Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
1.
Sci Rep ; 14(1): 12879, 2024 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839896

RESUMEN

Paneth cells (PCs), a subset of intestinal epithelial cells (IECs) found at the base of small intestinal crypts, play an essential role in maintaining intestinal homeostasis. Altered PCs function is associated with diverse intestinal pathologies, including ileal Crohn's disease (CD). CD patients with ileal involvement have been previously demonstrated to display impairment in PCs and decreased levels of anti-microbial peptides. Although the immunosuppressive drug Azathioprine (AZA) is widely used in CD therapy, the impact of AZA on IEC differentiation remains largely elusive. In the present study, we hypothesized that the orally administered drug AZA also exerts its effect through modulation of the intestinal epithelium and specifically via modulation of PC function. AZA-treated CD patients exhibited an ileal upregulation of AMPs on both mRNA and protein levels compared to non-AZA treated patients. Upon in vitro AZA stimulation, intestinal epithelial cell line MODE-K exhibited heightened expression levels of PC marker in concert with diminished cell proliferation but boosted mitochondrial OXPHOS activity. Moreover, differentiation of IECs, including PCs differentiation, was boosted in AZA-treated murine small intestinal organoids and was associated with decreased D-glucose consumption and decreased growth rates. Of note, AZA treatment strongly decreased Lgr5 mRNA expression as well as Ki67 positive cells. Further, AZA restored dysregulated PCs associated with mitochondrial dysfunction. AZA-dependent inhibition of IEC proliferation is accompanied by boosted mitochondria function and IEC differentiation into PC.


Asunto(s)
Azatioprina , Diferenciación Celular , Enfermedad de Crohn , Mucosa Intestinal , Células de Paneth , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/patología , Enfermedad de Crohn/metabolismo , Azatioprina/farmacología , Células de Paneth/metabolismo , Células de Paneth/efectos de los fármacos , Células de Paneth/patología , Humanos , Diferenciación Celular/efectos de los fármacos , Animales , Ratones , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Femenino , Masculino , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/patología , Adulto , Organoides/efectos de los fármacos , Organoides/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proliferación Celular/efectos de los fármacos , Persona de Mediana Edad , Línea Celular , Índice de Severidad de la Enfermedad
2.
EMBO J ; 42(21): e113975, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37718683

RESUMEN

Paneth cells (PCs), a specialized secretory cell type in the small intestine, are increasingly recognized as having an essential role in host responses to microbiome and environmental stresses. Whether and how commensal and pathogenic microbes modify PC composition to modulate inflammation remain unclear. Using newly developed PC-reporter mice under conventional and gnotobiotic conditions, we determined PC transcriptomic heterogeneity in response to commensal and invasive microbes at single cell level. Infection expands the pool of CD74+ PCs, whose number correlates with auto or allogeneic inflammatory disease progressions in mice. Similar correlation was found in human inflammatory disease tissues. Infection-stimulated cytokines increase production of reactive oxygen species (ROS) and expression of a PC-specific mucosal pentraxin (Mptx2) in activated PCs. A PC-specific ablation of MyD88 reduced CD74+ PC population, thus ameliorating pathogen-induced systemic disease. A similar phenotype was also observed in mice lacking Mptx2. Thus, infection stimulates expansion of a PC subset that influences disease progression.


Asunto(s)
Microbiota , Células de Paneth , Humanos , Animales , Ratones , Células de Paneth/metabolismo , Células de Paneth/patología , Intestino Delgado , Inflamación/patología , Citocinas/metabolismo
3.
World J Gastroenterol ; 29(21): 3222-3240, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37377591

RESUMEN

Crohn's disease (CD) is an inflammatory bowel disease characterized by immune-mediated flares affecting any region of the intestine alternating with remission periods. In CD, the ileum is frequently affected and about one third of patients presents with a pure ileal type. Moreover, the ileal type of CD presents epidemiological specificities like a younger age at onset and often a strong link with smoking and genetic susceptibility genes. Most of these genes are associated with Paneth cell dysfunction, a cell type found in the intestinal crypts of the ileum. Besides, a Western-type diet is associated in epidemiological studies with CD onset and increasing evidence shows that diet can modulate the composition of bile acids and gut microbiota, which in turn modulates the susceptibility of the ileum to inflammation. Thus, the interplay between environmental factors and the histological and anatomical features of the ileum is thought to explain the specific transcriptome profile observed in CD ileitis. Indeed, both immune response and cellular healing processes harbour differences between ileal and non-ileal CD. Taken together, these findings advocate for a dedicated therapeutic approach to managing ileal CD. Currently, interventional pharmacological studies have failed to clearly demonstrate distinct response profiles according to disease site. However, the high rate of stricturing disease in ileal CD requires the identification of new therapeutic targets to significantly change the natural history of this debilitating disease.


Asunto(s)
Enfermedad de Crohn , Enfermedades del Íleon , Ileítis , Humanos , Enfermedad de Crohn/epidemiología , Enfermedad de Crohn/genética , Enfermedad de Crohn/terapia , Íleon/patología , Ileítis/patología , Inflamación/patología , Células de Paneth/metabolismo , Células de Paneth/patología , Enfermedades del Íleon/patología
4.
Eur J Pediatr ; 182(6): 2775-2784, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37017768

RESUMEN

Previous studies suggest that Paneth cells are involved in NEC development. Defensin alpha 6 (DEFA6) and guanylate cyclase activator 2A (GUCA2A) are selective protein markers of Paneth cells. The objective was to explore DEFA6 and GUCA2A expression in intestinal tissue samples from newborn infants with and without NEC. Tissue samples from histologically intact intestine were analyzed from 70 infants: 43 underwent bowel resection due to NEC and 27 controls were operated due to conditions such as intestinal atresia, dysmotility, aganglionosis, pseudo-obstruction or volvulus. Each tissue sample was immunohistochemically stained for DEFA6 and GUCA2A. Semi-automated digital image analysis was performed to determine protein expression. Clinical data and protein expressions were compared between the groups. DEFA6 expression was lower in the NEC group (p = 0.006). Low DEFA6 correlated with risk of developing NEC in a logistic regression analysis, independently of gestational age and birth weight (OR 0.843 [CI 0.732-0.971]; p = 0.018). GUCA2A expression did not differ between the two groups. CONCLUSION: Lower expression of DEFA6 together with intact GUCA2A expression indicates that NEC patients have well-defined Paneth cells but diminished defensin activity. Our results suggest that DEFA6 could be used as a biomarker for NEC. WHAT IS KNOWN: • Previous studies of defensin activity in NEC have been inconsistent, showing that defensin levels may be increased or diminished in NEC. GUCA2A has to our knowledge never been studied in NEC. WHAT IS NEW: • This study benchmarks two specific Paneth cell markers (DEFA6 and GUCA2A) and their activity in individuals with and without NEC. • The key finding is that the NEC group had a lower DEFA6 expression compared to the Controls, while the expression of GUCA2A did not differ between the groups.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Lactante , Recién Nacido , Humanos , Células de Paneth/metabolismo , Células de Paneth/patología , Enterocolitis Necrotizante/diagnóstico , Peso al Nacer , Edad Gestacional , Defensinas/metabolismo
5.
Sci Rep ; 13(1): 3953, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36894646

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a chronic liver disease characterized by fibrosis that develops from fatty liver. Disruption of intestinal microbiota homeostasis, dysbiosis, is associated with fibrosis development in NASH. An antimicrobial peptide α-defensin secreted by Paneth cells in the small intestine is known to regulate composition of the intestinal microbiota. However, involvement of α-defensin in NASH remains unknown. Here, we show that in diet-induced NASH model mice, decrease of fecal α-defensin along with dysbiosis occurs before NASH onset. When α-defensin levels in the intestinal lumen are restored by intravenous administration of R-Spondin1 to induce Paneth cell regeneration or by oral administration of α-defensins, liver fibrosis is ameliorated with dissolving dysbiosis. Furthermore, R-Spondin1 and α-defensin improved liver pathologies together with different features in the intestinal microbiota. These results indicate that decreased α-defensin secretion induces liver fibrosis through dysbiosis, further suggesting Paneth cell α-defensin as a potential therapeutic target for NASH.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad del Hígado Graso no Alcohólico , alfa-Defensinas , Animales , Ratones , Aminoácidos , Colina , Dieta Alta en Grasa/métodos , Disbiosis/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Células de Paneth/patología
6.
Mucosal Immunol ; 16(4): 373-385, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36739089

RESUMEN

Interleukin (IL)-17 protects epithelial barriers by inducing the secretion of antimicrobial peptides. However, the effect of IL-17 on Paneth cells (PCs), the major producers of antimicrobial peptides in the small intestine, is unclear. Here, we show that the targeted ablation of the IL-17 receptor (IL-17R) in PCs disrupts their antimicrobial functions and decreases the frequency of ileal PCs. These changes become more pronounced after colonization with IL-17 inducing segmented filamentous bacteria. Mice with PCs that lack IL-17R show an increased inflammatory transcriptional profile in the ileum along with the severity of experimentally induced ileitis. These changes are associated with a decrease in the diversity of gut microbiota that induces a severe ileum pathology upon transfer to genetically susceptible mice, which can be prevented by the systemic administration of IL-17a/f in microbiota recipients. In an exploratory analysis of a small cohort of pediatric patients with Crohn's disease, we have found that a portion of these patients exhibits a low number of lysozyme-expressing ileal PCs and a high ileitis severity score, resembling the phenotype of mice with IL-17R-deficient PCs. Our study identifies IL-17R-dependent signaling in PCs as an important mechanism that maintains ileal homeostasis through the prevention of dysbiosis.


Asunto(s)
Ileítis , Microbiota , Receptores de Interleucina-17 , Animales , Niño , Humanos , Ratones , Péptidos Antimicrobianos , Disbiosis/microbiología , Ileítis/microbiología , Íleon/microbiología , Inflamación/patología , Interleucina-17 , Células de Paneth/patología , Receptores de Interleucina-17/genética
7.
J Gastroenterol ; 58(5): 444-457, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36739585

RESUMEN

BACKGROUND: Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. METHODS: To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. RESULTS: This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/ß-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. CONCLUSIONS: LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon , Sistema de Transporte de Aminoácidos y+L , Neoplasias Intestinales , Células de Paneth , Animales , Ratones , Transformación Celular Neoplásica/genética , Mucosa Intestinal/patología , Neoplasias Intestinales/metabolismo , Intestino Delgado/patología , Intestinos , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Sistema de Transporte de Aminoácidos y+L/metabolismo
8.
Nature ; 610(7932): 547-554, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36198790

RESUMEN

Loss of Paneth cells and their antimicrobial granules compromises the intestinal epithelial barrier and is associated with Crohn's disease, a major type of inflammatory bowel disease1-7. Non-classical lymphoid cells, broadly referred to as intraepithelial lymphocytes (IELs), intercalate the intestinal epithelium8,9. This anatomical position has implicated them as first-line defenders in resistance to infections, but their role in inflammatory disease pathogenesis requires clarification. The identification of mediators that coordinate crosstalk between specific IEL and epithelial subsets could provide insight into intestinal barrier mechanisms in health and disease. Here we show that the subset of IELs that express γ and δ T cell receptor subunits (γδ IELs) promotes the viability of Paneth cells deficient in the Crohn's disease susceptibility gene ATG16L1. Using an ex vivo lymphocyte-epithelium co-culture system, we identified apoptosis inhibitor 5 (API5) as a Paneth cell-protective factor secreted by γδ IELs. In the Atg16l1-mutant mouse model, viral infection induced a loss of Paneth cells and enhanced susceptibility to intestinal injury by inhibiting the secretion of API5 from γδ IELs. Therapeutic administration of recombinant API5 protected Paneth cells in vivo in mice and ex vivo in human organoids with the ATG16L1 risk allele. Thus, we identify API5 as a protective γδ IEL effector that masks genetic susceptibility to Paneth cell death.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Enfermedad de Crohn , Predisposición Genética a la Enfermedad , Linfocitos Intraepiteliales , Proteínas Nucleares , Células de Paneth , Animales , Humanos , Ratones , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Predisposición Genética a la Enfermedad/genética , Mucosa Intestinal/patología , Proteínas Nucleares/metabolismo , Células de Paneth/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Supervivencia Celular , Organoides , Alelos
9.
Korean J Gastroenterol ; 80(1): 34-37, 2022 07 25.
Artículo en Coreano | MEDLINE | ID: mdl-35879061

RESUMEN

Paneth cell carcinoma is a rare carcinoma composed predominantly or purely of malignant Paneth cells. An 83-year-old woman presented for evaluation of an elevated lesion in the stomach. On endoscopy, a 15 mm, discolored, elevated lesion with a central depression was found on the greater curvature of the gastric lower body. Endoscopic forceps biopsy revealed chronic gastritis with intestinal metaplasia. Magnifying endoscopy revealed an irregularly oval/tubular microsurface pattern and an irregular loop microvascular pattern with a demarcation line, suggestive of early gastric cancer. Therefore, endoscopic submucosal dissection was performed. Histopathological examination revealed a well-differentiated tubular adenocarcinoma limited to the muscularis mucosae and the tumor cells contained coarse eosinophilic granules in the cytoplasm. These tumor cells were diffusely and strongly stained for lysozyme, confirming the tumor diagnosis as Paneth cell carcinoma. Herein, we report a rare case of Paneth cell carcinoma and its endoscopic and histopathologic findings.


Asunto(s)
Carcinoma , Resección Endoscópica de la Mucosa , Neoplasias Gástricas , Anciano de 80 o más Años , Femenino , Humanos , Células de Paneth/patología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patología
10.
Histochem Cell Biol ; 158(1): 5-13, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35469099

RESUMEN

Paneth cells are antimicrobial peptide-secreting epithelial cells located at the bottom of the intestinal crypts of Lieberkühn. The crypts begin to form around postnatal day 7 (P7) mice, and Paneth cells usually appear within the first 2 weeks. Paneth cell dysfunction has been reported to correlate with Crohn's disease-like inflammation, showing narrow crypts or loss of crypt architecture in mice. The morphology of dysfunctional Paneth cells is similar to that of Paneth/goblet intermediate cells. However, it remains unclear whether the formation of the crypt is related to the maturation of Paneth cells. In this study, we investigated the histological changes including epigenetic modification in the mouse ileum postnatally and assessed the effect of the methyltransferase inhibitor on epithelium development using an organoid culture. The morphological and functional maturation of Paneth cells occurred in the first 2 weeks and was accompanied by histone H3 lysine 27 (H3K27) trimethylation, although significant differences in DNA methylation or other histone H3 trimethylation were not observed. Inhibition of H3K27 trimethylation in mouse ileal organoids suppressed crypt formation and Paneth cell maturation, until around P10. Overall, our findings show that post-transcriptional modification of histones, particularly H3K27 trimethylation, leads to the structural and functional maturation of Paneth cells during postnatal development.


Asunto(s)
Histonas , Células de Paneth , Animales , Diferenciación Celular , Epigénesis Genética/genética , Mucosa Intestinal , Ratones , Células de Paneth/patología , Células de Paneth/fisiología , Destete
11.
Histopathology ; 80(3): 529-537, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34608656

RESUMEN

AIMS: This study investigated the relationship between the differentiation of tumour cells into crypts, which is determined by cell differentiation into Paneth and neuroendocrine cells, and tumour infiltration in gastric dysplasia. METHODS AND RESULTS: The lesions were endoscopically biopsied low-grade dysplasia (LGD), endoscopically resected high-grade dysplasia (HGD) or cancer with submucosal invasion. LGD (n = 32) displayed crypt differentiation across the entire width of the tumour in all cases. Crypt differentiation was identified as a characteristic of tumours with low biological malignancy. HGD (n = 40) included tumours with a mixture of areas with and without crypt differentiation (n = 25) and tumours with crypt differentiation throughout the entire width (n = 15). Of the cancers with submucosal invasion (n = 30), the morphological progression of the HGD area with crypt differentiation, the HGD area without crypt differentiation and invasive cancer without crypt differentiation was confirmed for 23 samples. In two lesions, invasive cancer without crypt differentiation developed from HGD without crypt differentiation throughout the tumour width. In five samples, well-differentiated tubular adenocarcinoma with crypt differentiation developed from HGD with crypt differentiation and invaded with lamina propria-like stroma. CONCLUSIONS: Loss of crypt differentiation could be an objective indicator of infiltration in the progression of HGD to invasive cancer. The invasive potential of dysplasia depends upon the presence or absence of crypt differentiation.


Asunto(s)
Biopsia/clasificación , Diferenciación Celular , Células de Paneth/patología , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/patología , Anciano , Endoscopía Gastrointestinal , Femenino , Humanos , Masculino , Lesiones Precancerosas/clasificación , Estudios Retrospectivos , Neoplasias Gástricas/clasificación , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patología
12.
Cells ; 10(11)2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34831152

RESUMEN

BACKGROUND: Previous studies in mice indicated that Paneth cells and c-Kit-positive goblet cells represent the stem cell niche of the small intestine and colon, respectively, partly by supporting Wnt and Notch activation. Whether these cell populations play a similar role in human intestinal cancer remains unexplored. METHODS: We performed histopathological evaluation and immunohistochemical analysis of early colorectal adenomas and carcinoma adenoma from patients at the Hospital del Mar in Barcelona. We then determined the possible correlation between the different parameters analyzed and with patient outcomes. RESULTS: Paneth cells accumulate in a subset of human colorectal adenomas directly associated with Notch and Wnt/ß-catenin activation. Adenoma areas containing Paneth cells display increased vessel density in the lamina propria and higher levels of the stem cell marker EphB2. In an in-house cohort of 200 colorectal adenoma samples, we also observed a significant correlation between the presence of Paneth cells and Wnt activation. Kaplan-Meier analysis indicated that early adenoma patients carrying Paneth cell-positive tumors display reduced disease-free survival compared with patients with Paneth cell-free lesions. CONCLUSIONS: Our results indicate that Paneth cells contribute to the initial steps of cancer progression by providing the stem cell niche to adenoma cells, which could be therapeutically exploited.


Asunto(s)
Adenoma/metabolismo , Neoplasias Colorrectales/patología , Células de Paneth/patología , Transducción de Señal , beta Catenina/metabolismo , Humanos , Estimación de Kaplan-Meier , Pronóstico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor EphB2/metabolismo , Receptores Notch/metabolismo , Sinaptofisina/metabolismo , Proteínas Wnt/metabolismo
13.
Elife ; 102021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34633285

RESUMEN

Paneth cells constitutively produce antimicrobial peptides and growth factors that allow for intestinal homeostasis, host protection, and intestinal stem cell replication. Paneth cells rely heavily on the glycolytic metabolic program, which is in part controlled by the kinase complex Mechanistic target of rapamycin (mTORC1). Yet, little is known about mTOR importance in Paneth cell integrity under steady-state and inflammatory conditions. Our results demonstrate that IFN-γ, a crucial mediator of the intestinal inflammation, acts directly on murine Paneth cells to alter their mitochondrial integrity and membrane potential, resulting in an TORC1-dependent cell death mechanism distinct from canonical cell death pathways including apoptosis, necroptosis, and pyroptosis. These results were established with the purified cytokine and a physiologically relevant common Th1-inducing human parasite Toxoplasma gondii. Given the crucial role for IFN-γ, which is a cytokine frequently associated with the development of inflammatory bowel disease and compromised Paneth cell functions, the identified mechanisms underlying mTORC1-dependent Paneth cell death downstream of IFN-γ may provide promising novel approaches for treating intestinal inflammation.


Asunto(s)
Muerte Celular , Interferón gamma/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células de Paneth/patología , Animales , Femenino , Interferón gamma/genética , Intestino Delgado/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Toxoplasma , Toxoplasmosis/patología
14.
Cells ; 10(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34440876

RESUMEN

Paneth cells are specialized intestinal epithelial cells that are located at the base of small intestinal crypts and play a vital role in preserving the gut epithelium homeostasis. Paneth cells act as a safeguard from bacterial translocation across the epithelium and constitute the niche for intestinal stem cells in the small intestine by providing multiple niche signals. Recently, Paneth cells have become the focal point of investigations defining the mechanisms underlying the epithelium-microbiome interactions and pathogenesis of chronic gut mucosal inflammation and bacterial infection. Function of Paneth cells is tightly regulated by numerous factors at different levels, while Paneth cell defects have been widely documented in various gut mucosal diseases in humans. The post-transcription events, specific change in mRNA stability and translation by RNA-binding proteins (RBPs) and noncoding RNAs (ncRNAs) are implicated in many aspects of gut mucosal physiology by modulating Paneth cell function. Deregulation of RBPs and ncRNAs and subsequent Paneth cell defects are identified as crucial elements of gut mucosal pathologies. Here, we overview the posttranscriptional regulation of Paneth cells by RBPs and ncRNAs, with a particular focus on the increasing evidence of RBP HuR and long ncRNA H19 in this process. We also discuss the involvement of Paneth cell dysfunction in altered susceptibility of the intestinal epithelium to chronic inflammation and bacterial infection following disrupted expression of HuR and H19.


Asunto(s)
Células de Paneth/fisiología , ARN no Traducido/metabolismo , Proteínas de Unión al ARN/metabolismo , Regulación de la Expresión Génica , Homeostasis , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , ARN Circular/genética , ARN Circular/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN no Traducido/genética
15.
J Mol Med (Berl) ; 99(10): 1413-1426, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34129057

RESUMEN

Expression of the phosphatase of regenerating liver-3 (PRL-3) is known to promote tumor growth in gastrointestinal adenocarcinomas, and the incidence of tumor formation upon inflammatory events correlates with PRL-3 levels in mouse models. These carcinomas and their onset are associated with the impairment of intestinal cell homeostasis, which is regulated by a balanced number of Paneth cells and Lgr5 expressing intestinal stem cells (Lgr5+ ISCs). Nevertheless, the consequences of PRL-3 overexpression on cellular homeostasis and ISC fitness in vivo are unexplored. Here, we employ a doxycycline-inducible PRL-3 mouse strain to show that aberrant PRL-3 expression within a non-cancerous background leads to the death of Lgr5+ ISCs and to Paneth cell expansion. A higher dose of PRL-3, resulting from homozygous expression, led to mice dying early. A primary 3D intestinal culture model obtained from these mice confirmed the loss of Lgr5+ ISCs upon PRL-3 expression. The impaired intestinal organoid formation was rescued by a PRL inhibitor, providing a functional link to the observed phenotypes. These results demonstrate that elevated PRL-3 phosphatase activity in healthy intestinal epithelium impairs intestinal cell homeostasis, which correlates this cellular mechanism of tumor onset with PRL-3-mediated higher susceptibility to tumor formation upon inflammatory or mutational events.Key messages• Transgenic mice homozygous for PRL-3 overexpression die early.• PRL-3 heterozygous mice display disrupted intestinal self-renewal capacity.• PRL-3 overexpression alone does not induce tumorigenesis in the mouse intestine.• PRL-3 activity leads to the death of Lgr5+ ISCs and Paneth cell expansion.• Impairment of cell homeostasis correlates PRL-3 action with tumor onset mechanisms.


Asunto(s)
Homeostasis/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Intestinal/patología , Intestinos/patología , Masculino , Ratones , Ratones Transgénicos , Organoides/metabolismo , Organoides/patología , Células de Paneth/metabolismo , Células de Paneth/patología , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/patología
16.
Cells ; 10(6)2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072441

RESUMEN

Paneth cell defects in Crohn's disease (CD) patients (called the Type I phenotype) are associated with worse clinical outcomes. Recent studies have implicated mitochondrial dysfunction in Paneth cells as a mediator of ileitis in mice. We hypothesized that CD Paneth cells exhibit impaired mitochondrial health and that mitochondrial-targeted therapeutics may provide a novel strategy for ileal CD. Terminal ileal mucosal biopsies from adult CD and non-IBD patients were characterized for Paneth cell phenotyping and mitochondrial damage. To demonstrate the response of mitochondrial-targeted therapeutics in CD, biopsies were treated with vehicle or Mito-Tempo, a mitochondrial-targeted antioxidant, and RNA transcriptome was analyzed. During active CD inflammation, the epithelium exhibited mitochondrial damage evident in Paneth cells, goblet cells, and enterocytes. Independent of inflammation, Paneth cells in Type I CD patients exhibited mitochondrial damage. Mito-Tempo normalized the expression of interleukin (IL)-17/IL-23, lipid metabolism, and apoptotic gene signatures in CD patients to non-IBD levels. When stratified by Paneth cell phenotype, the global tissue response to Mito-Tempo in Type I patients was associated with innate immune, lipid metabolism, and G protein-coupled receptor (GPCR) gene signatures. Targeting impaired mitochondria as an underlying contributor to inflammation provides a novel treatment approach for CD.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/metabolismo , Inflamación/tratamiento farmacológico , Mitocondrias/metabolismo , Biopsia/métodos , Enterocitos/citología , Epitelio/efectos de los fármacos , Epitelio/patología , Humanos , Metabolismo de los Lípidos/fisiología , Células de Paneth/patología , Fenotipo
17.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G171-G184, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34159811

RESUMEN

Vitamin D deficiency is an environmental factor involved in the pathogenesis of inflammatory bowel disease (IBD); however, the mechanisms surrounding its role remain unclear. Previous studies conducted in an intestinal epithelial-specific vitamin D receptor (VDR) knockout model suggest that a lack of vitamin D signaling causes a reduction in intestinal autophagy. A potential link between vitamin D deficiency and dysregulated autophagy is microRNA (miR)-142-3p, which suppresses autophagy. In this study, we found that wild-type C57BL/6 mice fed a vitamin D-deficient diet for 5 wk had increased miR-142-3p expression in ileal tissues compared with mice that were fed a matched control diet. Interestingly, there was no difference in expression of key autophagy markers ATG16L1 and LC3II in the ileum whole tissue. However, Paneth cells of vitamin D-deficient mice were morphologically abnormal and had an accumulation of the autophagy adaptor protein p62, which was not present in the total crypt epithelium. These findings suggest that Paneth cells exhibit early markers of autophagy dysregulation within the intestinal epithelium in response to vitamin D deficiency and enhanced miR-142-3p expression. Finally, we demonstrated that treatment-naïve IBD patients with low levels of vitamin D have an increase in miR-142-3p expression in colonic tissues procured from "involved" areas of the disease. Taken together, our findings demonstrate that insufficient vitamin D levels alter expression of autophagy-regulating miR-142-3p in intestinal tissues of mice and patients with IBD, providing insight into the mechanisms by which vitamin D deficiency modulates IBD pathogenesis.NEW & NOTEWORTHY Vitamin D deficiency has a role in IBD pathogenesis, and although the mechanisms surrounding its role remain unclear, it has been suggested that autophagy dysregulation is involved. Here, we show increased ileal expression of autophagy-suppressing miR-142-3p in mice that were fed a vitamin D-deficient diet and in "involved" colonic biopsies from pediatric IBD patients with low vitamin D. miR-142-3p serves as a potential mechanism mediating vitamin D deficiency and reduced autophagy.


Asunto(s)
Íleon/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , MicroARNs/genética , Deficiencia de Vitamina D/metabolismo , Vitamina D/metabolismo , Adolescente , Animales , Autofagia , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Células Cultivadas , Niño , Células HCT116 , Células HeLa , Humanos , Íleon/patología , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , Deficiencia de Vitamina D/complicaciones
18.
Toxicol Appl Pharmacol ; 422: 115561, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33957193

RESUMEN

Arsenic is a global health concern that causes toxicity through ingestion of contaminated water and food. In vitro studies suggest that arsenic reduces stem and progenitor cell differentiation. Thus, this study determined if arsenic disrupted intestinal stem cell (ISC) differentiation, thereby altering the number, location, and/or function of intestinal epithelial cells. Adult male C57BL/6 mice were exposed to 0 or 100 ppb sodium arsenite (AsIII) through drinking water for 5 weeks. Duodenal sections were collected to assess changes in morphology, proliferation, and cell types. qPCR analysis revealed a 40% reduction in Lgr5 transcripts, an ISC marker, in the arsenic-exposed mice, although there were no changes in the protein expression of Olfm4. Secretory cell-specific transcript markers of Paneth (Defa1), Goblet (Tff3), and secretory transit amplifying (Math1) cells were reduced by 51%, 44%, and 30% respectively, in the arsenic-exposed mice, indicating significant impacts on the Wnt-dependent differentiation pathway. Further, protein levels of phosphorylated ß-catenin were reduced in the arsenic-exposed mice, which increased the expression of Wnt-dependent transcripts CD44 and c-myc. PCA analysis, followed by MANOVA and regression analyses, revealed significant changes and correlations between Lgr5 and the transit amplifying (TA) cell markers Math1 and Hes1, which are in the secretory cell pathway. Similar comparisons between Math1 and Defa1 show that terminal differentiation into Paneth cells is also reduced in the arsenic-exposed mice. The data suggests that ISCs are not lost following arsenic exposure, but rather, specific Wnt-dependent progenitor cell formation and terminal differentiation in the small intestine is reduced.


Asunto(s)
Arsenitos/toxicidad , Diferenciación Celular/efectos de los fármacos , Duodeno/efectos de los fármacos , Células de Paneth/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Compuestos de Sodio/toxicidad , Células Madre/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación hacia Abajo , Duodeno/metabolismo , Duodeno/patología , Masculino , Ratones Endogámicos C57BL , Células de Paneth/metabolismo , Células de Paneth/patología , Receptores Acoplados a Proteínas G/genética , Células Madre/metabolismo , Células Madre/patología , Factor Trefoil-3/genética , Factor Trefoil-3/metabolismo , Vía de Señalización Wnt , alfa-Defensinas/genética , alfa-Defensinas/metabolismo
19.
Cell Rep ; 35(3): 109026, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33882314

RESUMEN

Organoids allow the recapitulation of intestinal homeostasis and cancerogenesis in vitro; however, RNA sequencing (RNA-seq)-based methods for drug screens are missing. We develop targeted organoid sequencing (TORNADO-seq), a high-throughput, high-content drug discovery platform that uses targeted RNA-seq to monitor the expression of large gene signatures for the detailed evaluation of cellular phenotypes in organoids. TORNADO-seq is a fast, highly reproducible time- and cost-effective ($5 per sample) method that can probe cell mixtures and their differentiation state in the intestinal system. We apply this method to isolate drugs that enrich for differentiated cell phenotypes and show that these drugs are highly efficacious against cancer compared to wild-type organoids. Furthermore, TORNADO-seq facilitates in-depth insight into the mode of action of these drugs. Our technology can easily be adapted to many other systems and will allow for more systematic, large-scale, and quantitative approaches to study the biology of complex cellular systems.


Asunto(s)
Antineoplásicos/farmacología , Detección Precoz del Cáncer/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Organoides/efectos de los fármacos , Medicamentos bajo Prescripción/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Antineoplásicos/química , Antineoplásicos/clasificación , Diferenciación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Enterocitos/patología , Redes Reguladoras de Genes , Células Caliciformes/efectos de los fármacos , Células Caliciformes/metabolismo , Células Caliciformes/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Organoides/metabolismo , Organoides/patología , Células de Paneth/efectos de los fármacos , Células de Paneth/metabolismo , Células de Paneth/patología , Medicamentos bajo Prescripción/química , Medicamentos bajo Prescripción/clasificación , RNA-Seq , Análisis de Secuencia de ARN , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/clasificación
20.
Biochem Biophys Res Commun ; 545: 14-19, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33529805

RESUMEN

Paneth cells and Lgr5+ intestinal stem cells (Lgr5+ ISCs) constitute the stem cell niche and maintain small intestinal epithelial integrity by recognizing various niche factors derived from subepithelial cells and external antigens. Although it has been known that interferon-γ (IFN-γ), a Th1 cytokine, is associated with intestinal epithelial disruption during inflammation as a niche factor, dynamics of Paneth cells and Lgr5+ ISCs in response to IFN-γ remain to be understood. Here we show that CAG-tdTomato;Lgr5-EGFP (CT-LE) mice generated in this study enable to identify Paneth cells and Lgr5+ ISCs separately by fluorescence signals. Lgr5+ ISCs underwent cell death a little earlier than Paneth cells in response to IFN-γ by simultaneous tracking using CT-LE mice. In addition, the timing of cell death in most Paneth cells overlapped with Lgr5+ ISCs, suggesting that Paneth cell depletion is induced directly by IFN-γ. Taken together, we established a novel simultaneous stem cell niche tracking method and clarified the involvement of both Paneth cells and Lgr5+ ISCs in stem cell niche damage induced by IFN-γ, further contribute to understanding the mechanism for maintaining intestinal homeostasis by stem cell niche.


Asunto(s)
Interferón gamma/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Células de Paneth/efectos de los fármacos , Células de Paneth/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Sistemas de Computación , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Interferón gamma/fisiología , Mucosa Intestinal/fisiología , Ratones , Ratones Transgénicos , Células de Paneth/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interferón/metabolismo , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/fisiología , Células Madre/fisiología , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA