Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-36993731

RESUMO

Cell-to-cell signalling between niche and stem cells regulates tissue regeneration. While the identity of many mediating factors is known, it is largely unknown whether stem cells optimize their receptiveness to niche signals according to the niche organization. Here, we show that Lgr5+ small intestinal stem cells (ISCs) regulate the morphology and orientation of their secretory apparatus to match the niche architecture, and to increase transport efficiency of niche signal receptors. Unlike the progenitor cells lacking lateral niche contacts, ISCs orient Golgi apparatus laterally towards Paneth cells of the epithelial niche, and divide Golgi into multiple stacks reflecting the number of Paneth cell contacts. Stem cells with a higher number of lateral Golgi transported Epidermal growth factor receptor (Egfr) with a higher efficiency than cells with one Golgi. The lateral Golgi orientation and enhanced Egfr transport required A-kinase anchor protein 9 (Akap9), and was necessary for normal regenerative capacity in vitro . Moreover, reduced Akap9 in aged ISCs renders ISCs insensitive to niche-dependent modulation of Golgi stack number and transport efficiency. Our results reveal stem cell-specific Golgi complex configuration that facilitates efficient niche signal reception and tissue regeneration, which is compromised in the aged epithelium.

2.
Sci Adv ; 8(41): eabm1847, 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36240269

RESUMO

Niche-derived factors regulate tissue stem cells, but apart from the mechanosensory pathways, the effect of niche geometry is not well understood. We used organoids and bioengineered tissue culture platforms to demonstrate that the conical shape of Lgr5+ small intestinal stem cells (ISCs) facilitate their self-renewal and function. Inhibition of non-muscle myosin II (NM II)-driven apical constriction altered ISC shape and reduced niche curvature and stem cell capacity. Niche curvature is decreased in aged mice, suggesting that suboptimal interactions between old ISCs and their niche develop with age. We show that activation of NM IIC or physical restriction to young topology improves in vitro regeneration by old epithelium. We propose that the increase in lateral surface area of ISCs induced by apical constriction promotes interactions between neighboring cells, and the curved topology of the intestinal niche has evolved to maximize signaling between ISCs and neighboring cells.

3.
Hum Mol Genet ; 30(24): 2429-2440, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34274970

RESUMO

Many hereditary cancer syndromes are associated with an increased risk of small and large intestinal adenocarcinomas. However, conditions bearing a high risk to both adenocarcinomas and neuroendocrine tumors are yet to be described. We studied a family with 16 individuals in four generations affected by a wide spectrum of intestinal tumors, including hyperplastic polyps, adenomas, small intestinal neuroendocrine tumors, and colorectal and small intestinal adenocarcinomas. To assess the genetic susceptibility and understand the novel phenotype, we utilized multiple molecular methods, including whole genome sequencing, RNA sequencing, single cell sequencing, RNA in situ hybridization and organoid culture. We detected a heterozygous deletion at the cystic fibrosis locus (7q31.2) perfectly segregating with the intestinal tumor predisposition in the family. The deletion removes a topologically associating domain border between CFTR and WNT2, aberrantly activating WNT2 in the intestinal epithelium. These consequences suggest that the deletion predisposes to small intestinal neuroendocrine tumors and small and large intestinal adenocarcinomas, and reveals the broad tumorigenic effects of aberrant WNT activation in the human intestine.


Assuntos
Adenocarcinoma , Adenoma , Neoplasias Colorretais , Tumores Neuroendócrinos , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenoma/genética , Adenoma/patologia , Neoplasias Colorretais/genética , Humanos , Mucosa Intestinal/patologia , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Proteína Wnt2
4.
Development ; 148(12)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34128985

RESUMO

Epithelial attachment to the basement membrane (BM) is essential for mammary gland development, yet the exact roles of specific BM components remain unclear. Here, we show that Laminin α5 (Lama5) expression specifically in the luminal epithelial cells is necessary for normal mammary gland growth during puberty, and for alveologenesis during pregnancy. Lama5 loss in the keratin 8-expressing cells results in reduced frequency and differentiation of hormone receptor expressing (HR+) luminal cells. Consequently, Wnt4-mediated crosstalk between HR+ luminal cells and basal epithelial cells is compromised during gland remodeling, and results in defective epithelial growth. The effects of Lama5 deletion on gland growth and branching can be rescued by Wnt4 supplementation in the in vitro model of branching morphogenesis. Our results reveal a surprising role for BM-protein expression in the luminal mammary epithelial cells, and highlight the function of Lama5 in mammary gland remodeling and luminal differentiation.


Assuntos
Diferenciação Celular/genética , Epitélio/metabolismo , Laminina/genética , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Proteína Wnt4/genética , Animais , Biomarcadores , Células Epiteliais , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Laminina/metabolismo , Glândulas Mamárias Animais/embriologia , Camundongos , Modelos Biológicos , Morfogênese/genética , Organogênese/genética , Proteína Wnt4/metabolismo
5.
Nature ; 594(7863): 430-435, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34079124

RESUMO

The tumour suppressor APC is the most commonly mutated gene in colorectal cancer. Loss of Apc in intestinal stem cells drives the formation of adenomas in mice via increased WNT signalling1, but reduced secretion of WNT ligands increases the ability of Apc-mutant intestinal stem cells to colonize a crypt (known as fixation)2. Here we investigated how Apc-mutant cells gain a clonal advantage over wild-type counterparts to achieve fixation. We found that Apc-mutant cells are enriched for transcripts that encode several secreted WNT antagonists, with Notum being the most highly expressed. Conditioned medium from Apc-mutant cells suppressed the growth of wild-type organoids in a NOTUM-dependent manner. Furthermore, NOTUM-secreting Apc-mutant clones actively inhibited the proliferation of surrounding wild-type crypt cells and drove their differentiation, thereby outcompeting crypt cells from the niche. Genetic or pharmacological inhibition of NOTUM abrogated the ability of Apc-mutant cells to expand and form intestinal adenomas. We identify NOTUM as a key mediator during the early stages of mutation fixation that can be targeted to restore wild-type cell competitiveness and provide preventative strategies for people at a high risk of developing colorectal cancer.


Assuntos
Competição entre as Células , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Esterases/metabolismo , Genes APC , Mutação , Adenoma/genética , Adenoma/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Competição entre as Células/genética , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Meios de Cultivo Condicionados , Progressão da Doença , Esterases/antagonistas & inibidores , Esterases/genética , Feminino , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Organoides/citologia , Organoides/metabolismo , Organoides/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
6.
Mech Ageing Dev ; 191: 111330, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32805262

RESUMO

Like most tissues, intestine shows multiple alterations during aging. While the main function of nutrient absorption is relatively well maintained, capacity of the intestine to respond to abrupt changes or damage declines with age. The reduction in renewal and regeneration capacity results from alterations in the stem cells that renew the epithelium, and in the complex interactions stem cells have with their microenvironment, or the Niche. This review highlights recent evidence on age-associated changes in the intestinal stem cell function, and focuses on stem cell extrinsic mechanisms. Strategies targeting niche interactions have already shown promise in alleviating problems associated with intestinal aging in animal models, and may provide means to protect the elderly for example from chemotherapy induced gastrointestinal side-effects.


Assuntos
Envelhecimento/metabolismo , Mucosa Intestinal/metabolismo , Nicho de Células-Tronco , Células-Tronco/metabolismo , Animais , Humanos
7.
Gastroenterology ; 158(5): 1389-1401.e10, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31930988

RESUMO

BACKGROUND & AIMS: In addition to the Notch and Wnt signaling pathways, energy metabolism also regulates intestinal stem cell (ISC) function. Tumor suppressor and kinase STK11 (also called LKB1) regulates stem cells and cell metabolism. We investigated whether loss of LKB1 alters ISC homeostasis in mice. METHODS: We deleted LKB1 from ISCs in mice using Lgr5-regulated CRE-ERT2 (Lkb1Lgr5-KO mice) and the traced lineages by using a CRE-dependent TdTomato reporter. Intestinal tissues were collected and analyzed by immunohistochemical and immunofluorescence analyses. We purified ISCs and intestinal progenitors using flow cytometry and performed RNA-sequencing analysis. We measured organoid-forming capacity and ISC percentages using intestinal tissues from Lkb1Lgr5-KO mice. We analyzed human Ls174t cells with knockdown of LKB1 or other proteins by immunoblotting, real-time quantitative polymerase chain reaction, and the Seahorse live-cell metabolic assay. RESULTS: Some intestinal crypts from Lkb1Lgr5-KO mice lost ISCs compared with crypts from control mice. However, most crypts from Lkb1Lgr5-KO mice contained functional ISCs that expressed increased levels of Atoh1 messenger RNA (mRNA), acquired a gene expression signature associated with secretory cells, and generated more cells in the secretory lineage compared with control mice. Knockdown of LKB1 in Ls174t cells induced expression of Atoh1 mRNA and a phenotype of increased mucin production; knockdown of ATOH1 prevented induction of this phenotype. The increased expression of Atoh1 mRNA after LKB1 loss from ISCs or Ls174t cells did not involve Notch or Wnt signaling. Knockdown of pyruvate dehydrogenase kinase 4 (PDK4) or inhibition with dichloroacetate reduced the up-regulation of Atoh1 mRNA after LKB1 knockdown in Ls174t cells. Cells with LKB1 knockdown had a reduced rate of oxygen consumption, which was partially restored by PDK4 inhibition with dichloroacetate. ISCs with knockout of LKB1 increased the expression of PDK4 and had an altered metabolic profile. CONCLUSIONS: LKB1 represses transcription of ATOH1, via PDK4, in ISCs, restricting their differentiation into secretory lineages. These findings provide a connection between metabolism and the fate determination of ISCs.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Metabolismo Energético/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Células-Tronco/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Ácido Dicloroacético/farmacologia , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Camundongos , Camundongos Knockout , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil/antagonistas & inibidores , Piruvato Desidrogenase Quinase de Transferência de Acetil/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , RNA-Seq , Transcrição Gênica , Regulação para Cima/efeitos dos fármacos
8.
Nature ; 571(7765): 398-402, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31292548

RESUMO

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Assuntos
Envelhecimento , Senescência Celular , Esterases/metabolismo , Mucosa Intestinal/patologia , Celulas de Paneth/metabolismo , Regeneração , Envelhecimento/fisiologia , Animais , Senescência Celular/fisiologia , Esterases/antagonistas & inibidores , Esterases/biossíntese , Feminino , Humanos , Mucosa Intestinal/fisiologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , PPAR alfa/metabolismo , Celulas de Paneth/patologia , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células-Tronco , Células-Tronco/patologia , Proteínas Wnt/antagonistas & inibidores , Via de Sinalização Wnt
9.
J Clin Invest ; 128(1): 402-414, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202476

RESUMO

Germline mutations in the gene encoding tumor suppressor kinase LKB1 lead to gastrointestinal tumorigenesis in Peutz-Jeghers syndrome (PJS) patients and mouse models; however, the cell types and signaling pathways underlying tumor formation are unknown. Here, we demonstrated that mesenchymal progenitor- or stromal fibroblast-specific deletion of Lkb1 results in fully penetrant polyposis in mice. Lineage tracing and immunohistochemical analyses revealed clonal expansion of Lkb1-deficient myofibroblast-like cell foci in the tumor stroma. Loss of Lkb1 in stromal cells was associated with induction of an inflammatory program including IL-11 production and activation of the JAK/STAT3 pathway in tumor epithelia concomitant with proliferation. Importantly, treatment of LKB1-defcient mice with the JAK1/2 inhibitor ruxolitinib dramatically decreased polyposis. These data indicate that IL-11-mediated induction of JAK/STAT3 is critical in gastrointestinal tumorigenesis following Lkb1 mutations and suggest that targeting this pathway has therapeutic potential in Peutz-Jeghers syndrome.


Assuntos
Transformação Celular Neoplásica , Interleucina-11/metabolismo , Neoplasias Intestinais/metabolismo , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Interleucina-11/genética , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Janus Quinase 1/genética , Janus Quinase 2/genética , Camundongos , Camundongos Knockout , Mutação , Proteínas de Neoplasias/genética , Fator de Transcrição STAT3/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
10.
Science ; 348(6232): 340-3, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25837514

RESUMO

By dividing asymmetrically, stem cells can generate two daughter cells with distinct fates. However, evidence is limited in mammalian systems for the selective apportioning of subcellular contents between daughters. We followed the fates of old and young organelles during the division of human mammary stemlike cells and found that such cells apportion aged mitochondria asymmetrically between daughter cells. Daughter cells that received fewer old mitochondria maintained stem cell traits. Inhibition of mitochondrial fission disrupted both the age-dependent subcellular localization and segregation of mitochondria and caused loss of stem cell properties in the progeny cells. Hence, mechanisms exist for mammalian stemlike cells to asymmetrically sort aged and young mitochondria, and these are important for maintaining stemness properties.


Assuntos
Divisão Celular/fisiologia , Senescência Celular/fisiologia , Mitocôndrias/fisiologia , Células-Tronco/fisiologia , Células-Tronco/ultraestrutura , Divisão Celular/genética , Linhagem Celular , Senescência Celular/genética , Humanos , Mitocôndrias/ultraestrutura
11.
Curr Biol ; 24(21): 2533-40, 2014 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-25308079

RESUMO

Lamellipodia are dynamic actin-rich cellular extensions that drive advancement of the leading edge during cell migration. Lamellipodia undergo periodic extension and retraction cycles, but the molecular mechanisms underlying these dynamics and their role in cell migration have remained obscure. We show that glia-maturation factor (GMF), which is an Arp2/3 complex inhibitor and actin filament debranching factor, regulates lamellipodial protrusion dynamics in living cells. In cultured S2R(+) cells, GMF silencing resulted in an increase in the width of lamellipodial actin filament arrays. Importantly, live-cell imaging of mutant Drosophila egg chambers revealed that the dynamics of actin-rich protrusions in migrating border cells is diminished in the absence of GMF. Consequently, velocity of border cell clusters undergoing guided migration was reduced in GMF mutant flies. Furthermore, genetic studies demonstrated that GMF cooperates with the Drosophila homolog of Aip1 (flare) in promoting disassembly of Arp2/3-nucleated actin filament networks and driving border cell migration. These data suggest that GMF functions in vivo to promote the disassembly of Arp2/3-nucleated actin filament arrays, making an important contribution to cell migration within a 3D tissue environment.


Assuntos
Movimento Celular/fisiologia , Drosophila/metabolismo , Fator de Maturação da Glia/fisiologia , Pseudópodes/fisiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Drosophila/citologia , Drosophila/ultraestrutura , Proteínas de Drosophila/metabolismo , Fator de Maturação da Glia/genética , Fator de Maturação da Glia/metabolismo , Proteínas dos Microfilamentos/metabolismo , Pseudópodes/ultraestrutura
12.
Duodecim ; 130(19): 1965-72, 2014.
Artigo em Finlandês | MEDLINE | ID: mdl-25558617

RESUMO

The regeneration of human tissues requires actively dividing tissue-specific stem cells. These cells are maintained and functionally regulated by a specific microenvironment, the stem cell niche. The niche provides protection and produces signals that guide stem cell division and differentiation. As a consequence, the niche plays a significant role in maintaining the tissue throughout life, and thus impaired tissue regeneration associated with aging may be partly due to the non-functional stem cell niche.


Assuntos
Senescência Celular/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Humanos
13.
PLoS Biol ; 10(10): e1001407, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23091420

RESUMO

In adults, the growth of blood vessels, a process known as angiogenesis, is essential for organ growth and repair. In many disorders including cancer, angiogenesis becomes excessive. The cellular origin of new vascular endothelial cells (ECs) during blood vessel growth in angiogenic situations has remained unknown. Here, we provide evidence for adult vascular endothelial stem cells (VESCs) that reside in the blood vessel wall endothelium. VESCs constitute a small subpopulation within CD117+ (c-kit+) ECs capable of undergoing clonal expansion while other ECs have a very limited proliferative capacity. Isolated VESCs can produce tens of millions of endothelial daughter cells in vitro. A single transplanted c-kit-expressing VESC by the phenotype lin-CD31+CD105+Sca1+CD117+ can generate in vivo functional blood vessels that connect to host circulation. VESCs also have long-term self-renewal capacity, a defining functional property of adult stem cells. To provide functional verification on the role of c-kit in VESCs, we show that a genetic deficit in endothelial c-kit expression markedly decreases total colony-forming VESCs. In vivo, c-kit expression deficit resulted in impaired EC proliferation and angiogenesis and retardation of tumor growth. Isolated VESCs could be used in cell-based therapies for cardiovascular repair to restore tissue vascularization after ischemic events. VESCs also provide a novel cellular target to block pathological angiogenesis and cancer growth.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Células Endoteliais/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/metabolismo
14.
Angiogenesis ; 15(3): 511-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22581517

RESUMO

The role of Transforming growth factor ß (TGF-ß) as a regulator of blood vessel endothelium is complicated and controversial, and the mechanisms by which TGF-ß is able to induce angiogenesis in vivo are not well understood. Here we show that TGF-ß causes in vivo a massive recruitment of tissue infiltrating hematopoietic cells. Concurrently, TGF-ß induces strong vascular endothelial growth factor (VEGF) production in the recruited hematopoietic cells, resulting in activated angiogenesis and vascular remodeling. TGF-ß also promoted abnormalities of α-smooth muscle actin-expressing pericytes on angiogenic capillaries. TGF-ß-induced angiogenic effect was inhibited by a systemic treatment with VEGF-neutralizing antibodies. When studied in isolated human hematopoietic cells, physiological concentrations of TGF-ß stimulated VEGF mRNA and protein expression in a dose- and time-dependent manner. This induction was p38 and p44/p42 mitogen activated kinase dependent. p38 and p44/p42 activation was also observed in vivo in TGF-ß-treated angiogenic murine tissues. Taken together, our results provide a dual action mechanism by which TGF-ß promotes angiogenesis in vivo via recruitment of paracrine VEGF-expressing hematopoietic effector cells. This mechanism may activate vascular growth and remodeling during inflammatory conditions and tumor growth when TGF-ß activity is upregulated.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Células da Medula Óssea/metabolismo , Neovascularização Fisiológica/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...