Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Cell ; 186(21): 4694-4709.e16, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37832525

RESUMEN

Cytoplasmic divisions are thought to rely on nuclear divisions and mitotic signals. We demonstrate in Drosophila embryos that cytoplasm can divide repeatedly without nuclei and mitotic CDK/cyclin complexes. Cdk1 normally slows an otherwise faster cytoplasmic division cycle, coupling it with nuclear divisions, and when uncoupled, cytoplasm starts dividing before mitosis. In developing embryos where CDK/cyclin activity can license mitotic microtubule (MT) organizers like the spindle, cytoplasmic divisions can occur without the centrosome, a principal organizer of interphase MTs. However, centrosomes become essential in the absence of CDK/cyclin activity, implying that the cytoplasm can employ either the centrosome-based interphase or CDK/cyclin-dependent mitotic MTs to facilitate its divisions. Finally, we present evidence that autonomous cytoplasmic divisions occur during unperturbed fly embryogenesis and that they may help extrude mitotically stalled nuclei during blastoderm formation. We postulate that cytoplasmic divisions occur in cycles governed by a yet-to-be-uncovered clock mechanism autonomous from CDK/cyclin complexes.


Asunto(s)
Citocinesis , Embrión no Mamífero , Animales , Núcleo Celular , Centrosoma , Ciclinas/metabolismo , Drosophila , Mitosis , Huso Acromático/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(10): e2320559121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38408237

RESUMEN

Basal progenitor cells serve as a stem cell pool to maintain the homeostasis of the epithelium of the foregut, including the esophagus and the forestomach. Aberrant genetic regulation in these cells can lead to carcinogenesis, such as squamous cell carcinoma (SCC). However, the underlying molecular mechanisms regulating the function of basal progenitor cells remain largely unknown. Here, we use mouse models to reveal that Hippo signaling is required for maintaining the homeostasis of the foregut epithelium and cooperates with p53 to repress the initiation of foregut SCC. Deletion of Mst1/2 in mice leads to epithelial overgrowth in both the esophagus and forestomach. Further molecular studies find that Mst1/2-deficiency promotes epithelial growth by enhancing basal cell proliferation in a Yes-associated protein (Yap)-dependent manner. Moreover, Mst1/2 deficiency accelerates the onset of foregut SCC in a carcinogen-induced foregut SCC mouse model, depending on Yap. Significantly, a combined deletion of Mst1/2 and p53 in basal progenitor cells sufficiently drives the initiation of foregut SCC. Therefore, our studies shed light on the collaborative role of Hippo signaling and p53 in maintaining squamous epithelial homeostasis while suppressing malignant transformation of basal stem cells within the foregut.


Asunto(s)
Carcinoma de Células Escamosas , Transducción de Señal , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Homeostasis , Transducción de Señal/genética , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Señalizadoras YAP
3.
Development ; 150(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37746871

RESUMEN

The stem/progenitor cell pool is indispensable for the development, homeostasis and regeneration of the gastric epithelium, owing to its defining ability to self-renew whilst supplying the various functional epithelial lineages needed to digest food efficiently. A detailed understanding of the intricacies and complexities surrounding the behaviours and roles of these stem cells offers insights, not only into the physiology of gastric epithelial development and maintenance, but also into the pathological consequences following aberrations in stem cell regulation. Here, we provide an insightful synthesis of the existing knowledge on gastric epithelial stem cell biology, including the in vitro and in vivo experimental techniques that have advanced such studies. We highlight the contributions of stem/progenitor cells towards patterning the developing stomach, specification of the differentiated cell lineages and maintenance of the mature epithelium during homeostasis and following injury. Finally, we discuss gaps in our understanding and identify key research areas for future work.


Asunto(s)
Células Madre , Estómago , Homeostasis , Diferenciación Celular , Linaje de la Célula
4.
Genes Dev ; 32(2): 181-193, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440247

RESUMEN

Epithelial tissues rely on a highly coordinated balance between self-renewal, proliferation, and differentiation, disruption of which may drive carcinogenesis. The epigenetic regulator KMT2D (MLL4) is one of the most frequently mutated genes in all cancers, particularly epithelial cancers, yet its normal function in these tissues is unknown. Here, we identify a novel role for KMT2D in coordinating this fine balance, as depletion of KMT2D from undifferentiated epidermal keratinocytes results in reduced proliferation, premature spurious activation of terminal differentiation genes, and disorganized epidermal stratification. Genome-wide, KMT2D interacts with p63 and is enriched at its target enhancers. Depletion of KMT2D results in a broad loss of enhancer histone modifications H3 Lys 4 (H3K4) monomethylation (H3K4me1) and H3K27 acetylation (H3K27ac) as well as reduced expression of p63 target genes, including key genes involved in epithelial development and adhesion. Together, these results reveal a critical role for KMT2D in the control of epithelial enhancers and p63 target gene expression, including the requirement of KMT2D for the maintenance of epithelial progenitor gene expression and the coordination of proper terminal differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos , Queratinocitos/metabolismo , Proteínas de Neoplasias/fisiología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Código de Histonas , Homeostasis , Humanos , Proteínas de Neoplasias/metabolismo
5.
Am J Respir Crit Care Med ; 209(11): 1338-1350, 2024 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-38259174

RESUMEN

Rationale: Pharmacological improvement of cystic fibrosis transmembrane conductance regulator (CFTR) function with elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes in patients with cystic fibrosis (CF). However, ETI effects on impaired mucosal homeostasis and host defense at the molecular and cellular levels in the airways of patients with CF remain unknown. Objectives: To investigate effects of ETI on the transcriptome of nasal epithelial and immune cells from children with CF at the single-cell level. Methods: Nasal swabs from 13 children with CF and at least one F508del allele aged 6 to 11 years were collected at baseline and 3 months after initiation of ETI, subjected to single-cell RNA sequencing, and compared with swabs from 12 age-matched healthy children. Measurements and Main Results: Proportions of CFTR-positive cells were decreased in epithelial basal, club, and goblet cells, but not in ionocytes, from children with CF at baseline and were restored by ETI therapy to nearly healthy levels. Single-cell transcriptomics revealed an impaired IFN signaling and reduced expression of major histocompatibility complex classes I and II encoding genes in epithelial cells of children with CF at baseline, which was partially restored by ETI. In addition, ETI therapy markedly reduced the inflammatory phenotype of immune cells, particularly of neutrophils and macrophages. Conclusions: Pharmacological improvement of CFTR function improves innate mucosal immunity and reduces immune cell inflammatory responses in the upper airways of children with CF at the single-cell level, highlighting the potential to restore epithelial homeostasis and host defense in CF airways by early initiation of ETI therapy.


Asunto(s)
Aminofenoles , Benzodioxoles , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Homeostasis , Humanos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Niño , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Masculino , Benzodioxoles/uso terapéutico , Benzodioxoles/farmacología , Aminofenoles/uso terapéutico , Aminofenoles/farmacología , Quinolonas/uso terapéutico , Quinolonas/farmacología , Indoles/uso terapéutico , Indoles/farmacología , Combinación de Medicamentos , Quinolinas/uso terapéutico , Quinolinas/farmacología , Pirazoles/uso terapéutico , Pirazoles/farmacología , Pirroles/uso terapéutico , Pirroles/farmacología , Mucosa Nasal/inmunología , Piridinas/uso terapéutico , Piridinas/farmacología
6.
Mol Cell ; 63(6): 976-89, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27594448

RESUMEN

Prostate inflammation has been suggested as an etiology for benign prostatic hyperplasia (BPH). We show that decreased expression of the androgen receptor (AR) in luminal cells of human BPH specimens correlates with a higher degree of regional prostatic inflammation. However, the cause-and-effect relationship between the two events remains unclear. We investigated specifically whether attenuating AR activity in prostate luminal cells induces inflammation. Disrupting luminal cell AR signaling in mouse models promotes cytokine production cell-autonomously, impairs epithelial barrier function, and induces immune cell infiltration, which further augments local production of cytokines and chemokines including Il-1 and Ccl2. This inflammatory microenvironment promotes AR-independent prostatic epithelial proliferation, which can be abolished by ablating IL-1 signaling or depleting its major cellular source, the macrophages. This study demonstrates that disrupting luminal AR signaling promotes prostate inflammation, which may serve as a mechanism for resistance to androgen-targeted therapy for prostate-related diseases.


Asunto(s)
Células Epiteliales/metabolismo , Homeostasis/genética , Macrófagos/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/genética , Receptores Androgénicos/genética , Animales , Proliferación Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica , Homeostasis/inmunología , Humanos , Inflamación , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Infiltración Neutrófila , Próstata/inmunología , Próstata/patología , Hiperplasia Prostática/inmunología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Receptores Androgénicos/inmunología , Transducción de Señal , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología
7.
J Virol ; 96(5): e0118121, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35019722

RESUMEN

Papillomaviruses exclusively infect stratified epithelial tissues and cause chronic infections. To achieve this, infected cells must remain in the epithelial basal layer alongside their uninfected neighbors for years or even decades. To examine how papillomaviruses achieve this, we used the in vivo MmuPV1 (Mus musculus papillomavirus 1) model of lesion formation and persistence. During early lesion formation, an increased cell density in the basal layer, as well as a delay in the infected cells' commitment to differentiation, was apparent in cells expressing MmuPV1 E6/E7 RNA. Using cell culture models, keratinocytes exogenously expressing MmuPV1 E6, but not E7, recapitulated this delay in differentiation postconfluence and also grew to a significantly higher density. Cell competition assays further showed that MmuPV1 E6 expression led to a preferential persistence of the cell in the first layer, with control cells accumulating almost exclusively in the second layer. Interestingly, the disruption of MmuPV1 E6 binding to MAML1 protein abrogated these phenotypes. This suggests that the interaction between MAML1 and E6 is necessary for the lower (basal)-layer persistence of MmuPV1 E6-expressing cells. Our results indicate a role for E6 in lesion establishment by facilitating the persistence of infected cells in the epithelial basal layer, a mechanism that is most likely shared by other papillomavirus types. Interruption of this interaction is predicted to impede persistent papillomavirus infection and consequently provides a novel treatment target. IMPORTANCE Persistent infection with high-risk HPV types can lead to development of HPV-associated cancers, and persistent low-risk HPV infection causes problematic diseases, such as recurrent respiratory papillomatosis. The management and treatment of these conditions pose a considerable economic burden. Maintaining a reservoir of infected cells in the basal layer of the epithelium is critical for the persistence of infection in the host, and our studies using the mouse papillomavirus model suggest that E6 gene expression leads to the preferential persistence of epithelial cells in the lower layers during stratification. The E6 interaction with MAML1, a component of the Notch pathway, is required for this phenotype and is linked to E6 effects on cell density and differentiation. These observations are likely to reflect a common E6 role that is preserved among papillomaviruses and provide us with a novel therapeutic target for the treatment of recalcitrant lesions.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Animales , Diferenciación Celular , Epitelio/metabolismo , Epitelio/virología , Queratinocitos/virología , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Int J Mol Sci ; 24(18)2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37762681

RESUMEN

Mucus secreted by goblet cells (GCs) may play an important role in intestinal transit function. Our previous study found that Piezo1 protein is essential for GC function; however, the effect of GC Piezo1 on intestinal transit function is unclear. Our study aimed to investigate the effect of Piezo1 in GCs on intestinal transit and the potential mechanism. We compared intestinal mucus, fecal form, intestinal transit time, intestinal epithelial cell composition, and stem cell function in WT and GC-specific Piezo1-deficient (Piezo1ΔGC) mice. Our results revealed a correlation between mucus and intestinal transit: the less mucus there was, the slower the intestinal transit. Piezo1 deficiency in GCs led to decreased mucus synthesis and also disrupted the ecological niche of colon stem cells (CSCs). Through organoid culture, we found that the capacity of proliferation and differentiation in Piezo1ΔGC mouse CSCs was significantly decreased, which also led to a reduced source of GCs. Further studies found that the reduced Wnt and Notch signals in colon crypts might be the potential mechanism. These results indicated the importance of GC Piezo1 in intestinal transit function, which acts by maintaining the homeostasis of intestinal epithelial cells and mucus.

9.
Immunol Rev ; 286(1): 86-101, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30294961

RESUMEN

Innate lymphoid cells (ILC) are a recently identified group of tissue-resident innate lymphocytes. Available data support the view that ILC or their progenitors are deposited and retained in tissues early during ontogeny. Thereby, ILC become an integral cellular component of tissues and organs. Here, we will review the intriguing relationships between ILC and basic developmental and homeostatic processes within tissues. Studying ILC has already led to the appreciation of the integral roles of immune cells in tissue homeostasis, morphogenesis, metabolism, regeneration, and growth. This area of immunology has not yet been studied in-depth but is likely to reveal important networks contributing to disease tolerance and may be harnessed for future therapeutic approaches.


Asunto(s)
Inmunidad Innata , Inmunoterapia/tendencias , Linfocitos/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Plasticidad de la Célula , Homeostasis , Humanos , Tolerancia Inmunológica
10.
Physiology (Bethesda) ; 35(5): 328-337, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32783609

RESUMEN

Polyamines regulate a variety of physiological functions and are involved in pathogenesis of diverse human diseases. The epithelium of the mammalian gut mucosa is a rapidly self-renewing tissue in the body, and its homeostasis is preserved through well-controlled mechanisms. Here, we highlight the roles of cellular polyamines in maintaining the integrity of the gut epithelium, focusing on the emerging evidence of polyamines in the regulation of gut epithelial renewal and barrier function. Gut mucosal growth depends on the available supply of polyamines to the dividing cells in the crypts, and polyamines are also essential for normal gut epithelial barrier function. Polyamines modulate expression of various genes encoding growth-associated proteins and intercellular junctions via distinct mechanisms involving RNA-binding proteins and noncoding RNAs. With the rapid advance of polyamine biology, polyamine metabolism and transport are promising therapeutic targets in our efforts to protect the gut epithelium and barrier function in patients with critical illnesses.


Asunto(s)
Proliferación Celular , Autorrenovación de las Células , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Poliaminas/metabolismo , Animales , Células Epiteliales/patología , Humanos , Mucosa Intestinal/patología , Permeabilidad , Transducción de Señal
11.
J Endocrinol Invest ; 44(8): 1571-1579, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33683663

RESUMEN

Thyroid hormones (THs) are key endocrine regulators of tissue development and homeostasis. They are constantly released into the bloodstream and help to regulate many cell functions. The principal products released by the follicular epithelial cells are T3 and T4. T4, which is the less active form of TH, is produced in greater amounts than T3, which is the most active form of TH. This mechanism highlights the importance of the peripheral regulation of TH levels that goes beyond the central axis. Skin, muscle, liver, bone and heart are finely regulated by TH. In particular, skin is among the target organs most influenced by TH, which is essential for skin homeostasis. Accordingly, skin diseases are associated with an altered thyroid status. Alopecia, dermatitis and vitiligo are associated with thyroiditis and alopecia and eczema are frequently correlated with the Graves' disease. However, only in recent decades have studies started to clarify the molecular mechanisms underlying the effects of TH in epidermal homeostasis. Herein, we summarize the most frequent clinical epidermal alterations linked to thyroid diseases and review the principal mechanisms involved in TH control of keratinocyte proliferation and functional differentiation. Our aim is to define the open questions in this field that are beginning to be elucidated thanks to the advent of mouse models of altered TH metabolism and to obtain novel insights into the physiopathological consequences of TH metabolism on the skin.


Asunto(s)
Enfermedades de la Piel , Enfermedades de la Tiroides , Hormonas Tiroideas/metabolismo , Animales , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Epitelio/fisiopatología , Homeostasis/fisiología , Humanos , Modelos Animales , Enfermedades de la Piel/etiología , Enfermedades de la Piel/patología , Enfermedades de la Tiroides/complicaciones , Enfermedades de la Tiroides/metabolismo
12.
Proc Natl Acad Sci U S A ; 115(48): 12218-12223, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30404917

RESUMEN

Epithelial homeostasis requires the precise balance of epithelial stem/progenitor proliferation and differentiation. While many signaling pathways that regulate epithelial stem cells have been identified, it is probable that other regulators remain unidentified. Here, we use gene-expression profiling by targeted DamID to identify the stem/progenitor-specific transcription and signaling factors in the Drosophila midgut. Many signaling pathway components, including ligands of most major pathways, exhibit stem/progenitor-specific expression and have regulatory regions bound by both intrinsic and extrinsic transcription factors. In addition to previously identified stem/progenitor-derived ligands, we show that both the insulin-like factor Ilp6 and TNF ligand eiger are specifically expressed in the stem/progenitors and regulate normal tissue homeostasis. We propose that intestinal stem cells not only integrate multiple signals but also contribute to and regulate the homeostatic signaling microenvironmental niche through the expression of autocrine and paracrine factors.


Asunto(s)
Drosophila/fisiología , Intestinos/citología , Nicho de Células Madre , Células Madre/citología , Animales , Drosophila/citología , Proteínas de Drosophila/metabolismo , Homeostasis , Transducción de Señal , Células Madre/metabolismo
13.
Gastroenterology ; 157(3): 731-743, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31103627

RESUMEN

BACKGROUND & AIMS: Paneth cells secrete antimicrobial proteins including lysozyme via secretory autophagy as part of the mucosal protective response. The ELAV like RNA-binding protein 1 (ELAVL1, also called HuR) regulates stability and translation of messenger RNAs (mRNAs) and many aspects of mucosal physiology. We studied the posttranscriptional mechanisms by which HuR regulates Paneth cell function. METHODS: Intestinal mucosal tissues were collected from mice with intestinal epithelium (IE)-specific disruption of HuR (IE-HuR-/-), HuRfl/fl-Cre- mice (controls), and patients with inflammatory bowel diseases and analyzed by histology and immunohistochemistry. Paneth cell functions were determined by lysozyme-immunostaining assays. We isolated primary enterocytes from IE-HuR-/- and control mice and derived intestinal organoids. HuR and the chaperone CNPY3 were overexpressed from transgenes in intestinal epithelial cells (IECs) or knocked down with small interfering RNAs. We performed RNA pulldown assays to investigate interactions between HuR and its target mRNAs. RESULTS: Intestinal tissues from IE-HuR-/- mice had reduced numbers of Paneth cells, and Paneth cells had fewer lysozyme granules per cell, compared with tissues from control mice, but there were no effects on Goblet cells or enterocytes. Intestinal mucosa from patients with inflammatory bowel diseases had reduced levels of HuR and fewer Paneth cells. IE-HuR-/- mice did not have the apical distribution of TLR2 in the intestinal mucosa as observed in control mice. IECs from IE-HuR-/- mice expressed lower levels of CNPY3. Intestinal organoids from IE-HuR-/- mice were smaller and contained fewer buds compared with those generated from controls, and had fewer lysozyme-positive cells. In IECs, knockdown of HuR decreased levels of the autophagy proteins LC3-I and LC3-II, compared with control cells, and prevented rapamycin-induced autophagy. We found HuR to interact directly with the Cnpy3 mRNA coding region and increase levels of CNPY3 by increasing the stability and translation of Cnpy3 mRNA. CNPY3 bound TLR2, and cells with knockdown of CNPY3 or HuR lost membrane localization of TLR2, but increased cytoplasmic levels of TLR2. CONCLUSIONS: In studies of mice, IECs, and human tissues, we found HuR to increase expression of CNPY3 at the posttranscriptional level. CNPY3 is required for membrane localization of TLR2 and Paneth cell function.


Asunto(s)
Membrana Celular/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Intestino Delgado/metabolismo , Chaperonas Moleculares/metabolismo , Células de Paneth/metabolismo , Procesamiento Postranscripcional del ARN , Receptor Toll-Like 2/metabolismo , Animales , Estudios de Casos y Controles , Células Cultivadas , Proteína 1 Similar a ELAV/deficiencia , Proteína 1 Similar a ELAV/genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Intestino Delgado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Células de Paneth/patología , Transporte de Proteínas , Transducción de Señal , Regulación hacia Arriba
14.
FASEB J ; 33(12): 14067-14082, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31657630

RESUMEN

Glucocorticoids (GCs) are important hormones involved in the regulation of multiple physiologic functions. GCs are also widely used in anti-inflammatory/immunosuppressant drugs. GCs are synthesized by the adrenal cortex as part of the hypothalamus-pituitary-adrenal axis and also by intestinal epithelial cells, among other peripheral sites. GCs are one of the main therapy choices for the exacerbations of inflammatory bowel disease, but they are not useful to prolong remission, and development of tolerance with secondary treatment failure is frequent. Thus, GC actions at the intestinal epithelial level are of great importance, both physiologically and pharmacologically. We generated a tamoxifen-inducible nuclear receptor subfamily 3 group C member 1 (NR3C1)ΔIEC mouse model to study the effects of GCs on epithelial cells in vivo. Nr3c1 deletion in epithelial cells of the small intestine and colon was associated with limited colonic inflammation at 1 wk postdeletion, involving augmented epithelial proliferation and mucus production, plus local and systemic immune/inflammatory changes. This phenotype regressed substantially, but not completely, after 2 wk. The mechanism may involve augmented inflammatory signaling by epithelial cells or defective barrier function. We conclude that the epithelial GC receptor plays a significant role in colonic homeostasis in basal conditions, but its deficiency can be compensated in the short term. Future studies are required to assess the impact of Nr3c1 deletion in other conditions such as experimental colitis.-Aranda, C. J., Arredondo-Amador, M., Ocón, B., Lavín, J. L., Aransay, A. M., Martínez-Augustin, O., Sánchez de Medina, F. Intestinal epithelial deletion of the glucocorticoid receptor NR3C1 alters expression of inflammatory mediators and barrier function.


Asunto(s)
Células Epiteliales/metabolismo , Inflamación/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Antagonistas de Estrógenos/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores de Glucocorticoides/genética , Tamoxifeno/toxicidad
15.
Int J Biol Sci ; 20(2): 554-568, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38169732

RESUMEN

The vertebrate adult intestinal epithelium has a high self-renewal rate driven by intestinal stem cells (ISCs) in the crypts, which play central roles in maintaining intestinal integrity and homeostasis. However, the underlying mechanisms remain elusive. Here we showed that protein arginine methyltransferase 1 (PRMT1), a major arginine methyltransferase that can also function as a transcription co-activator, was highly expressed in the proliferating cells of adult mouse intestinal crypts. Intestinal epithelium-specific knockout of PRMT1, which ablates PRMT1 gene starting during embryogenesis, caused distinct, region-specific effects on small intestine and colon: increasing and decreasing the goblet cell number in the small intestinal and colonic crypts, respectively, leading to elongation of the crypts in small intestine but not colon, while increasing crypt cell proliferation in both regions. We further generated a tamoxifen-inducible intestinal epithelium-specific PRMT1 knockout mouse model and found that tamoxifen-induced knockout of PRMT1 in the adult mice resulted in the same region-specific intestinal phenotypes. Thus, our studies have for the first time revealed that the epigenetic enzyme PRMT1 has distinct, region-specific roles in the maintenance of intestinal epithelial architecture and homeostasis, although PRMT1 may influence intestinal development.


Asunto(s)
Intestino Delgado , Proteína-Arginina N-Metiltransferasas , Animales , Ratones , Arginina , Proliferación Celular/genética , Células Epiteliales/metabolismo , Homeostasis/genética , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Ratones Noqueados , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Tamoxifeno
16.
Front Cell Infect Microbiol ; 14: 1346087, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736751

RESUMEN

Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.


Asunto(s)
Homeostasis , Inmunidad Innata , Mucosa Intestinal , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Animales , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/inmunología , Células Epiteliales/microbiología , Transducción de Señal , Inmunidad Adaptativa , Macrófagos/inmunología , Macrófagos/microbiología , Interacciones Huésped-Patógeno
17.
Hum Cell ; 37(4): 1080-1090, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38814518

RESUMEN

Airway epithelium represents a physical barrier against toxic substances and pathogens but also presents pattern recognition receptors on the epithelial cells that detect pathogens leading to molecule release and sending signals that activate both the innate and adaptive immune responses. Thus, impaired airway epithelial function and poor integrity may increase the recurrence of infections. Probiotic use in respiratory diseases as adjuvant of traditional therapy is increasingly widespread. There is growing interest in the use of non-viable heat-killed bacteria, such as tyndallized bacteria (TB), due to safety concerns and to their immunomodulatory properties. This study explores in vitro the effects of a TB blend on the immune activation of airway epithelium. 16HBE bronchial epithelial cells were exposed to different concentrations of TB. Cell viability, TB internalization, TLR2 expression, IL-6, IL-8 and TGF-ßl expression/release, E-cadherin expression and wound healing were assessed. We found that TB were tolerated, internalized, increased TLR2, E-cadherin expression, IL-6 release and wound healing but decreased both IL-8 and TGF-ßl release. In conclusion, TB activate TLR2 pathway without inducing a relevant pro-inflammatory response and improve barrier function, leading to the concept that TB preserve epithelial homeostasis and could be used as strategy to prevent and to manage respiratory infection, exacerbations included.


Asunto(s)
Bronquios , Células Epiteliales , Inmunidad Innata , Receptor Toll-Like 2 , Humanos , Receptor Toll-Like 2/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Bronquios/citología , Bronquios/inmunología , Interleucina-6/metabolismo , Probióticos , Mucosa Respiratoria/inmunología , Cadherinas/metabolismo , Expresión Génica , Células Cultivadas , Interleucina-8/metabolismo , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Supervivencia Celular
18.
Mol Cell Biol ; : 1-18, 2024 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-39155414

RESUMEN

The histone variant H2A.Z plays important functions in the regulation of gene expression. In mammals, it is encoded by two genes, giving rise to two highly related isoforms named H2A.Z.1 and H2A.Z.2, which can have similar or antagonistic functions depending on the promoter. Knowledge of the physiopathological consequences of such functions emerges, but how the balance between these isoforms regulates tissue homeostasis is not fully understood. Here, we investigated the relative role of H2A.Z isoforms in intestinal epithelial homeostasis. Through genome-wide analysis of H2A.Z genomic localization in differentiating Caco-2 cells, we uncovered an enrichment of H2A.Z isoforms on the bodies of genes which are induced during enterocyte differentiation, stressing the potential importance of H2A.Z isoforms dynamics in this process. Through a combination of in vitro and in vivo experiments, we further demonstrated the two isoforms cooperate for stem and progenitor cells proliferation, as well as for secretory lineage differentiation. However, we found that they antagonistically regulate enterocyte differentiation, with H2A.Z.1 preventing terminal differentiation and H2A.Z.2 favoring it. Altogether, these data indicate that H2A.Z isoforms are critical regulators of intestine homeostasis and may provide a paradigm of how the balance between two isoforms of the same chromatin structural protein can control physiopathological processes.

19.
J Hazard Mater ; 479: 135734, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39244982

RESUMEN

Intestinal epithelium has the largest surface of human body, contributes dramatically to defense of toxicant-associated intestinal injury. Triclosan (TCS) and triclocarban (TCC), extensively employed as antibacterial agents in personal care products (PCPs) and healthcare facilities, caused serious damage to human intestine. However, the role of the intestinal epithelium in TCS/TCC-induced intestinal toxicity and its underlying toxic mechanisms remain incompletely understood. In this study, a novel 3D intestinal organoid model was utilized to investigate that exposure to TCS/TCC led to a compromised self-renewal and differentiation of intestinal stem cells (ISCs). Consequently, this disrupted intestinal epithelial homeostasis ultimately caused a reduction in nutrient absorption and deficient of epithelial defense to exogenous and endogenous pathogens stimulation. The inhibition of the Wnt signaling pathway in intestinal stem cell was contributed to the intestinal toxicity of TCS/TCC. These results were further confirmed in vivo with mice exposed to TCS/TCC. The findings of this study provide evidence that TCS/TCC possess the capacity to disturb the homeostasis of the intestinal epithelium, and emphasize the credibility of organoids as a valuable model for toxicological studies, as they could faithfully recapitulate in vivo phenomena.


Asunto(s)
Carbanilidas , Homeostasis , Mucosa Intestinal , Intestino Delgado , Organoides , Células Madre , Triclosán , Triclosán/toxicidad , Carbanilidas/toxicidad , Organoides/efectos de los fármacos , Animales , Homeostasis/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Células Madre/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Antiinfecciosos Locales/toxicidad , Ratones , Ratones Endogámicos C57BL , Humanos , Masculino , Diferenciación Celular/efectos de los fármacos
20.
Elife ; 132024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39320349

RESUMEN

The ß-catenin-dependent canonical Wnt signaling is pivotal in organ development, tissue homeostasis, and cancer. Here, we identified an upstream enhancer of Ctnnb1 - the coding gene for ß-catenin, named ieCtnnb1 (intestinal enhancer of Ctnnb1), which is crucial for intestinal homeostasis. ieCtnnb1 is predominantly active in the base of small intestinal crypts and throughout the epithelia of large intestine. Knockout of ieCtnnb1 led to a reduction in Ctnnb1 transcription, compromising the canonical Wnt signaling in intestinal crypts. Single-cell sequencing revealed that ieCtnnb1 knockout altered epithelial compositions and potentially compromised functions of small intestinal crypts. While deletion of ieCtnnb1 hampered epithelial turnovers in physiologic conditions, it prevented occurrence and progression of Wnt/ß-catenin-driven colorectal cancers. Human ieCTNNB1 drove reporter gene expression in a pattern highly similar to mouse ieCtnnb1. ieCTNNB1 contains a single-nucleotide polymorphism associated with CTNNB1 expression levels in human gastrointestinal epithelia. The enhancer activity of ieCTNNB1 in colorectal cancer tissues was stronger than that in adjacent normal tissues. HNF4α and phosphorylated CREB1 were identified as key trans-factors binding to ieCTNNB1 and regulating CTNNB1 transcription. Together, these findings unveil an enhancer-dependent mechanism controlling the dosage of Wnt signaling and homeostasis in intestinal epithelia.


Asunto(s)
Homeostasis , Mucosa Intestinal , Vía de Señalización Wnt , beta Catenina , beta Catenina/metabolismo , beta Catenina/genética , Mucosa Intestinal/metabolismo , Animales , Humanos , Ratones , Carcinogénesis/genética , Elementos de Facilitación Genéticos/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda