppression of adhesion interactions, which are essential to preserve the stem cells retained within their BM niches. Importantly, homing of HSCs to the BM following clinical transplantation is a crucial first step for the repopulation of ablated BM as in the case of curative treatment strategies for hematologic malignancies. The homing process ends with selective access and anchorage of HSCs to their specialized niches within the BM. Adhesion molecules are targets to either enhance homing in cases of stem cell transplantation or reduce BM retention to harvest mobilized HSCs from the blood of matched donors. A major adhesion protein which is functionally expressed on HSCs and is involved in their homing and retention is the integrin alpha4beta1 (Very late antigen-4; VLA4). In this protocol we introduce an adhesion assay optimized for VLA4 expressing murine bone marrow stem cells. This assay quantifies adherent HSCs by flow cytometry with HSC enriching cell surface markers subsequent to the isolation of VLA4 expressing adherent cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA