Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Immunity ; 55(12): 2300-2317.e6, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36473468

RESUMO

Intestinal stem cell maturation and development coincide with gut microbiota exposure after birth. Here, we investigated how early life microbial exposure, and disruption of this process, impacts the intestinal stem cell niche and development. Single-cell transcriptional analysis revealed impaired stem cell differentiation into Paneth cells and macrophage specification upon antibiotic treatment in early life. Mouse genetic and organoid co-culture experiments demonstrated that a CD206+ subset of intestinal macrophages secreted Wnt ligands, which maintained the mesenchymal niche cells important for Paneth cell differentiation. Antibiotics and reduced numbers of Paneth cells are associated with the deadly infant disease, necrotizing enterocolitis (NEC). We showed that colonization with Lactobacillus or transfer of CD206+ macrophages promoted Paneth cell differentiation and reduced NEC severity. Together, our work defines the gut microbiota-mediated regulation of stem cell niches during early postnatal development.


Assuntos
Enterocolite Necrosante , Microbioma Gastrointestinal , Camundongos , Animais , Celulas de Paneth/fisiologia , Diferenciação Celular/fisiologia , Macrófagos
2.
Cell Mol Gastroenterol Hepatol ; 12(4): 1239-1250, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34153524

RESUMO

Paneth cells are professional secretory cells that classically play a role in the innate immune system by secreting antimicrobial factors into the lumen to control enteric bacteria. In this role, Paneth cells are able to sense cues from luminal bacteria and respond by changing production of these factors to protect the epithelial barrier. Paneth cells rely on autophagy to regulate their secretory capability and capacity. Disruption of this pathway through mutation of genes, such as Atg16L1, results in decreased Paneth cell function, dysregulated enteric microbiota, decreased barrier integrity, and increased risk of diseases such as Crohn's disease in humans. Upon differentiation Paneth cells migrate downward and intercalate among active intestinal stem cells at the base of small intestinal crypts. This localization puts them in a unique position to interact with active intestinal stem cells, and recent work shows that Paneth cells play a critical role in influencing the intestinal stem cell niche. This review discusses the numerous ways Paneth cells can influence intestinal stem cells and their niche. We also highlight the ways in which Paneth cells can alter cells and other organ systems.


Assuntos
Homeostase , Mucosa Intestinal/fisiologia , Celulas de Paneth/fisiologia , Regeneração , Animais , Diferenciação Celular , Microambiente Celular , Doença de Crohn/etiologia , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Suscetibilidade a Doenças , Interações entre Hospedeiro e Microrganismos , Humanos , Mucosa Intestinal/microbiologia , Microbiota , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Cicatrização
3.
FASEB J ; 34(8): 10299-10315, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32725957

RESUMO

Although previous studies show that exogenous nutrients regulate the stem cell function, little is known about the effects of L-arginine on intestinal stem cells (ISCs). In this study, we utilize mice, small intestinal (SI) organoids, and ISC-Paneth cell co-cultured models to clarify the role of L-arginine in ISC function. We find that exogenous L-arginine is essential for ISCs proliferation and intestinal epithelial renewal. Our data show that Paneth cells, a critical component of the ISCs niche, augment the ISCs function in response to L-arginine. Moreover, enhanced the expression of Wnt3a in Paneth cells, which is a ligand of the Wnt/ß-catenin signaling pathway, mediates the effects of L-arginine on ISCs function. Pre-treatment with L-arginine enhances the ISCs pool and protects the gut in response to injury provoked by murine tumor necrosis factor α (TNF-α) and 5-Fluorouracil (5-FU). Our findings establish that the regulation of Wnt3a in the Paneth cell niche by exogenous L-arginine couples ISCs function and favours a model in which the ISCs niche couples the nutrient levels to ISCs function.


Assuntos
Arginina/metabolismo , Intestino Delgado/metabolismo , Celulas de Paneth/metabolismo , Nicho de Células-Tronco/fisiologia , Células-Tronco/metabolismo , Animais , Proliferação de Células/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiologia , Intestino Delgado/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Organoides/metabolismo , Organoides/fisiologia , Celulas de Paneth/fisiologia , Células-Tronco/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Via de Sinalização Wnt/fisiologia
4.
Front Immunol ; 11: 646, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32351509

RESUMO

The Paneth cells reside in the small intestine at the bottom of the crypts of Lieberkühn, intermingled with stem cells, and provide a niche for their neighbors by secreting growth and Wnt-factors as well as different antimicrobial peptides including defensins, lysozyme and others. The most abundant are the human Paneth cell α-defensin 5 and 6 that keep the crypt sterile and control the local microbiome. In ileal Crohn's disease various mechanisms including established genetic risk factors contribute to defects in the production and ordered secretion of these peptides. In addition, life-style risk factors for Crohn's disease like tobacco smoking also impact on Paneth cell function. Taken together, current evidence suggest that defective Paneth cells may play the key role in initiating inflammation in ileal, and maybe ileocecal, Crohn's disease by allowing bacterial attachment and invasion.


Assuntos
Células-Tronco Adultas/fisiologia , Doença de Crohn/patologia , Microbioma Gastrointestinal/fisiologia , Doenças do Íleo/patologia , Inflamação/patologia , Celulas de Paneth/fisiologia , alfa-Defensinas/metabolismo , Animais , Autofagia , Doença de Crohn/imunologia , Humanos , Doenças do Íleo/imunologia , Inflamação/imunologia , Necroptose , Nicho de Células-Tronco
5.
Gastroenterol Clin North Am ; 48(4): 585-605, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31668185

RESUMO

Intestinal failure is defined as the inability to maintain fluid, nutrition, energy, and micronutrient balance that leads to the inability to gain or maintain weight, resulting in malnutrition and dehydration. Causes of intestinal failure include short bowel syndrome (ie, the physical loss of intestinal surface area and severe intestinal dysmotility). For patients with intestinal failure who fail to achieve enteral autonomy through intestinal rehabilitation programs, the current treatment options are expensive and associated with severe complications. Therefore, the need persists for next-generation therapies, including cell-based therapy, to increase intestinal regeneration, and development of the tissue-engineered small intestine.


Assuntos
Órgãos Artificiais , Intestino Delgado , Síndrome do Intestino Curto/cirurgia , Engenharia Tecidual/métodos , Proteínas Morfogenéticas Ósseas/metabolismo , Sistema Nervoso Entérico/citologia , Fator de Crescimento Epidérmico/fisiologia , Motilidade Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/fisiologia , Organoides/fisiologia , Celulas de Paneth/fisiologia , Receptores Notch/fisiologia , Regeneração , Transdução de Sinais/fisiologia , Transplante de Células-Tronco , Alicerces Teciduais , Via de Sinalização Wnt/fisiologia
6.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G580-G591, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31433211

RESUMO

Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.


Assuntos
Diarreia Infantil/genética , Molécula de Adesão da Célula Epitelial/genética , Mucosa Intestinal/citologia , Síndromes de Malabsorção/genética , Técnicas de Cultura de Tecidos/métodos , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Diarreia Infantil/patologia , Molécula de Adesão da Célula Epitelial/metabolismo , Feminino , Células Caliciformes/citologia , Células Caliciformes/metabolismo , Células Caliciformes/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Síndromes de Malabsorção/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , Celulas de Paneth/fisiologia
7.
J Pediatr Surg ; 54(11): 2402-2407, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30857731

RESUMO

PURPOSE: Necrotizing enterocolitis is associated with decreased intestinal perfusion and ischemia. Paneth cells, specialized epithelial cells, have been shown to regulate the intestinal vasculature and disruption of these cells has been associated with NEC. We hypothesized that Paneth cell disruption in immature mice intestine would decrease the perfusion of the intestinal microvasculature. METHODS: Paneth cells were disrupted in P14-16 mice using chemical (dithizone) and transgenic (diphtheria toxin) methodology. Six hours after Paneth cell disruption, Dylight 488 was injected directly into the left ventricle and allowed to perfuse for 5 minutes prior to intestinal harvesting. Tissue samples were evaluated with confocal fluorescence microscopy to quantify intestinal perfusion and samples were quantified by real time RT-PCR for gene expression. RESULTS: Dithizone treatment significantly decreased intestinal perfusion compared to controls (p < 0.01). However, diphtheria toxin treatment demonstrated no significant difference in perfusion (p > 0.21). Intestines from all treatment groups had similar PECAM staining, but intestines treated with dithizone had significantly decreased nNOS and iNOS gene expression compared to controls (p < 0.007). CONCLUSIONS: Paneth cell disruption significantly decreases the perfusion of the small intestinal microvasculature in a dithizone-specific manner. Dithizone has no effect on the amount of microvasculature, but does impact genes critical to nitric oxide signaling likely contributing to mesenteric vasoconstriction.


Assuntos
Ditizona/farmacologia , Intestino Delgado/irrigação sanguínea , Microcirculação/efeitos dos fármacos , Celulas de Paneth/efeitos dos fármacos , Animais , Toxina Diftérica/farmacologia , Modelos Animais de Doenças , Enterocolite Necrosante/etiologia , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Isquemia/induzido quimicamente , Camundongos , Óxido Nítrico/metabolismo , Celulas de Paneth/metabolismo , Celulas de Paneth/fisiologia , Transdução de Sinais
8.
Int Immunopharmacol ; 67: 54-61, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30530169

RESUMO

Complement activation is associated with regional inflammation during acute gastrointestinal injury (AGI). This study is designed to explore how intracellular C3 activation in Paneth cells (PCs) affects regeneration of intestinal epithelium during AGI. AGI was induced in wildtype C57BL/6 mice, with sham operation employed as control. Exogenous C3 (1 mg, I.P.) was applied at 6 h post-surgery. Intestinal crypts harvested from ileum were cultured with presence or absence of C3 (20 µg/ml), with small interfering RNA against BST1 and complement activation inhibitor selectively applied in vitro. The intestinal integrity, percentage of PCs and intestinal stem cells (ISCs) were evaluated. Importantly, cADPR, C3 fragments, and S6-related proteins were detected in PCs to inspect the mammalian target of rapamycin complex 1 (mTORC1) signaling. AGI caused breakdown of intestinal mucosa integrity and regional inflammation. Exogenous C3 by itself failed to promote the growth of intestinal epithelium, but distinctly enhanced the activity of PCs via intracellular activation, which subsequently supported the expansion of ISCs inside of intestinal crypts. Inhibition of C3 activation was associated with decreased expressions of S6, S6K1 and cADPR, with blocking BST1 found to depress cADPR only. Collectively, these data confirmed intracellular activation of C3 in PCs enhanced expansion of ISCs in response to acute injury. The mTORC1 signaling pathway in PCs contributed to this crosstalk during exogenous C3 treatment.


Assuntos
Complemento C3/metabolismo , Gastroenteropatias/imunologia , Mucosa Intestinal/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Celulas de Paneth/fisiologia , Ferimentos e Lesões/imunologia , Animais , Autorrenovação Celular , Células Cultivadas , ADP-Ribose Cíclica/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Ferimentos e Lesões/cirurgia
9.
Immunotherapy ; 10(15): 1325-1336, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30381988

RESUMO

AIM: To explore whether Paneth cells (PCs) and complement system collaborate in the repair of enteric epithelia during acute gastrointestinal injury (AGI). METHODS: Wild-type C57BL/6 mice were employed to induce AGI by performing colon ascendens stent surgery, with sham-operated as control. Exogenous C3 treatment was applied at 6-h postsurgery. After 48 h, overall survival, intestinal damage severity, and C3 intracellular activation were assessed in both epithelial cells and PCs. RESULTS: AGI caused a high mortality, while C3 therapy significantly attenuated epithelial damages and improved survival. Besides, exogenous C3 in vitro enhanced the proliferation and activity of PCs. Importantly, intracellular C3 activation was observed inside of PCs under C3 co-stimulation in vitro. CONCLUSION: C3 immunotherapy might play a valuable role in turnover of gut epithelia through intracellular activation in PCs.


Assuntos
Complemento C3/uso terapêutico , Gastroenteropatias/terapia , Imunoterapia/métodos , Mucosa Intestinal/efeitos dos fármacos , Celulas de Paneth/efeitos dos fármacos , Animais , Proliferação de Células , Células Cultivadas , Colo/cirurgia , Ativação do Complemento , Modelos Animais de Doenças , Feminino , Gastroenteropatias/imunologia , Humanos , Mucosa Intestinal/fisiologia , Espaço Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Celulas de Paneth/fisiologia , Cicatrização
10.
Innate Immun ; 23(6): 537-545, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28770667

RESUMO

Impaired Paneth cell expression of antimicrobial protein (AMP) lysozyme is found in patients with Crohn's disease with the autophagy gene ATG16L1 risk allele, in mice with mutations in autophagy genes Atg16L1, Atg5 and Atg7, and in Irgm1 knockout mice. Defective autophagy is also associated with expansion of resident Gram-negative bacteria in the intestinal lumen. These findings suggest that autophagy may control extracellular resident microbes by governing expression of lysozyme. To test the hypothesis that autophagy may have a defensive role in host response to resident extracellular microbes, we investigated the relationship between gut microbes, autophagy, and lysozyme. RAW 264.7 macrophages were treated with fecal slurry (FS), representing the resident microbial community; lipopolysaccharide (LPS); or butyrate, representing microbial products; or a representative resident Gram-negative bacterium Desulfovibrio vulgaris (DSV). FS, LPS, and DSV inhibited lysozyme expression, whereas butyrate had no effect. Induction of autophagy by rapamycin countered this inhibition, whereas silencing of the autophagy gene Irgm1 exacerbated the inhibitory effects of LPS on lysozyme expression. LPS also inhibited lysozyme activity against DSV and autophagy reversed this effect. Our results provide a novel insight into an interaction between gut bacteria, autophagy and AMP whereby autophagy may defend the host by countering the suppression of antimicrobial protein by Gram-negative bacteria.


Assuntos
Doença de Crohn/imunologia , Desulfovibrio vulgaris/imunologia , Infecções por Desulfovibrionaceae/imunologia , Microbioma Gastrointestinal/imunologia , Macrófagos/fisiologia , Muramidase/metabolismo , Celulas de Paneth/fisiologia , Animais , Autofagia , Fezes , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Humanos , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Muramidase/genética , Células RAW 264.7 , RNA Interferente Pequeno/genética , Sirolimo/farmacologia
11.
Sci Rep ; 6: 26129, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27188450

RESUMO

Recent studies have linked appearance of Paneth cells in colorectal adenomas to adenoma burden and male gender. However, the clinical importance of Paneth cells' associations with synchronous advanced adenoma (AA) and colorectal carcinoma (CRC) is currently unclear. We performed a comprehensive case-control study using 1,900 colorectal adenomas including 785 from females, and 1,115 from males. We prospectively reviewed and recorded Paneth cell status in the colorectal adenomas consecutively collected between February 2014 and June 2015. Multivariable logistic regression analyses revealed that, in contrast to the adenomas without Paneth cells, the Paneth cell-containing adenomas at distal colorectum were inversely associated with presence of a synchronous AA or CRC (odds ratio [OR] 0.39, P = 0.046), whereas no statistical significance was reached for Paneth cell-containing proximal colorectal adenomas (P = 0.33). Synchronous AA and CRC were significantly associated with older age (60 + versus <60 years, OR 1.60, P = 0.002), male gender (OR 1.42, P = 0.021), and a history of AA or CRC (OR 2.31, P < 0.001). However, synchronous CRC was not associated with Paneth cell status, or a history of AA or CRC. Paneth cell presence in the adenomas of distal colorectum may be a negative indicator for synchronous AA and CRC, and seems to warrant further studies.


Assuntos
Adenoma/complicações , Adenoma/patologia , Carcinoma/epidemiologia , Carcinoma/patologia , Neoplasias Colorretais/patologia , Celulas de Paneth/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Comorbidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
12.
Immunity ; 44(3): 553-567, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26982364

RESUMO

Intestinal epithelial cells (IECs) regulate gut immune homeostasis, and impaired epithelial responses are implicated in the pathogenesis of inflammatory bowel diseases (IBD). IEC-specific ablation of nuclear factor κB (NF-κB) essential modulator (NEMO) caused Paneth cell apoptosis and impaired antimicrobial factor expression in the ileum, as well as colonocyte apoptosis and microbiota-driven chronic inflammation in the colon. Combined RelA, c-Rel, and RelB deficiency in IECs caused Paneth cell apoptosis but not colitis, suggesting that NEMO prevents colon inflammation by NF-κB-independent functions. Inhibition of receptor-interacting protein kinase 1 (RIPK1) kinase activity or combined deficiency of Fas-associated via death domain protein (FADD) and RIPK3 prevented epithelial cell death, Paneth cell loss, and colitis development in mice with epithelial NEMO deficiency. Therefore, NEMO prevents intestinal inflammation by inhibiting RIPK1 kinase activity-mediated IEC death, suggesting that RIPK1 inhibitors could be effective in the treatment of colitis in patients with NEMO mutations and possibly in IBD.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Celulas de Paneth/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Apoptose/genética , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelB/genética
13.
Nihon Rinsho Meneki Gakkai Kaishi ; 39(6): 522-527, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-28049961

RESUMO

The intestinal epithelium constitutes a physical barrier between inner and outer side of our body. It also functions as a "hub" which connects factors that determine the development of inflammatory bowel disease, such as microbiota, susceptibility genes, and host immune response. Accordingly, recent studies have implicated and further featured the role of intestinal epithelial cell dysfunction in the pathophysiology of inflammatory bowel disease. For example, mucin producing goblet cells are usually "depleted" in ulcerative colitis patients. Studies have shown that those goblet cells exhibit various immune-regulatory functions in addition to mucin production, such as antigen presentation or cytokine production. Paneth cells are another key cell lineage that has been deeply implicated in the pathophysiology of Crohn's disease. Several susceptibility genes for Crohn's disease may lead to impairment of anti-bacterial peptide production and secretion by Paneth cells. Also, other susceptibility genes may determine the survival of Paneth cells, which leads to reduced Paneth cell function in the patient small intestinal mucosa. Further studies may reveal other unexpected roles of the intestinal epithelium in the pathophysiology of inflammatory bowel disease, and may help to develop alternative therapies targeted to intestinal epithelial cell functions.


Assuntos
Células Epiteliais/fisiologia , Doenças Inflamatórias Intestinais/etiologia , Intestinos/citologia , Apresentação de Antígeno , Peptídeos Catiônicos Antimicrobianos/metabolismo , Citocinas/biossíntese , Microbioma Gastrointestinal , Predisposição Genética para Doença/genética , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Células Caliciformes/fisiologia , Humanos , Imunidade , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/terapia , Terapia de Alvo Molecular , Mucinas/biossíntese , Celulas de Paneth/metabolismo , Celulas de Paneth/fisiologia
15.
J Gastrointest Surg ; 18(5): 1040-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24643495

RESUMO

BACKGROUND: Colorectal cancer remains the most common gastrointestinal cancer. While screening combined with effective surgical treatment has reduced its mortality, we still do not have effective means to prevent recurrence nor to treat metastatic disease. What we know about cancer biology has gone through revolutionary changes in recent decades. The advent of the cancer stem cell theory has accelerated our understanding of the cancer cell. However, there is increasing evidence that cancer cells are influenced by their surrounding microenvironment. PURPOSE: This review divides the tumor microenvironment into four functional components-the stem cell niche, cancer stroma, immune cells, and vascular endothelia-and examines their individual and collective influence on the growth and metastasis of the colon cancer stem cell. The discussion will highlight the need to fully exploit the tumor microenvironment when designing future prognostic tools and therapies.


Assuntos
Neoplasias do Colo/patologia , Fibroblastos/fisiologia , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral , Transformação Celular Neoplásica , Neoplasias do Colo/terapia , Células Dendríticas/fisiologia , Células Endoteliais/fisiologia , Humanos , Leucócitos/fisiologia , Células Progenitoras Mieloides/fisiologia , Metástase Neoplásica , Neovascularização Patológica/tratamento farmacológico , Celulas de Paneth/fisiologia
16.
Integr Biol (Camb) ; 6(3): 243-57, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24480852

RESUMO

The definition, regulation and function of intestinal stem cells (ISCs) has been hotly debated. Recent discoveries have started to clarify the nature of ISCs, but many questions remain. This review discusses the current advances and controversies of ISC biology as well as theoretical compartmental models that have been coupled with in vivo experimentation to investigate the mechanisms of ISC dynamics during homeostasis, tumorigenesis, repair and development. We conclude our review by discussing the key lingering questions in the field and proposing how many of these questions can be addressed using both compartmental models and experimental techniques.


Assuntos
Células-Tronco Adultas/citologia , Intestinos/citologia , Modelos Biológicos , Células-Tronco Adultas/classificação , Células-Tronco Adultas/fisiologia , Animais , Biomarcadores/metabolismo , Carcinogênese , Diferenciação Celular , Homeostase , Humanos , Intestinos/fisiologia , Intestinos/efeitos da radiação , Celulas de Paneth/citologia , Celulas de Paneth/fisiologia , Nicho de Células-Tronco
18.
Neonatology ; 103(1): 10-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23006982

RESUMO

Current models of necrotizing enterocolitis (NEC) propose that intraluminal microbes destroy intestinal mucosa and activate an inflammatory cascade that ends in necrosis. We suggest an alternate hypothesis wherein NEC is caused by injury to Paneth cells (PCs) in the intestinal crypts. PCs are specialized epithelia that protect intestinal stem cells from pathogens, stimulate stem cell differentiation, shape the intestinal microbiota, and assist in repairing the gut. Our novel model of NEC uses neonatal mice and ablates PCs followed by enteral infection. We contrast this model with other animal examples of NEC and the clinical disease. Selective destruction of PCs using dithizone likely releases tumor necrosis factor-α and other inflammatory mediators. We propose that this event produces inflammation in the submucosa, generates platelet-activating factor, and induces a coagulopathy. The role of PCs in NEC is consistent with the onset of disease in preterm infants after a period of PC-related maturation, the central role of PCs in crypt-related homeostasis, the anatomic location of pneumatosis intestinalis close to the crypts, and the proximity of PCs to occluded blood vessels that cause coagulation necrosis of the intestinal villi. We offer this hypothesis to promote new thoughts about how NEC occurs and its potential prevention.


Assuntos
Enterocolite Necrosante/etiologia , Celulas de Paneth/patologia , Celulas de Paneth/fisiologia , Animais , Formação de Conceito , Doença/etiologia , Enterocolite Necrosante/patologia , Enterocolite Necrosante/prevenção & controle , Saúde , Humanos , Recém-Nascido , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Modelos Biológicos
19.
Gastroenterology ; 143(6): 1518-1529.e7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22922422

RESUMO

BACKGROUND & AIMS: Wnt signaling regulates multiple aspects of intestinal physiology, including stem cell maintenance. Paneth cells support stem cells by secreting Wnt, but little is known about the exact sources and primary functions of individual Wnt family members. METHODS: We analyzed intestinal tissues and cultured epithelial cells from adult mice with conditional deletion of Wnt3 (Vil-CreERT2;Wnt3fl/fl mice). We also analyzed intestinal tissues and cells from Atoh1 mutant mice, which lack secretory cells. RESULTS: Unexpectedly, Wnt3 was dispensable for maintenance of intestinal stem cells in mice, indicating a redundancy of Wnt signals. By contrast, cultured crypt organoids required Paneth cell-derived Wnt3. Addition of exogenous Wnt, or coculture with mesenchymal cells, restored growth of Vil-CreERT2;Wnt3fl/fl crypt organoids. Intestinal organoids from Atoh1 mutant mice did not grow or form Paneth cells; addition of Wnt3 allowed growth in the absence of Paneth cells. Wnt signaling had a synergistic effect with the Lgr4/5 ligand R-spondin to induce formation of Paneth cells. Mosaic expression of Wnt3 in organoids using a retroviral vector promoted differentiation of Paneth cells in a cell-autonomous manner. CONCLUSIONS: Wnt is part of a signaling loop that affects homeostasis of intestinal stem and Paneth cells in mice. Wnt3 signaling is required for growth and development of organoid cultures, whereas nonepithelial Wnt signals could provide a secondary physiological source of Wnt.


Assuntos
Diferenciação Celular/fisiologia , Intestino Delgado/citologia , Celulas de Paneth/citologia , Células-Tronco/citologia , Proteína Wnt3/fisiologia , Animais , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Homeostase/fisiologia , Técnicas In Vitro , Intestino Delgado/fisiologia , Camundongos , Camundongos Transgênicos , Modelos Animais , Celulas de Paneth/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia
20.
FEBS J ; 279(18): 3475-87, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22632461

RESUMO

In vitro culture of intestinal tissue has been attempted for decades. Only recently did Sato et al. [Sato, T., Vries, R. G., Snippert, H. J., van de Wetering, M., Barker, N., Stange, D. E., van Es, J. H., Abo, A., Kujala, P., Peters, P. J., et al. (2009) Nature 459, 262-265] succeed in establishing long-term intestinal culture, demonstrating that cells expressing the Lgr5 gene can give rise to organoids with crypt-like domains similar to those found in vivo. In these cultures, Paneth cells provide essential signals supporting stem cell function. We have recently developed an individual cell-based computational model of the intestinal tissue [Buske, P., Galle, J., Barker, N., Aust, G., Clevers, H. & Loeffler, M. (2011) PLoS Comput Biol 7, e1001045]. The model is capable of quantitatively reproducing a comprehensive set of experimental data on intestinal cell organization. Here, we present a significant extension of this model that allows simulation of intestinal organoid formation in silico. For this purpose, we introduce a flexible basal membrane that assigns a bending modulus to the organoid surface. This membrane may be re-organized by cells attached to it depending on their differentiation status. Accordingly, the morphology of the epithelium is self-organized. We hypothesize that local tissue curvature is a key regulatory factor in stem cell organization in the intestinal tissue by controlling Paneth cell specification. In simulation studies, our model closely resembles the spatio-temporal organization of intestinal organoids. According to our results, proliferation-induced shape fluctuations are sufficient to induce crypt-like domains, and spontaneous tissue curvature induced by Paneth cells can control cell number ratios. Thus, stem cell expansion in an organoid depends sensitively on its biomechanics. We suggest a number of experiments that will enable new insights into mechano-transduction in the intestine, and suggest model extensions in the field of gland formation.


Assuntos
Intestinos/citologia , Mecanotransdução Celular/fisiologia , Organoides/metabolismo , Nicho de Células-Tronco , Fenômenos Biomecânicos , Contagem de Células , Diferenciação Celular , Proliferação de Células , Simulação por Computador , Células Epiteliais/fisiologia , Modelos Biológicos , Celulas de Paneth/fisiologia , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA