Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Trends Immunol ; 40(11): 1053-1070, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31645297

RESUMEN

Hematopoietic stem cells (HSCs) self-renew or differentiate into blood cell lineages following extrinsic cues propagated in specialized niches. Support cells and soluble factors in the niche respond to stress and enable progenitor activity. Metalloproteases (MMPs, ADAMs, ADAMTSs) and their inhibitors (TIMPs) control certain physical and biochemical features of the niche by altering protease-dependent bioavailability of local niche factors (e.g., CXCL12, SCF, TGFß, VEGF), matrix turnover, and cellular interactions. With over 40 examples of diverse metalloprotease substrates known to trigger fate-changing decisions, the spatially confined activity of this multi-member protease family is ideally positioned to constitute a higher order control over hematopoiesis. Comprehension of regulated proteolysis in the bone marrow may fuel innovative strategies to harness HSC fate and function.


Asunto(s)
Matriz Extracelular/metabolismo , Células Madre Hematopoyéticas/fisiología , Metaloproteasas/metabolismo , Animales , Diferenciación Celular , Autorrenovación de las Células , Hematopoyesis , Humanos , Proteolisis , Nicho de Células Madre
2.
Nature ; 465(7299): 803-7, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20445538

RESUMEN

Reproductive history is the strongest risk factor for breast cancer after age, genetics and breast density. Increased breast cancer risk is entwined with a greater number of ovarian hormone-dependent reproductive cycles, yet the basis for this predisposition is unknown. Mammary stem cells (MaSCs) are located within a specialized niche in the basal epithelial compartment that is under local and systemic regulation. The emerging role of MaSCs in cancer initiation warrants the study of ovarian hormones in MaSC homeostasis. Here we show that the MaSC pool increases 14-fold during maximal progesterone levels at the luteal dioestrus phase of the mouse. Stem-cell-enriched CD49fhi cells amplify at dioestrus, or with exogenous progesterone, demonstrating a key role for progesterone in propelling this expansion. In aged mice, CD49fhi cells display stasis upon cessation of the reproductive cycle. Progesterone drives a series of events where luminal cells probably provide Wnt4 and RANKL signals to basal cells which in turn respond by upregulating their cognate receptors, transcriptional targets and cell cycle markers. Our findings uncover a dynamic role for progesterone in activating adult MaSCs within the mammary stem cell niche during the reproductive cycle, where MaSCs are putative targets for cell transformation events leading to breast cancer.


Asunto(s)
Envejecimiento/fisiología , Glándulas Mamarias Animales/citología , Progesterona/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Animales , Recuento de Células , División Celular/efectos de los fármacos , Transformación Celular Neoplásica , Estrógenos/farmacología , Ciclo Estral/sangre , Ciclo Estral/fisiología , Femenino , Homeostasis/efectos de los fármacos , Integrina alfa6/metabolismo , Ratones , Ovariectomía , Comunicación Paracrina/efectos de los fármacos , Progesterona/sangre , Progesterona/metabolismo , Ligando RANK/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/metabolismo , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt4
3.
Cell Rep Methods ; 4(4): 100741, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38569541

RESUMEN

Deep proteomic profiling of rare cell populations has been constrained by sample input requirements. Here, we present DROPPS (droplet-based one-pot preparation for proteomic samples), an accessible low-input platform that generates high-fidelity proteomic profiles of 100-2,500 cells. By applying DROPPS within the mammary epithelium, we elucidated the connection between mitochondrial activity and clonogenicity, identifying CD36 as a marker of progenitor capacity in the basal cell compartment. We anticipate that DROPPS will accelerate biology-driven proteomic research for a multitude of rare cell populations.


Asunto(s)
Biomarcadores , Antígenos CD36 , Glándulas Mamarias Animales , Proteómica , Células Madre , Proteómica/métodos , Antígenos CD36/metabolismo , Animales , Femenino , Células Madre/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Biomarcadores/metabolismo , Biomarcadores/análisis , Epitelio/metabolismo , Ratones , Humanos , Mitocondrias/metabolismo
4.
Cell Rep ; 42(10): 113256, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37847590

RESUMEN

It is widely assumed that all normal somatic cells can equally perform homologous recombination (HR) and non-homologous end joining in the DNA damage response (DDR). Here, we show that the DDR in normal mammary gland inherently depends on the epithelial cell lineage identity. Bioinformatics, post-irradiation DNA damage repair kinetics, and clonogenic assays demonstrated luminal lineage exhibiting a more pronounced DDR and HR repair compared to the basal lineage. Consequently, basal progenitors were far more sensitive to poly(ADP-ribose) polymerase inhibitors (PARPis) in both mouse and human mammary epithelium. Furthermore, PARPi sensitivity of murine and human breast cancer cell lines as well as patient-derived xenografts correlated with their molecular resemblance to the mammary progenitor lineages. Thus, mammary epithelial cells are intrinsically divergent in their DNA damage repair capacity and PARPi vulnerability, potentially influencing the clinical utility of this targeted therapy.


Asunto(s)
Antineoplásicos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Animales , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Antineoplásicos/farmacología , Reparación del ADN , Recombinación Homóloga , Daño del ADN
5.
Blood Adv ; 5(20): 3960-3974, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34500457

RESUMEN

Bone marrow (BM) is the primary site of hematopoiesis and is responsible for a lifelong supply of all blood cell lineages. The process of hematopoiesis follows key intrinsic programs that also integrate instructive signals from the BM niche. First identified as an erythropoietin-potentiating factor, the tissue inhibitor of metalloproteinase (TIMP) protein family has expanded to 4 members and has widely come to be viewed as a classical regulator of tissue homeostasis. By virtue of metalloprotease inhibition, TIMPs not only regulate extracellular matrix turnover but also control growth factor bioavailability. The 4 mammalian TIMPs possess overlapping enzyme-inhibition profiles and have never been studied for their cumulative role in hematopoiesis. Here, we show that TIMPs are critical for postnatal B lymphopoiesis in the BM. TIMP-deficient mice have defective B-cell development arising at the pro-B-cell stage. Expression analysis of TIMPless hematopoietic cell subsets pointed to an altered B-cell program in the Lineage-Sca-1+c-Kit+ (LSK) cell fraction. Serial and competitive BM transplants identified a defect in TIMP-deficient hematopoietic stem and progenitor cells for B lymphopoiesis. In parallel, reverse BM transplants uncovered the extrinsic role of stromal TIMPs in pro- and pre-B-cell development. TIMP deficiency disrupted CXCL12 localization to LepR+ cells, and increased soluble CXCL12 within the BM niche. It also compromised the number and morphology of LepR+ cells. These data provide new evidence that TIMPs control the cellular and biochemical makeup of the BM niche and influence the LSK transcriptional program required for optimal B lymphopoiesis.


Asunto(s)
Células de la Médula Ósea , Médula Ósea , Animales , Linfocitos B , Hematopoyesis , Ratones , Inhibidores Tisulares de Metaloproteinasas/genética
7.
Nat Commun ; 10(1): 1760, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988300

RESUMEN

The mammary gland experiences substantial remodeling and regeneration during development and reproductive life, facilitated by stem cells and progenitors that act in concert with physiological stimuli. While studies have focused on deciphering regenerative cells within the parenchymal epithelium, cell lineages in the stroma that may directly contribute to epithelial biology is unknown. Here we identify, in mouse, the transition of a PDGFRα+ mesenchymal cell population into mammary epithelial progenitors. In addition to being adipocyte progenitors, PDGFRα+ cells make a de novo contribution to luminal and basal epithelia during mammary morphogenesis. In the adult, this mesenchymal lineage primarily generates luminal progenitors within lobuloalveoli during sex hormone exposure or pregnancy. We identify cell migration as a key molecular event that is activated in mesenchymal progenitors in response to epithelium-derived chemoattractant. These findings demonstrate a stromal reservoir of epithelial progenitors and provide insight into cell origins and plasticity during mammary tissue growth.


Asunto(s)
Adipocitos/citología , Glándulas Mamarias Animales/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/citología , Animales , Diferenciación Celular , Linaje de la Célula , Células Epiteliales , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones
8.
Commun Biol ; 2: 192, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31123716

RESUMEN

The heterogeneity of breast cancer makes current therapies challenging. Metformin, the anti-diabetic drug, has shown promising anti-cancer activities in epidemiological studies and breast cancer models. Yet, how metformin alters the normal adult breast tissue remains elusive. We demonstrate metformin intake at a clinically relevant dose impacts the hormone receptor positive (HR+) luminal cells in the normal murine mammary gland. Metformin decreases total cell number, progenitor capacity and specifically reduces DNA damage in normal HR+ luminal cells, decreases oxygen consumption rate and increases cell cycle length of luminal cells. HR+ luminal cells demonstrate the lowest levels of mitochondrial respiration and capacity to handle oxidative stress compared to the other fractions, suggesting their intrinsic susceptibility to long-term metformin exposure. Uncovering HR+ luminal cells in the normal mammary gland as the major cell target of metformin exposure could identify patients that would most benefit from repurposing this anti-diabetic drug for cancer prevention/therapy purposes.


Asunto(s)
Hipoglucemiantes/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Metformina/farmacología , Animales , Apoptosis , Ciclo Celular , Linaje de la Célula , Separación Celular , Daño del ADN , Femenino , Citometría de Flujo , Ratones , Receptores de Estrógenos/metabolismo
9.
J Cell Biol ; 218(9): 3134-3152, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31371388

RESUMEN

Regulated growth plate activity is essential for postnatal bone development and body stature, yet the systems regulating epiphyseal fusion are poorly understood. Here, we show that the tissue inhibitors of metalloprotease (TIMP) gene family is essential for normal bone growth after birth. Whole-body quadruple-knockout mice lacking all four TIMPs have growth plate closure in long bones, precipitating limb shortening, epiphyseal distortion, and widespread chondrodysplasia. We identify TIMP/FGF-2/IHH as a novel nexus underlying bone lengthening where TIMPs negatively regulate the release of FGF-2 from chondrocytes to allow IHH expression. Using a knock-in approach that combines MMP-resistant or ADAMTS-resistant aggrecans with TIMP deficiency, we uncouple growth plate activity in axial and appendicular bones. Thus, natural metalloprotease inhibitors are crucial regulators of chondrocyte maturation program, growth plate integrity, and skeletal proportionality. Furthermore, individual and combinatorial TIMP-deficient mice demonstrate the redundancy of metalloprotease inhibitor function in embryonic and postnatal development.


Asunto(s)
Desarrollo Óseo , Huesos/metabolismo , Condrocitos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Placa de Crecimiento/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Animales , Factor 2 de Crecimiento de Fibroblastos/genética , Ratones , Ratones Noqueados , Inhibidores Tisulares de Metaloproteinasas/genética
10.
Commun Biol ; 1: 111, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271991

RESUMEN

Breast cancer is the most common cancer in females. The number of years menstruating and length of an individual menstrual cycle have been implicated in increased breast cancer risk. At present, the proliferative changes within an individual reproductive cycle or variations in the estrous cycle in the normal mammary gland are poorly understood. Here we use Fucci2 reporter mice to demonstrate actively proliferating mammary epithelial cells have shorter G1 lengths, whereas more differentiated/non-proliferating cells have extended G1 lengths. We find that cells enter into the cell cycle mainly during diestrus, yet the expansion is erratic and does not take place every reproductive cycle. Single cell expression analyses feature expected proliferation markers (Birc5, Top2a), while HR+ luminal cells exhibit fluctuations of key differentiation genes (ER, Gata3) during the cell cycle. We highlight the proliferative heterogeneity occurring within the normal mammary gland during a single-estrous cycle, indicating that the mammary gland undergoes continual dynamic proliferative changes.

12.
Stem Cell Reports ; 5(1): 31-44, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26095608

RESUMEN

Systemic and local signals must be integrated by mammary stem and progenitor cells to regulate their cyclic growth and turnover in the adult gland. Here, we show RANK-positive luminal progenitors exhibiting WNT pathway activation are selectively expanded in the human breast during the progesterone-high menstrual phase. To investigate underlying mechanisms, we examined mouse models and found that loss of RANK prevents the proliferation of hormone receptor-negative luminal mammary progenitors and basal cells, an accompanying loss of WNT activation, and, hence, a suppression of lobuloalveologenesis. We also show that R-spondin1 is depleted in RANK-null progenitors, and that its exogenous administration rescues key aspects of RANK deficiency by reinstating a WNT response and mammary cell expansion. Our findings point to a novel role of RANK in dictating WNT responsiveness to mediate hormone-induced changes in the growth dynamics of adult mammary cells.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Células Madre/citología , Trombospondinas/genética , Animales , Proliferación Celular/genética , Femenino , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Trombospondinas/biosíntesis , Vía de Señalización Wnt/genética
13.
Stem Cell Reports ; 4(3): 313-322, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-28447939

RESUMEN

Progesterone drives mammary stem and progenitor cell dynamics through paracrine mechanisms that are currently not well understood. Here, we demonstrate that CXCR4, the receptor for stromal-derived factor 1 (SDF-1; CXC12), is a crucial instructor of hormone-induced mammary stem and progenitor cell function. Progesterone elicits specific changes in the transcriptome of basal and luminal mammary epithelial populations, where CXCL12 and CXCR4 represent a putative ligand-receptor pair. In situ, CXCL12 localizes to progesterone-receptor-positive luminal cells, whereas CXCR4 is induced in both basal and luminal compartments in a progesterone-dependent manner. Pharmacological inhibition of CXCR4 signaling abrogates progesterone-directed expansion of basal (CD24+CD49fhi) and luminal (CD24+CD49flo) subsets. This is accompanied by a marked reduction in CD49b+SCA-1- luminal progenitors, their functional capacity, and lobuloalveologenesis. These findings uncover CXCL12 and CXCR4 as novel paracrine effectors of hormone signaling in the adult mammary gland, and present a new avenue for potentially targeting progenitor cell growth and malignant transformation in breast cancer.

14.
Nat Cell Biol ; 16(9): 889-901, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25150980

RESUMEN

Cancer-associated fibroblasts (CAFs) drive tumour progression, but the emergence of this cell state is poorly understood. A broad spectrum of metalloproteinases, controlled by the Timp gene family, influence the tumour microenvironment in human cancers. Here, we generate quadruple TIMP knockout (TIMPless) fibroblasts to unleash metalloproteinase activity within the tumour-stromal compartment and show that complete Timp loss is sufficient for the acquisition of hallmark CAF functions. Exosomes produced by TIMPless fibroblasts induce cancer cell motility and cancer stem cell markers. The proteome of these exosomes is enriched in extracellular matrix proteins and the metalloproteinase ADAM10. Exosomal ADAM10 increases aldehyde dehydrogenase expression in breast cancer cells through Notch receptor activation and enhances motility through the GTPase RhoA. Moreover, ADAM10 knockdown in TIMPless fibroblasts abrogates their CAF function. Importantly, human CAFs secrete ADAM10-rich exosomes that promote cell motility and activate RhoA and Notch signalling in cancer cells. Thus, Timps suppress cancer stroma where activated-fibroblast-secreted exosomes impact tumour progression.


Asunto(s)
Fibroblastos/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Inhibidores Tisulares de Metaloproteinasas/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Exosomas/fisiología , Femenino , Fibroblastos/patología , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Mamarias Experimentales/enzimología , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Trasplante de Neoplasias , Fenotipo , Transducción de Señal , Inhibidores Tisulares de Metaloproteinasas/deficiencia , Carga Tumoral
15.
Nat Genet ; 46(9): 964-72, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25129143

RESUMEN

Creating spontaneous yet genetically tractable human tumors from normal cells presents a fundamental challenge. Here we combined retroviral and transposon insertional mutagenesis to enable cancer gene discovery starting with human primary cells. We used lentiviruses to seed gain- and loss-of-function gene disruption elements, which were further deployed by Sleeping Beauty transposons throughout the genome of human bone explant mesenchymal cells. De novo tumors generated rapidly in this context were high-grade myxofibrosarcomas. Tumor insertion sites were enriched in recurrent somatic copy-number aberration regions from multiple cancer types and could be used to pinpoint new driver genes that sustain somatic alterations in patients. We identified HDLBP, which encodes the RNA-binding protein vigilin, as a candidate tumor suppressor deleted at 2q37.3 in greater than one out of ten tumors across multiple tissues of origin. Hybrid viral-transposon systems may accelerate the functional annotation of cancer genomes by enabling insertional mutagenesis screens in higher eukaryotes that are not amenable to germline transgenesis.


Asunto(s)
Mutagénesis Insercional , Sarcoma/genética , Línea Celular , Elementos Transponibles de ADN , Vectores Genéticos/genética , Genoma Humano , Células HEK293 , Humanos , Proteínas de Unión al ARN/genética , Retroviridae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA