Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 37(6): e22975, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37159340

RESUMO

Intestinal epithelial stem cells (ISCs) are responsible for intestinal epithelial barrier renewal; thereby, ISCs play a critical role in intestinal pathophysiology research. While transgenic ISC reporter mice are available, advanced translational studies lack a large animal model. This study validates ISC isolation in a new porcine Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5) reporter line and demonstrates the use of these pigs as a novel colorectal cancer (CRC) model. We applied histology, immunofluorescence, fluorescence-activated cell sorting, flow cytometry, gene expression quantification, and 3D organoid cultures to whole tissue and single cells from the duodenum, jejunum, ileum, and colon of LGR5-H2B-GFP and wild-type pigs. Ileum and colon LGR5-H2B-GFP, healthy human, and murine biopsies were compared by mRNA fluorescent in situ hybridization (FISH). To model CRC, adenomatous polyposis coli (APC) mutation was induced by CRISPR/Cas9 editing in porcine LGR5-H2B-GFP colonoids. Crypt-base, green fluorescent protein (GFP) expressing cells co-localized with ISC biomarkers. LGR5-H2B-GFPhi cells had significantly higher LGR5 expression (p < .01) and enteroid forming efficiency (p < .0001) compared with LGR5-H2B-GFPmed/lo/neg cells. Using FISH, similar LGR5, OLFM4, HOPX, LYZ, and SOX9 expression was identified between human and LGR5-H2B-GFP pig crypt-base cells. LGR5-H2B-GFP/APCnull colonoids had cystic growth in WNT/R-spondin-depleted media and significantly upregulated WNT/ß-catenin target gene expression (p < .05). LGR5+ ISCs are reproducibly isolated in LGR5-H2B-GFP pigs and used to model CRC in an organoid platform. The known anatomical and physiologic similarities between pig and human, and those shown by crypt-base FISH, underscore the significance of this novel LGR5-H2B-GFP pig to translational ISC research.


Assuntos
Intestinos , Humanos , Suínos , Animais , Camundongos , Hibridização in Situ Fluorescente , Células-Tronco , Íleo , Colo , Proteínas de Fluorescência Verde/genética , Receptores Acoplados a Proteínas G/genética
2.
Gut Microbes ; 14(1): 2018898, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35012435

RESUMO

Acute intestinal mucositis is a common off-target effect of chemotherapy, leading to co-morbidities such as vomiting, diarrhea, sepsis, and death. We previously demonstrated that the presence of enteric bacteria modulates the extent of jejunal epithelial damage induced by doxorubicin (DXR) in mice. Despite conventional thinking of the crypt as a sterile environment, recent evidence suggests that bacterial signaling influences aISC function. In this study, we labeled aISCs using transgenic Lgr5-driven fluorescence or with immunostaining for OLFM4. We examined the effect of DXR in both germ free (GF) mice and mice depleted of microbiota using an established antimicrobial treatment protocol (AMBx). We found differences in DXR-induced loss of aISCs between GF mice and mice treated with AMBx. aISCs were decreased after DXR in GF mice, whereas AMBx mice retained aISC expression after DXR. Neither group of mice exhibited an inflammatory response to DXR, suggesting the difference in aISC retention was not due to differences in local tissue inflammation. Therefore, we suspected that there was a protective microbial signal present in the AMBx mice that was not present in the GF mice. 16S rRNA sequencing of jejunal luminal contents demonstrated that AMBx altered the fecal and jejunal microbiota. In the jejunal contents, AMBx mice had increased abundance of Ureaplasma and Burkholderia. These results suggest pro-survival signaling from microbiota in AMBx-treated mice to the aISCs, and that this signaling maintains aISCs in the face of chemotherapeutic injury. Manipulation of the enteric microbiota presents a therapeutic target for reducing the severity of chemotherapy-associated mucositis.


Assuntos
Antineoplásicos/efeitos adversos , Doxorrubicina/efeitos adversos , Jejuno/efeitos dos fármacos , Mucosite/prevenção & controle , Células-Tronco/efeitos dos fármacos , Administração Oral , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Antineoplásicos/administração & dosagem , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Vida Livre de Germes , Humanos , Jejuno/citologia , Jejuno/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosite/microbiologia , Células-Tronco/citologia , Fatores de Tempo
3.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G668-G681, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34643097

RESUMO

MicroRNA-mediated regulation is critical for the proper development and function of the small intestinal (SI) epithelium. However, it is not known which microRNAs are expressed in each of the cell types of the SI epithelium. To bridge this important knowledge gap, we performed comprehensive microRNA profiling in all major cell types of the mouse SI epithelium. We used flow cytometry and fluorescence-activated cell sorting with multiple reporter mouse models to isolate intestinal stem cells, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, tuft cells, and secretory progenitors. We then subjected these cell populations to small RNA-sequencing. The resulting atlas revealed highly enriched microRNA markers for almost every major cell type (https://sethupathy-lab.shinyapps.io/SI_miRNA/). Several of these lineage-enriched microRNAs (LEMs) were observed to be embedded in annotated host genes. We used chromatin-run-on sequencing to determine which of these LEMs are likely cotranscribed with their host genes. We then performed single-cell RNA-sequencing to define the cell type specificity of the host genes and embedded LEMs. We observed that the two most enriched microRNAs in secretory progenitors are miR-1224 and miR-672, the latter of which we found is deleted in hominin species. Finally, using several in vivo models, we established that miR-152 is a Paneth cell-specific microRNA.NEW & NOTEWORTHY In this study, first, microRNA atlas (and searchable web server) across all major small intestinal epithelial cell types is presented. We have demonstrated microRNAs that uniquely mark several lineages, including enteroendocrine and tuft. Identification of a key marker of mouse secretory progenitor cells, miR-672, which we show is deleted in humans. We have used several in vivo models to establish miR-152 as a specific marker of Paneth cells, which are highly understudied in terms of microRNAs.


Assuntos
Linhagem da Célula , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , MicroRNAs/genética , Transcriptoma , Animais , Biomarcadores/metabolismo , Separação Celular , Células Cultivadas , Biologia Computacional , Cães , Feminino , Citometria de Fluxo , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Organoides , RNA-Seq , Análise de Célula Única
4.
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
5.
Cell Mol Gastroenterol Hepatol ; 12(1): 119-140, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33571711

RESUMO

BACKGROUND & AIMS: aISCs (aISCs) are sensitive to acute insults including chemotherapy and irradiation. Regeneration after aISC depletion has primarily been explored in irradiation (IR). However, the cellular origin of epithelial regeneration after doxorubicin (DXR), a common chemotherapeutic, is poorly understood. METHODS: We monitored DXR's effect on aISCs by enumerating Lgr5-eGFP+ and Olfm4+ crypts, cleaved caspase-3 (CASP3+) immunofluorescence, and time-lapse organoid imaging. Lineage tracing from previously identified regenerative cell populations (Bmi1+, Hopx+, Dll1+, and Defa6+) was performed with DXR damage. Lineage tracing from aISCs was compared with lineage tracing from early progeny cells (transit-amplifying cells arising from aISCs 1 day predamage) in the context of DXR and IR. We compared stem cell and DNA damage response (DDR) transcripts in isolated aISCs and early progeny cells 6 and 24 hours after DXR. RESULTS: Epithelial regeneration after DXR primarily arose from early progeny cells generated by aISCs. Early progeny cells upregulated stem cell gene expression and lacked apoptosis induction (6 hours DXR: 2.5% of CASP3+ cells, p<0.0001). aISCs downregulated stem cell gene expression and underwent rapid apoptosis (6 hours DXR: 63.4% of CASP3+ cells). There was minimal regenerative contribution from Bmi1+, Hopx+, Dll1+, and Defa6+-expressing populations. In homeostasis, 48.4% of early progeny cells were BrdU+, and expressed low levels of DDR transcripts. CONCLUSIONS: We show that DXR effectively depleted aISCs in the small intestine and subsequent epithelial regeneration depended on nonquiescent early progeny cells of aISCs. The chemoresistant phenotype of the early progeny cells may rely on a dampened DDR in contrast to aISCs' robust DDR, which facilitates expeditious apoptosis.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Células Epiteliais/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Intestinos/metabolismo , Regeneração/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia
6.
Sci Rep ; 10(1): 21486, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33293626

RESUMO

Enteric bacteria and/or their products are necessary for doxorubicin (DXR)-induced small intestine mucosal damage. While DXR does not induce gross loss of epithelium, others have shown elevated serum endotoxin after DXR administration. However, the mechanism of movement is unknown. We hypothesized that DXR treatment resulted in increased paracellular translocation of bacteria or bacterial products through the small intestinal epithelium. We measured permeability after DXR administration using transepithelial resistance and macromolecular flux and assessed tight junctional gene expression and protein localization both in vitro using T84 cells and ex vivo using murine jejunum. DXR treatment increased flux of 4 kDa dextrans in mouse jejenum, but increased flux of 4, 10 and 20 kDa dextrans in T84 cells. Following DXR, we observed increased permeability, both in vitro and ex vivo, independent of bacteria. DXR induced increased expression of Cldn2 and Cldn4 in murine small intestine but increased only CLDN2 expression in T84 cells. DXR treatment induced disorganization of tight junctional proteins. We conclude that DXR increases paracellular transit of small macromolecules, including bacterial products, through the epithelium, by altering expression of tight junctional components and dynamic loosening of cellular tight junctions.


Assuntos
Doxorrubicina/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/metabolismo , Animais , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Doxorrubicina/metabolismo , Duodeno/metabolismo , Epitélio/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Jejuno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade/efeitos dos fármacos , Junções Íntimas/metabolismo
7.
Cell Mol Gastroenterol Hepatol ; 8(4): 549-560, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31330316

RESUMO

BACKGROUND: Paneth cells are professional secretory cells found within the small intestinal crypt epithelium. Although their role as part of the innate immune complex providing antimicrobial secretory products is well-known, the mechanisms that control secretory capacity are not well-understood. MIST1 is a scaling factor that is thought to control secretory capacity of exocrine cells. METHODS: Mist1+/+ and Mist1-/- mice were used to evaluate the function of MIST1 in small intestinal Paneth cells. We used histologic and immunofluorescence staining to evaluate small intestinal tissue for proliferation and lineage allocation. Total RNA was isolated to evaluate gene expression. Enteroid culture was used to evaluate the impact of the absence of MIST1 expression on intestinal stem cell function. RESULTS: Absence of MIST1 resulted in increased numbers of Paneth cells exhibiting an intermediate cell phenotype but otherwise did not alter overall epithelial cell lineage allocation. Muc2 and lysozyme staining confirmed the presence of intermediate cells at the crypt base of Mist1-/- mice. These changes were not associated with changes in mRNA expression of transcription factors associated with lineage allocation, and they were not abrogated by inhibition of Notch signaling. However, the absence of MIST1 expression was associated with alterations in Paneth cell morphology including decreased granule size and distended rough endoplasmic reticulum. Absence of MIST1 was associated with increased budding of enteroid cultures; however, there was no evidence of increased intestinal stem cell numbers in vivo. CONCLUSIONS: MIST1 plays an important role in organization of the Paneth cell secretory apparatus and managing endoplasmic reticulum stress. This role occurs downstream of Paneth cell lineage allocation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Celulas de Paneth/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Linhagem da Célula , Estresse do Retículo Endoplasmático , Retículo Endoplasmático Rugoso/fisiologia , Epitélio/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Intestino Delgado/fisiologia , Intestinos/fisiologia , Camundongos , Camundongos Knockout , Celulas de Paneth/fisiologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcriptoma
10.
Artigo em Inglês | MEDLINE | ID: mdl-26595884

RESUMO

BACKGROUND: Prior studies have shown the prognostic utility of measuring L-lactate in critically ill neonatal foals, both as single (at admission) and serial measurements. Greater prognostic sensitivity and specificity may be achieved by use of the area under the L-lactate versus time curve (LACArea ) over the first 24 hours of hospitalization, which captures both severity and duration of hyperlactatemia. Prior to application of this concept in sick equine neonates, a reference interval for LACArea should be determined. METHODS: The concentration of lactate [LAC] was measured in blood obtained via direct jugular venipuncture from clinically normal foals on Days 1 (birth-24 h of age), 3, 7, and 14 following birth at 6-hour intervals for each 24-hour period. LACArea was calculated using the trapezoidal method. Differences in LACArea by Day were determined by MANOVA with a priori Bonferroni correction, P ≤ 0.05. RESULTS: LACArea differed by Day (P = 0.001), being largest on Day 1, followed by Day 3. Days 7 and 14 were smallest and not different from each other. CONCLUSION: LACArea decreases substantially and predictably over the first week of life in normal neonatal foals. Knowing how LACArea normally changes over the first 2 weeks of life will aid in future study of LACArea as it applies to sick neonatal foals, allowing for consideration of maturational changes potentially unrelated to disease.


Assuntos
Biomarcadores/sangue , Cavalos/fisiologia , Ácido Láctico/sangue , Animais , Animais Recém-Nascidos , Estado Terminal , Cavalos/sangue , Hospitalização , Prognóstico , Valores de Referência , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...