Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Biol Int ; 45(7): 1523-1532, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33724613

RESUMO

Radiotherapy is a leading treatment for various types of cancer. However, exposure to high-dose ionizing radiation causes acute gastrointestinal injury and gastrointestinal syndrome. This has significant implications for human health, and therefore, radioprotection is a major area of research. Radiation induces the loss of intestinal stem cells; hence, the protection of stem cells expressing LGR5 (a marker of intestinal epithelial stem cells) is a key strategy for the prevention of radiation-induced injury. In this study, we identified valproic acid (VPA) as a potent radioprotector using an intestinal organoid culture system. VPA treatment increased the number of LGR5+ stem cells and organoid regeneration after irradiation. N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT, an inhibitor of NOTCH signaling) blocked the radioprotective effects of VPA, indicating that NOTCH signaling is a likely mechanism underlying the observed effects of VPA. In addition, VPA acted as a radiosensitizer via the inhibition of histone deacetylase (HDAC) in a colorectal cancer organoid. These results demonstrate that VPA exerts strong protective effects on LGR5+ stem cells via NOTCH signaling and that the inhibition of NOTCH signaling reduces these protective effects, providing a basis for the improved management of radiation injury.


Assuntos
Neoplasias/radioterapia , Organoides/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Lesões por Radiação/tratamento farmacológico , Ácido Valproico/farmacologia , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Notch/metabolismo
2.
Adv Exp Med Biol ; 908: 11-25, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27573765

RESUMO

The field of gastrointestinal epithelial stem cells is a rapidly developing area of adult stem cell research. The discovery of Lgr5(+) intestinal stem cells has enabled us to study many hidden aspects of the biology of gastrointestinal adult stem cells. Marked by Lgr5 and Troy, several novel endodermal stem cells have been identified in the gastrointestinal tract. A precise working model of stem cell propagation, dynamics, and plasticity has been revealed by a genetic labeling method, termed lineage tracing. This chapter introduces the reidentification of crypt base columnar cells as Lgr5(+) stem cells in the intestine. Subsequently, it will discuss dynamic clonal evolution and cellular plasticity in the intestinal stem cell zone, as well as in stem cell zones of stomach glands.


Assuntos
Evolução Clonal , Trato Gastrointestinal/citologia , Mucosa Intestinal/citologia , Células-Tronco/citologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores Tumorais/genética , Linhagem da Célula/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Trato Gastrointestinal/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Células-Tronco/metabolismo
3.
Inflamm Regen ; 43(1): 14, 2023 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-36803580

RESUMO

BACKGROUND: The hair follicles (HFs) are barely regenerated after loss in injuries in mammals as well as in human beings. Recent studies have shown that the regenerative ability of HFs is age-related; however, the relationship between this phenomenon and the stem cell niche remains unclear. This study aimed to find a key secretory protein that promotes the HFs regeneration in the regenerative microenvironment. METHODS: To explore why age affects HFs de novo regeneration, we established an age-dependent HFs regeneration model in leucine-rich repeat G protein-coupled receptor 5 (Lgr5) + /mTmG mice. Proteins in tissue fluids were analyzed by high-throughput sequencing. The role and mechanism of candidate proteins in HFs de novo regeneration and hair follicle stem cells (HFSCs) activation were investigated through in vivo experiments. The effects of candidate proteins on skin cell populations were investigated by cellular experiments. RESULTS: Mice under 3-week-old (3W) could regenerate HFs and Lgr5 HFSCs, which were highly correlated with the immune cells, cytokines, IL-17 signaling pathway, and IL-1α level in the regeneration microenvironment. Additionally, IL-1α injection induced de novo regeneration of HFs and Lgr5 HFSCs in 3W mouse model with a 5 mm wound, as well as promoted activation and proliferation of Lgr5 HFSCs in 7-week-old (7W) mice without wound. Dexamethasone and TEMPOL inhibited the effects of IL-1α. Moreover, IL-1α increased skin thickness and promoted the proliferation of human epidermal keratinocyte line (HaCaT) and skin-derived precursors (SKPs) in vivo and in vitro, respectively. CONCLUSIONS: In conclusion, injury-induced IL-1α promotes HFs regeneration by modulating inflammatory cells and oxidative stress-induced Lgr5 HFSCs regeneration as well as promoting skin cell populations proliferation. This study uncovers the underlying molecular mechanisms enabling HFs de novo regeneration in an age-dependent model.

4.
Cell Mol Gastroenterol Hepatol ; 13(1): 275-287, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34438113

RESUMO

BACKGROUND & AIMS: Notch pathway signaling maintains gastric epithelial cell homeostasis by regulating stem cell proliferation and differentiation. We previously identified NOTCH1 and NOTCH2 as the key Notch receptors controlling gastric stem cell function. Here, we identify the niche cells and critical Notch ligand responsible for regulating stem cell proliferation in the distal mouse stomach. METHODS: Expression of Notch ligands in the gastric antrum was determined by quantitative reverse-transcriptase polymerase chain reaction and cellular localization was determined by in situ hybridization and immunostaining. The contribution of specific Notch ligands to regulate epithelial cell proliferation in adult mice was determined by inducible gene deletion, or by pharmacologic inhibition using antibodies directed against specific Notch ligands. Mouse gastric organoid cultures were used to confirm that Notch ligand signaling was epithelial specific. RESULTS: Delta-like 1 (DLL1) and Jagged 1 (JAG1) were the most abundantly expressed Notch ligands in the adult mouse stomach, with DLL1 restricted to the antral gland base and JAG1 localized to the upper gland region. Inhibition of DLL1 alone or in combination with other Notch ligands significantly reduced epithelial cell proliferation and the growth of gastric antral organoids, while inhibition of the other Notch ligands, DLL4, JAG1, and JAG2, did not affect proliferation or organoid growth. Similarly, DLL1, and not DLL4, regulated proliferation of LGR5+ antral stem cells, which express the NOTCH1 receptor. CONCLUSIONS: DLL1 is the key Notch ligand regulating epithelial cell proliferation in the gastric antrum. We propose that DLL1-expressing cells at the gland base are Notch niche cells that signal to adjacent LGR5+ antral stem cells to regulate stem cell proliferation and epithelial homeostasis.


Assuntos
Proteínas de Ligação ao Cálcio , Antro Pilórico , Células-Tronco , Animais , Proteínas de Ligação ao Cálcio/fisiologia , Proliferação de Células , Camundongos , Antro Pilórico/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Células-Tronco/metabolismo
5.
Int J Biol Sci ; 16(11): 1876-1887, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32398956

RESUMO

Ulcerative colitis (UC) is a recurrent intestinal inflammatory disease. Slit2, a secreted protein, interacts with its receptor Robo1 to regulate the differentiation of intestinal stem cells and participate in inflammation and tumor development. However, whether Slit2/Robo1involved in the pathogenesis of UC is not known. We investigated Slit2/Robo1-mediated UC using a dextran sodium sulfate (DSS)-induced model. Eight-week-old male Slit2-Tg (Slit2 transgene) mice, Robo1/2+/- (Robo1+/- Robo2+/-) mice, and their WT littermates were allocated into two groups: (I) control group (n=10), of mice fed a normal diet and tap water and (II) DSS group (n=10), of mice fed a normal diet and drinking water with 2% DSS for 7 days. Colon tissues were collected and analyzed by qPCR, immunohistochemistry, western blot, and immunofluorescence. Slit2-Tg DSS mice showed less body weight loss, less blood in the stool, and less viscous stool compared to those of WTSlit DSS mice. Robo1/2+/- DSS mice displayed a heavier degree of blood in the stool and a more apparent viscosity of the stool compared to those of WTRobo1/2 DSS mice. Slit2 overexpression maintained Lgr5+ stem cell proliferation in the crypt after DSS treatment, significantly increased the LC3II/I ratio, and slightly stimulated p62 expression in the crypt compared to those of DSS-induced WTSlit mice. Robo1/2 partial knockout reduced the number of Lgr5+ stem cells, decreased the LC3II/I ratio, and markedly increased p62 expression in the crypt compare to those of DSS-treated WTRobo1/2 mice. Our findings suggest that Slit2/Robo1 mediates DSS-induced UC probably by activating the autophagy of Lgr5+ stem cells.


Assuntos
Autofagia/fisiologia , Colite Ulcerativa/induzido quimicamente , Sulfato de Dextrana/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Células-Tronco/fisiologia , Animais , Colite Ulcerativa/metabolismo , Citocinas/genética , Citocinas/metabolismo , Bases de Dados Genéticas , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Intestinos/citologia , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Transdução de Sinais , Proteínas Roundabout
6.
Int J Biol Sci ; 15(1): 34-43, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662345

RESUMO

Lgr5-expressing stem cells contribute to the epithelial turnover of the gastric antrum. However, the mechanism controlling the homeostasis of Lgr5+ antral stem cells is not fully understood. Here, we demonstrate the key role of E-cadherin in the homeostasis of Lgr5+ gastric antral stem cells. The deletion of E-cadherin in these cells results in their apoptosis, thereby leading to a marked decrease in their number. A reduced Lgr5+ stem cell pool caused by the loss of E-cadherin impairs gastric antral epithelial homeostasis in vivo and organoid growth in vitro. Furthermore, p53 contributes to the apoptosis of Lgr5+ stem cells following E-cadherin loss, while the simultaneous deletion of p53 rescues the phenotype in E-cadherin mutants. Our study reveals the critical pro-survival function of E-cadherin in Lgr5+ gastric antral stem cells and the key role of the Lgr5+ stem cell pool in the maintenance of gastric epithelial homeostasis.


Assuntos
Caderinas/metabolismo , Antro Pilórico/citologia , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Caderinas/genética , Células Cultivadas , Homeostase/genética , Homeostase/fisiologia , Camundongos , Microscopia Confocal , Receptores Acoplados a Proteínas G/genética
7.
Methods Mol Biol ; 1576: 113-121, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29589266

RESUMO

The intestinal epithelium is one of the most rapidly self-renewing tissues throughout life in mammals. A small population of stem cells at the base of crypt in the epithelium can continually self-renew and give rise to differentiated epithelial cells. The self-renewal and differentiation of intestinal stem cells are under a tight control during homeostasis, and disruption of this balancing regulation leads to intestinal degeneration or tumorigenesis. Accordingly, exploration of the mechanism underlying the regulation of stem cells is essential for the understanding and treatment of intestinal disorders. As traditional methods using mice models are costly and time-consuming, the recently established ex vivo intestinal organoids model provides an ideal tool to investigate the mechanisms regulating the self-renewal and differentiation of intestinal stem cells. The intestinal organoids recapitulate major characteristics in both structure and function of intestinal epithelium in vivo. Here, we describe a new protocol to generate the intestinal organoids from both crypts and single stem cells with a higher efficiency using the small molecule blebbistatin and provide an approach to assess the self-renewal and differentiation of stem cells in intestinal organoids.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Intestinos/citologia , Organoides/citologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Células Cultivadas , Intestinos/efeitos dos fármacos , Camundongos , Organoides/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
8.
Biomedicines ; 7(2)2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31248166

RESUMO

The Wnt signaling pathway is one of the most prominent developmental signals. In addition to its functions in development, there is emerging evidence that it is also crucial for various organ functions in adult organisms, where Wnt signaling controls tissue stem cell behavior, proliferation and differentiation. Deregulation of Wnt signaling is involved in various pathological conditions and has been linked to malignant tissue transformation in different organ systems. The study of the Wnt signaling pathway has revealed a complex regulatory network that tightly balances the quality and strength of Wnt signaling in tissues. In this context, R-spondins are secreted proteins that stabilize Wnt receptors and enhance Wnt signaling. In this review we focus on new insights into the regulatory function of Wnt and R-spondin signaling in the stomach. In addition to its function in the healthy state, we highlight the connection between Wnt signaling and infection with Helicobacter pylori (H. pylori), a pathogen that colonizes the stomach and is the main risk factor for gastric cancer. In addition to experimental data that link Wnt signaling to carcinogenesis, we discuss that Wnt signaling is affected in a substantial proportion of patients with gastric cancer, and provide examples for potential clinical implications for altered Wnt signaling in gastric cancer.

9.
Cell Rep ; 25(3): 585-597.e7, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332640

RESUMO

Epithelial tissues, such as the skin, rely on cellular plasticity of stem cells (SCs) from different niches to restore tissue function after injury. How these molecularly and functionally diverse SC populations respond to injury remains elusive. Here, we genetically labeled Lgr5- or Lgr6-expressing cells from the hair follicle bulge and interfollicular epidermis (IFE), respectively, and monitored their individual transcriptional adaptations during wound healing using single-cell transcriptomics. Both Lgr5 and Lgr6 progeny rapidly induced a genetic wound signature that, for Lgr5 progeny, included the remodeling of receptors to permit interactions with the wound environment, a property that Lgr6 progeny possessed even before wounding. When contributing to re-epithelialization, Lgr5 progeny gradually replaced their bulge identity with an IFE identity, and this process started already before Lgr5 progeny left the bulge. Altogether, this study reveals how different SCs respond and adapt to a new environment, potentially explaining cellular plasticity of many epithelial tissues.


Assuntos
Epiderme/crescimento & desenvolvimento , Folículo Piloso/citologia , Análise de Célula Única/métodos , Pele/citologia , Células-Tronco/citologia , Transcriptoma , Cicatrização , Animais , Proliferação de Células , Células Cultivadas , Epiderme/lesões , Epiderme/metabolismo , Feminino , Folículo Piloso/lesões , Folículo Piloso/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reepitelização , Receptores Acoplados a Proteínas G/fisiologia , Pele/lesões , Pele/metabolismo , Células-Tronco/metabolismo
10.
Cell Rep ; 24(9): 2312-2328.e7, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30157426

RESUMO

IBD syndromes such as Crohn's disease and ulcerative colitis result from the inflammation of specific intestinal segments. Although many studies have reported on the regenerative response of intestinal progenitor and stem cells to tissue injury, very little is known about the response of differentiated lineages to inflammatory cues. Here, we show that acute inflammation of the mouse small intestine is followed by a dramatic loss of Lgr5+ stem cells. Instead, Paneth cells re-enter the cell cycle, lose their secretory expression signature, and acquire stem-like properties, thus contributing to the tissue regenerative response to inflammation. Stem cell factor secretion upon inflammation triggers signaling through the c-Kit receptor and a cascade of downstream events culminating in GSK3ß inhibition and Wnt activation in Paneth cells. Hence, the plasticity of the intestinal epithelium in response to inflammation goes well beyond stem and progenitor cells and extends to the fully differentiated and post-mitotic Paneth cells.


Assuntos
Inflamação/metabolismo , Intestino Delgado/fisiopatologia , Regeneração Nervosa/fisiologia , Celulas de Paneth/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Camundongos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais
11.
Environ Toxicol Pharmacol ; 43: 44-53, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26950395

RESUMO

The intestinal epithelium plays a central role in immune homeostasis in the intestine. AhR, a ligand-activated transcription factor, plays an important role in diverse physiological processes. The intestines are exposed to various exogenous and endogenous AhR ligands. Thus, AhR may regulate the intestinal homeostasis, directly acting on the development of intestinal epithelial cells (IEC). In this study, we demonstrated that 6-formylindolo[3,2-b]carbazole (FICZ) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibited the in vitro development of mouse intestinal organoids. The number of Paneth cells in the small intestine and the depth of crypts of the small and large intestines were reduced in mice administrated with FICZ. Immunohistochemical and flow cytometric assays revealed that AhR was highly expressed in Lgr5(+) stem cells. FICZ inhibited Wnt signaling lowering the level of ß-catenin protein. Gene expression analyses demonstrated that FICZ increased expression of Lgr5, Math1, BMP4, and Indian Hedgehog while inhibiting that of Lgr4.


Assuntos
Carbazóis/toxicidade , Células Epiteliais/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Células Epiteliais/fisiologia , Inibidores do Crescimento/toxicidade , Camundongos
12.
Cancer Lett ; 369(2): 336-43, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26455770

RESUMO

There is an urgent need to elucidate the mechanistic links between obesity and colon cancer. Convincing evidence for the role of Lgr5(+) stem cells in colon tumorigenesis has been established; however, the influence of obesity on stem cell maintenance is unknown. We assessed the effects of high fat (HF) feeding on colonic stem cell maintenance during cancer initiation (AOM induced) and the responsiveness of stem cells to adipokine signaling pathways. The number of colonic GFP(+) stem cells was significantly higher in the AOM-injected HF group compared to the LF group. The Lgr5(+) stem cells of the HF fed mice exhibited statistically significant increases in cell proliferation and decreases in apoptosis in response to AOM injection compared to the LF group. Colonic organoid cultures from lean mice treated with an adiponectin receptor agonist exhibited a reduction in Lgr5-GPF(+) stem cell number and an increase in apoptosis; however, this response was diminished in the organoid cultures from obese mice. These results suggest that the responsiveness of colonic stem cells to adiponectin in diet-induced obesity is impaired and may contribute to the stem cell accumulation observed in obesity.


Assuntos
Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Neoplasias do Colo/etiologia , Obesidade/complicações , Animais , Apoptose , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/patologia , Humanos , Masculino , Camundongos , Obesidade/metabolismo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA