Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.315
Filtrar
1.
J Nanobiotechnology ; 22(1): 331, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38867284

RESUMO

BACKGROUND: In the context of increasing exposure to silica nanoparticles (SiNPs) and ensuing respiratory health risks, emerging evidence has suggested that SiNPs can cause a series of pathological lung injuries, including fibrotic lesions. However, the underlying mediators in the lung fibrogenesis caused by SiNPs have not yet been elucidated. RESULTS: The in vivo investigation verified that long-term inhalation exposure to SiNPs induced fibroblast activation and collagen deposition in the rat lungs. In vitro, the uptake of exosomes derived from SiNPs-stimulated lung epithelial cells (BEAS-2B) by fibroblasts (MRC-5) enhanced its proliferation, adhesion, and activation. In particular, the mechanistic investigation revealed SiNPs stimulated an increase of epithelium-secreted exosomal miR-494-3p and thereby disrupted the TGF-ß/BMPR2/Smad pathway in fibroblasts via targeting bone morphogenetic protein receptor 2 (BMPR2), ultimately resulting in fibroblast activation and collagen deposition. Conversely, the inhibitor of exosomes, GW4869, can abolish the induction of upregulated miR-494-3p and fibroblast activation in MRC-5 cells by the SiNPs-treated supernatants of BEAS-2B. Besides, inhibiting miR-494-3p or overexpression of BMPR2 could ameliorate fibroblast activation by interfering with the TGF-ß/BMPR2/Smad pathway. CONCLUSIONS: Our data suggested pulmonary epithelium-derived exosomes serve an essential role in fibroblast activation and collagen deposition in the lungs upon SiNPs stimuli, in particular, attributing to exosomal miR-494-3p targeting BMPR2 to modulate TGF-ß/BMPR2/Smad pathway. Hence, strategies targeting exosomes could be a new avenue in developing therapeutics against lung injury elicited by SiNPs.


Assuntos
Colágeno , Epigênese Genética , Exossomos , Fibroblastos , Pulmão , MicroRNAs , Nanopartículas , Transdução de Sinais , Dióxido de Silício , Fator de Crescimento Transformador beta , Exossomos/metabolismo , Animais , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Dióxido de Silício/química , Transdução de Sinais/efeitos dos fármacos , Ratos , Pulmão/metabolismo , Pulmão/patologia , Colágeno/metabolismo , Humanos , Nanopartículas/química , MicroRNAs/metabolismo , MicroRNAs/genética , Linhagem Celular , Fator de Crescimento Transformador beta/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/induzido quimicamente , Masculino , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Ratos Sprague-Dawley , Epitélio/metabolismo , Epitélio/efeitos dos fármacos
2.
Sci Rep ; 14(1): 12744, 2024 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-38830931

RESUMO

Transforming growth factor ß (TGF-ß) is implicated in both mesothelial-to-mesenchymal transition (MMT) and cellular senescence of human peritoneal mesothelial cells (HPMCs). We previously showed that senescent HPMCs could spontaneously acquire some phenotypic features of MMT, which in young HPMCs were induced by TGF-ß. Here, we used electron microscopy, as well as global gene and protein profiling to assess in detail how exposure to TGF-ß impacts on young and senescent HPMCs in vitro. We found that TGF-ß induced structural changes consistent with MMT in young, but not in senescent HPMCs. Of all genes and proteins identified reliably in HPMCs across all treatments and states, 4,656 targets represented overlapping genes and proteins. Following exposure to TGF-ß, 137 proteins and 46 transcripts were significantly changed in young cells, compared to 225 proteins and only 2 transcripts in senescent cells. Identified differences between young and senescent HPMCs were related predominantly to wound healing, integrin-mediated signalling, production of proteases and extracellular matrix components, and cytoskeleton structure. Thus, the response of senescent HPMCs to TGF-ß differs or is less pronounced compared to young cells. As a result, the character and magnitude of the postulated contribution of HPMCs to TGF-ß-induced peritoneal remodelling may change with cell senescence.


Assuntos
Senescência Celular , Células Epiteliais , Peritônio , Fator de Crescimento Transformador beta , Humanos , Senescência Celular/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Peritônio/citologia , Peritônio/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células Cultivadas , Epitélio/metabolismo , Epitélio/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Perfilação da Expressão Gênica
3.
Cell ; 187(12): 2898-2900, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38848672

RESUMO

Epithelial folding is a fundamental biological process that requires epithelial interactions with the underlying mesenchyme. In this issue of Cell, Huycke et al. investigate intestinal villus formation. They discover that water-droplet-like behavior of mesenchymal cells drives their coalescence into uniformly patterned aggregates, which generate forces on the epithelium to initiate folding.


Assuntos
Epitélio , Mesoderma , Animais , Humanos , Células Epiteliais/metabolismo , Células Epiteliais/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citologia , Mesoderma/metabolismo , Mesoderma/citologia , Epitélio/metabolismo
4.
Int J Mol Sci ; 25(11)2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38891804

RESUMO

The periodontium comprising periodontal ligament (PDL), gingiva, and epithelium play crucial roles in maintaining tooth integrity and function. Understanding tissue cellular composition and gene expression is crucial for illuminating periodontal pathophysiology. This study aimed to identify tissue-specific markers via scRNA-Seq. Primary human PDL, gingiva, and epithelium tissues (n = 7) were subjected to cell hashing and sorting. scRNA-Seq library preparation using 10× Genomics protocol and Illumina sequencing was conducted. The analysis was performed using Cellranger (v3.1.0), with downstream analysis via R packages Seurat (v5.0.1) and SCORPIUS (v1.0.9). Investigations identified eight distinct cellular clusters, revealing the ubiquitous presence of epithelial and gingival cells. PDL cells evolved in two clusters with numerical superiority. The other clusters showed varied predominance regarding gingival and epithelial cells or an equitable distribution of both. The cluster harboring most cells mainly consisted of PDL cells and was present in all donors. Some of the other clusters were also tissue-inherent, while the presence of others was environmentally influenced, revealing variability across donors. Two clusters exhibited genetic profiles associated with tissue development and cellular integrity, respectively, while all other clusters were distinguished by genes characteristic of immune responses. Developmental trajectory analysis uncovered that PDL cells may develop after epithelial and gingival cells, suggesting the inherent PDL cell-dominated cluster as a final developmental stage. This single-cell RNA sequencing study delineates the hierarchical organization of periodontal tissue development, identifies tissue-specific markers, and reveals the influence of environmental factors on cellular composition, advancing our understanding of periodontal biology and offering potential insights for therapeutic interventions.


Assuntos
Gengiva , Ligamento Periodontal , Análise de Célula Única , Transcriptoma , Humanos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/citologia , Gengiva/metabolismo , Análise de Célula Única/métodos , Análise de Sequência de RNA/métodos , Perfilação da Expressão Gênica , Epitélio/metabolismo , Células Epiteliais/metabolismo , Feminino , Masculino
5.
Life Sci ; 348: 122695, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38710285

RESUMO

AIMS: To evaluate the basal release of 6-nitrodopamine (6-ND) from human isolated seminal vesicles (HISV) and to characterize its action and origin. MAIN METHODS: Left HISV obtained from patients undergoing prostatectomy surgery was suspended in a 3-mL organ bath containing warmed (37 °C) and gassed (95%O2:5%CO2) Krebs-Henseleit's solution (KHS) with ascorbic acid. An aliquot of 2 mL of the supernatant was used to quantify catecholamines by LC-MS/MS. For functional studies, concentration-responses curves to catecholamines were obtained, and pEC50 and Emax values were calculated. Detection of tyrosine hydroxylase and S100 protein were also carried out by both immunohistochemistry and fluorescence in-situ hybridization assays (FISH). KEY FINDINGS: Basal release of 6-ND was higher than the other catecholamines (14.76 ± 14.54, 4.99 ± 6.92, 3.72 ± 4.35 and 5.13 ± 5.76 nM for 6-ND, noradrenaline, adrenaline, and dopamine, respectively). In contrast to the other catecholamines, the basal release of 6-ND was not affected by the sodium current (Nav) channel inhibitor tetrodotoxin (1 µM; 10.4 ± 8.9 and 10.4 ± 7.9 nM, before and after tetrodotoxin, respectively). All the catecholamines produced concentration-dependent HISV contractions (pEC50 4.1 ± 0.2, 4.9 ± 0.3, 5.0 ± 0.3, and 3.9 ± 0.8 for 6-ND, noradrenaline, adrenaline, and dopamine, respectively), but 6-ND was 10-times less potent than noradrenaline and adrenaline. However, preincubation with very low concentration of 6-ND (10-8 M, 30 min) produced significant leftward shifts of the concentration-response curves to noradrenaline. Immunohistochemical and FISH assays identified tyrosine hydroxylase in tissue epithelium of HISV strips. SIGNIFICANCE: Epithelium-derived 6-ND is the major catecholamine released from human isolated seminal vesicles and that modulates smooth muscle contractility by potentiating noradrenaline-induced contractions.


Assuntos
Dopamina , Norepinefrina , Glândulas Seminais , Humanos , Masculino , Norepinefrina/farmacologia , Norepinefrina/metabolismo , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/metabolismo , Dopamina/metabolismo , Dopamina/farmacologia , Pessoa de Meia-Idade , Epitélio/metabolismo , Epitélio/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Idoso , Catecolaminas/metabolismo
6.
Sci Rep ; 14(1): 10678, 2024 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724551

RESUMO

Mutations in LRBA, a BEACH domain protein, cause severe immune deficiency in humans. LRBA is expressed in many tissues and organs according to biochemical analysis, but little is known about its cellular and subcellular localization, and its deficiency phenotype outside the immune system. By LacZ histochemistry of Lrba gene-trap mice, we performed a comprehensive survey of LRBA expression in numerous tissues, detecting it in many if not all epithelia, in exocrine and endocrine cells, and in subpopulations of neurons. Immunofluorescence microscopy of the exocrine and endocrine pancreas, salivary glands, and intestinal segments, confirmed these patterns of cellular expression and provided information on the subcellular localizations of the LRBA protein. Immuno-electron microscopy demonstrated that in neurons and endocrine cells, which co-express LRBA and its closest relative, neurobeachin, both proteins display partial association with endomembranes in complementary, rather than overlapping, subcellular distributions. Prominent manifestations of human LRBA deficiency, such as inflammatory bowel disease or endocrinopathies, are believed to be primarily due to immune dysregulation. However, as essentially all affected tissues also express LRBA, it is possible that LRBA deficiency enhances their vulnerability and contributes to the pathogenesis.


Assuntos
Glândulas Endócrinas , Epitélio , Glândulas Exócrinas , Síndromes de Imunodeficiência , Neurônios , Animais , Humanos , Camundongos , Glândulas Endócrinas/metabolismo , Epitélio/metabolismo , Glândulas Exócrinas/metabolismo , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/patologia , Mutação , Neurônios/metabolismo
7.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38775023

RESUMO

Regenerative ability often declines as animals mature past embryonic and juvenile stages, suggesting that regeneration requires redirection of growth pathways that promote developmental growth. Intriguingly, the Drosophila larval epithelia require the hormone ecdysone (Ec) for growth but require a drop in circulating Ec levels to regenerate. Examining Ec dynamics more closely, we find that transcriptional activity of the Ec-receptor (EcR) drops in uninjured regions of wing discs, but simultaneously rises in cells around the injury-induced blastema. In parallel, blastema depletion of genes encoding Ec biosynthesis enzymes blocks EcR activity and impairs regeneration but has no effect on uninjured wings. We find that local Ec/EcR signaling is required for injury-induced pupariation delay following injury and that key regeneration regulators upd3 and Ets21c respond to Ec levels. Collectively, these data indicate that injury induces a local source of Ec within the wing blastema that sustains a transcriptional signature necessary for developmental delay and tissue repair.


Assuntos
Proteínas de Drosophila , Ecdisona , Regeneração , Asas de Animais , Animais , Ecdisona/metabolismo , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Larva/metabolismo , Larva/crescimento & desenvolvimento , Transdução de Sinais , Drosophila , Receptores de Esteroides/metabolismo , Receptores de Esteroides/genética
8.
Pathol Res Pract ; 258: 155320, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38728794

RESUMO

The objective of this study to examine the effects of curcumin and gallic acid use against oxidative stress damage in the autologous intraperitoneal ovarian transplantation model created in rats on ovarian follicle reserve, ovarian surface epithelium, and oxidant-antioxidant systems. 42 adult female Sprague Dawley rats (n=7) were allocated into 6 groups. Group 1 served as the control. In Group 2, rats underwent ovarian transplantation (TR) to their peritoneal walls. Group 3 received corn oil (CO) (0.5 ml/day) one day before and 14 days after transplantation. Group 4 was administered curcumin (CUR) (100 mg/kg/day), Group 5 received gallic acid (GA) (20 mg/kg/day), and Group 6 was treated with a combination of curcumin and gallic acid via oral gavage after transplantation. Rats were sacrificed on the 14th postoperative day, and blood along with ovaries were collected for analysis. The removed ovaries were analyzed at light microscopic, fluorescence microscopic, and biochemical levels. In Group 2 and Group 3, while serum and tissue Total Oxidant Levels (TOS) and Oxidative Stress Index (OSI) increased, serum Total Antioxidant Levels (TAS) decreased statistically significantly (p˂0.05) compared to the other groups (Groups 1, 4, 5, and 6). The ovarian follicle reserve was preserved and the changes in the ovarian surface epithelium and histopathological findings were reduced in the antioxidant-treated groups (Groups 4, 5, and 6). In addition, immunofluorescence examination revealed that the expression of Cytochrome C and Caspase 3 was stronger and Ki-67 was weaker in Groups 2 and 3, in comparison to the groups that were given antioxidants. It can be said that curcumin and gallic acid have a histological and biochemical protective effect against ischemia-reperfusion injury due to ovarian transplantation, and this effect is stronger when these two antioxidants are applied together compared to individual use.


Assuntos
Antioxidantes , Curcumina , Ácido Gálico , Folículo Ovariano , Reserva Ovariana , Ovário , Estresse Oxidativo , Ratos Sprague-Dawley , Animais , Feminino , Ácido Gálico/farmacologia , Curcumina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/patologia , Ovário/metabolismo , Ratos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Reserva Ovariana/efeitos dos fármacos , Antioxidantes/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/patologia , Epitélio/metabolismo , Transplante Autólogo , Sinergismo Farmacológico
9.
Int J Mol Sci ; 25(9)2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38732189

RESUMO

Sjögren's Disease (SjD) is an autoimmune disease of the exocrine tissues. Etiological events result in the loss of epithelial homeostasis alongside extracellular matrix (ECM) destruction within the salivary and lacrimal glands, followed by immune cell infiltration. In this review, we have assessed the current understanding of epithelial-mesenchymal transition (EMT)-associated changes within the salivary epithelium potentially involved in salivary dysfunction and SjD pathogenesis. We performed a PubMed literature review pertaining to the determination of pathogenic events that lead to EMT-related epithelial dysfunction and signaling in SjD. Molecular patterns of epithelial dysfunction in SjD salivary glands share commonalities with EMT mediating wound healing. Pathological changes altering salivary gland integrity and function may precede direct immune involvement while perpetuating MMP9-mediated ECM destruction, inflammatory mediator expression, and eventual immune cell infiltration. Dysregulation of EMT-associated factors is present in the salivary epithelium of SjD and may be significant in initiating and perpetuating the disease. In this review, we further highlight the gap regarding mechanisms that drive epithelial dysfunction in salivary glands in the early or subclinical pre-lymphocytic infiltration stages of SjD.


Assuntos
Transição Epitelial-Mesenquimal , Glândulas Salivares , Síndrome de Sjogren , Humanos , Síndrome de Sjogren/patologia , Síndrome de Sjogren/metabolismo , Glândulas Salivares/patologia , Glândulas Salivares/metabolismo , Animais , Epitélio/patologia , Epitélio/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transdução de Sinais , Matriz Extracelular/metabolismo
10.
J Photochem Photobiol B ; 255: 112908, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663336

RESUMO

The prevalence of Light-emitting diodes (LEDs) has exposed us to an excessive amount of blue light (BL) which causes various ophthalmic diseases. Previous studies have shown that conjunctiva is vulnerable to BL. In this study, we aimed to investigate the underlying mechanism of BL-induced injury in conjunctiva. We placed C57BL/6 mice and human conjunctival epithelial cell lines (HCECs) under BL (440 nm ± 15 nm, 0.2 mW/cm2) to establish a BL injury model in vivo and in vitro. Immunohistochemistry and MDA assay were used to identify lipid peroxidation (LPO) in vivo. HE staining was applied to detect morphological damage of conjunctival epithelium. DCFH-DA, C11-BODIPY 581/591, Calcein-AM, and FeRhoNox™-1 probes were performed to identify ferroptosis levels in vitro. Real-time qPCR and Western blotting techniques were employed to uncover signaling pathways of blue light-induced ferroptosis. Our findings demonstrated that BL affected tear film instability and induced conjunctival epithelium injury in vivo. Ferrostatin-1 significantly alleviated blue light-induced ferroptosis in vivo and in vitro. BL downregulates the levels of solute carrier family 7 member 11 (SLC7A11), Ferritin heavy chain (FTH1), and glutathione peroxidase (GPX4) by inhibiting the activation and translocation of the Signal transducer and activator of transcription 3 (STAT3) from inducing Fe2+ burst, ROS and LPO accumulation, ultimately resulting in ferroptosis. This study will offer new insight into BL-induced conjunctival injury and LED-induced dry eye.


Assuntos
Túnica Conjuntiva , Ferroptose , Luz , Camundongos Endogâmicos C57BL , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Fator de Transcrição STAT3 , Animais , Túnica Conjuntiva/metabolismo , Túnica Conjuntiva/efeitos da radiação , Túnica Conjuntiva/patologia , Camundongos , Ferroptose/efeitos da radiação , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Humanos , Fator de Transcrição STAT3/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Peroxidação de Lipídeos/efeitos da radiação , Linhagem Celular , Epitélio/efeitos da radiação , Epitélio/metabolismo , Epitélio/patologia , Transdução de Sinais/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Células Epiteliais/patologia , Espécies Reativas de Oxigênio/metabolismo , Fenilenodiaminas/farmacologia , Luz Azul , Cicloexilaminas
11.
Cell Rep Methods ; 4(4): 100741, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38569541

RESUMO

Deep proteomic profiling of rare cell populations has been constrained by sample input requirements. Here, we present DROPPS (droplet-based one-pot preparation for proteomic samples), an accessible low-input platform that generates high-fidelity proteomic profiles of 100-2,500 cells. By applying DROPPS within the mammary epithelium, we elucidated the connection between mitochondrial activity and clonogenicity, identifying CD36 as a marker of progenitor capacity in the basal cell compartment. We anticipate that DROPPS will accelerate biology-driven proteomic research for a multitude of rare cell populations.


Assuntos
Biomarcadores , Antígenos CD36 , Glândulas Mamárias Animais , Proteômica , Células-Tronco , Proteômica/métodos , Antígenos CD36/metabolismo , Animais , Feminino , Células-Tronco/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Biomarcadores/metabolismo , Biomarcadores/análise , Epitélio/metabolismo , Camundongos , Humanos , Mitocôndrias/metabolismo
12.
Am J Pathol ; 194(7): 1294-1305, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38657836

RESUMO

Mesothelial cells with reactive hyperplasia are difficult to distinguish from malignant mesothelioma cells based on cell morphology. This study aimed to identify and validate potential biomarkers that distinguish mesothelial cells from mesothelioma cells through machine learning combined with immunohistochemistry. It integrated the gene expression matrix from three Gene Expression Omnibus data sets (GSE2549, GSE12345, and GSE51024) to analyze the differently expressed genes between normal and mesothelioma tissues. Then, three machine learning algorithms, least absolute shrinkage and selection operator, support vector machine recursive feature elimination, and random forest were used to screen and obtain four shared candidate markers, including ACADL, EMP2, GPD1L, and HMMR. The receiver operating characteristic curve analysis showed that the area under the curve for distinguishing normal mesothelial cells from mesothelioma was 0.976, 0.943, 0.962, and 0.956, respectively. The expression and diagnostic performance of these candidate genes were validated in two additional independent data sets (GSE42977 and GSE112154), indicating that the performances of ACADL, GPD1L, and HMMR were consistent between the training and validation data sets. Finally, the optimal candidate marker ACADL was verified by immunohistochemistry assay. Acyl-CoA dehydrogenase long chain (ACADL) was stained strongly in mesothelial cells, especially for reactive hyperplasic mesothelial cells, but was negative in malignant mesothelioma cells. Therefore, ACADL has the potential to be used as a specific marker of reactive hyperplasic mesothelial cells in the differential diagnosis of mesothelioma.


Assuntos
Biomarcadores Tumorais , Biologia Computacional , Aprendizado de Máquina , Mesotelioma Maligno , Mesotelioma , Humanos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Mesotelioma/genética , Mesotelioma/patologia , Mesotelioma/diagnóstico , Mesotelioma/metabolismo , Biologia Computacional/métodos , Mesotelioma Maligno/genética , Mesotelioma Maligno/patologia , Mesotelioma Maligno/metabolismo , Mesotelioma Maligno/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/diagnóstico , Epitélio/metabolismo , Epitélio/patologia
13.
Dev Cell ; 59(10): 1269-1283.e6, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38565145

RESUMO

Progenitor cells adapt their behavior in response to tissue demands. However, the molecular mechanisms controlling esophageal progenitor decisions remain largely unknown. Here, we demonstrate the presence of a Troy (Tnfrsf19)-expressing progenitor subpopulation localized to defined regions along the mouse esophageal axis. Lineage tracing and mathematical modeling demonstrate that Troy-positive progenitor cells are prone to undergoing symmetrical fate choices and contribute to esophageal tissue homeostasis long term. Functionally, TROY inhibits progenitor proliferation and enables commitment to differentiation without affecting fate symmetry. Whereas Troy expression is stable during esophageal homeostasis, progenitor cells downregulate Troy in response to tissue stress, enabling proliferative expansion of basal cells refractory to differentiation and reestablishment of tissue homeostasis. Our results demonstrate functional, spatially restricted progenitor heterogeneity in the esophageal epithelium and identify how dynamic regulation of Troy coordinates tissue generation.


Assuntos
Diferenciação Celular , Proliferação de Células , Esôfago , Receptores do Fator de Necrose Tumoral , Células-Tronco , Animais , Camundongos , Linhagem da Célula , Epitélio/metabolismo , Mucosa Esofágica/metabolismo , Mucosa Esofágica/citologia , Esôfago/citologia , Esôfago/metabolismo , Proteínas de Homeodomínio , Homeostase , Células-Tronco/metabolismo , Células-Tronco/citologia , Receptores do Fator de Necrose Tumoral/análise , Receptores do Fator de Necrose Tumoral/metabolismo
14.
Epigenetics Chromatin ; 17(1): 10, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643244

RESUMO

BACKGROUND: Nuclear organization of interphase chromosomes involves individual chromosome territories, "open" and "closed" chromatin compartments, topologically associated domains (TADs) and chromatin loops. The DNA- and RNA-binding transcription factor CTCF together with the cohesin complex serve as major organizers of chromatin architecture. Cellular differentiation is driven by temporally and spatially coordinated gene expression that requires chromatin changes of individual loci of various complexities. Lens differentiation represents an advantageous system to probe transcriptional mechanisms underlying tissue-specific gene expression including high transcriptional outputs of individual crystallin genes until the mature lens fiber cells degrade their nuclei. RESULTS: Chromatin organization between mouse embryonic stem (ES) cells, newborn (P0.5) lens epithelium and fiber cells were analyzed using Hi-C. Localization of CTCF in both lens chromatins was determined by ChIP-seq and compared with ES cells. Quantitative analyses show major differences between number and size of TADs and chromatin loop size between these three cell types. In depth analyses show similarities between lens samples exemplified by overlaps between compartments A and B. Lens epithelium-specific CTCF peaks are found in mostly methylated genomic regions while lens fiber-specific and shared peaks occur mostly within unmethylated DNA regions. Major differences in TADs and loops are illustrated at the ~ 500 kb Pax6 locus, encoding the critical lens regulatory transcription factor and within a larger ~ 15 Mb WAGR locus, containing Pax6 and other loci linked to human congenital diseases. Lens and ES cell Hi-C data (TADs and loops) together with ATAC-seq, CTCF, H3K27ac, H3K27me3 and ENCODE cis-regulatory sites are shown in detail for the Pax6, Sox1 and Hif1a loci, multiple crystallin genes and other important loci required for lens morphogenesis. The majority of crystallin loci are marked by unexpectedly high CTCF-binding across their transcribed regions. CONCLUSIONS: Our study has generated the first data on 3-dimensional (3D) nuclear organization in lens epithelium and lens fibers and directly compared these data with ES cells. These findings generate novel insights into lens-specific transcriptional gene control, open new research avenues to study transcriptional condensates in lens fiber cells, and enable studies of non-coding genetic variants linked to cataract and other lens and ocular abnormalities.


Assuntos
Cromatina , Cristalinas , Animais , Camundongos , Humanos , Células-Tronco Embrionárias Murinas/metabolismo , Cromossomos/metabolismo , Fatores de Transcrição/metabolismo , DNA/metabolismo , Epitélio/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Fator de Ligação a CCCTC/metabolismo
15.
Hear Res ; 446: 109006, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38583350

RESUMO

Hair cells in the cochlear sensory epithelia serve as mechanosensory receptors, converting sound into neuronal signals. The basal sensory epithelia are responsible for transducing high-frequency sounds, while the apex handles low-frequency sounds. Age-related hearing loss predominantly affects hearing at high frequencies and is indicative of damage to the basal sensory epithelia. However, the precise mechanism underlying this site-selective injury remains unclear. In this study, we employed a microscale proteomics approach to examine and compare protein expression in different regions of the cochlear sensory epithelia (upper half and lower half) in 1.5-month-old (normal hearing) and 6-month-old (severe high-frequency hearing loss without hair cell loss) C57BL/6J mice. A total of 2,386 proteins were detected, and no significant differences in protein expression were detected in the upper half of the cochlear sensory epithelia between the two age groups. The expression of 20 proteins in the lower half of the cochlear sensory epithelia significantly differed between the two age groups (e.g., MATN1, MATN4, and AQP1). Moreover, there were 311 and 226 differentially expressed proteins between the upper and lower halves of the cochlear sensory epithelia in 1.5-month-old and 6-month-old mice, respectively. The expression levels of selected proteins were validated by Western blotting. These findings suggest that the spatial differences in protein expression within the cochlear sensory epithelia may play a role in determining the susceptibility of cells at different sites of the cochlea to age-related damage.


Assuntos
Cóclea , Camundongos Endogâmicos C57BL , Presbiacusia , Proteômica , Animais , Cóclea/metabolismo , Cóclea/patologia , Presbiacusia/metabolismo , Presbiacusia/patologia , Presbiacusia/fisiopatologia , Presbiacusia/genética , Fatores Etários , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Modelos Animais de Doenças , Audição , Epitélio/metabolismo , Masculino , Camundongos
16.
Sci Adv ; 10(14): eadj7666, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38569041

RESUMO

Inflammation-associated fibroblasts (IAFs) are associated with progression and drug resistance of chronic inflammatory diseases such as inflammatory bowel disease (IBD), but their direct impact on epithelial cells is unknown. Here, we developed an in vitro model whereby human colon fibroblasts are induced by specific cytokines and recapitulate key features of IAFs in vivo. When cocultured with patient-derived colon organoids (colonoids), IAFs induced rapid colonoid expansion and barrier disruption due to swelling and rupture of individual epithelial cells. Colonoids cocultured with IAFs also show increased DNA damage, mitotic errors, and proliferation arrest. These IAF-induced epithelial defects are mediated by a paracrine pathway involving prostaglandin E2 and its receptor EP4, leading to protein kinase A -dependent activation of the cystic fibrosis transmembrane conductance regulator. EP4-specific chemical inhibitors effectively prevented IAF-induced colonoid swelling and restored normal proliferation and genome stability. These findings reveal a mechanism by which IAFs could promote and perpetuate IBD and suggest a therapeutic avenue to mitigate inflammation-associated epithelial injury.


Assuntos
Doenças Inflamatórias Intestinais , Prostaglandinas , Humanos , Epitélio/metabolismo , Inflamação , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Fibroblastos/metabolismo
17.
Nat Commun ; 15(1): 3000, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589403

RESUMO

Actomyosin networks constrict cell area and junctions to alter cell and tissue shape. However, during cell expansion under mechanical stress, actomyosin networks are strengthened and polarized to relax stress. Thus, cells face a conflicting situation between the enhanced actomyosin contractile properties and the expansion behaviour of the cell or tissue. To address this paradoxical situation, we study late Drosophila oogenesis and reveal an unusual epithelial expansion wave behaviour. Mechanistically, Rac1 and Rho1 integrate basal pulsatile actomyosin networks with ruffles and focal adhesions to increase and then stabilize basal area of epithelial cells allowing their flattening and elongation. This epithelial expansion behaviour bridges cell changes to oocyte growth and extension, while oocyte growth in turn deforms the epithelium to drive cell spreading. Basal pulsatile actomyosin networks exhibit non-contractile mechanics, non-linear structures and F-actin/Myosin-II spatiotemporal signal separation, implicating unreported expanding properties. Biophysical modelling incorporating these expanding properties well simulates epithelial cell expansion waves. Our work thus highlights actomyosin expanding properties as a key mechanism driving tissue morphogenesis.


Assuntos
Actomiosina , Proteínas de Drosophila , Animais , Actomiosina/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliais/metabolismo , Citoesqueleto de Actina/metabolismo , Drosophila/metabolismo , Epitélio/metabolismo , Morfogênese
18.
Int J Dev Biol ; 68(1): 39-45, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38591692

RESUMO

Keratin 17 (K17) is thought to be a candidate target gene for regulation by Lymphoid Enhancer Factor-1 (Lef-1). K17 is a marker that distinguishes junctional epithelium (JE) from epithelial rests of Malassez (ERM). However, the relationship of Lef-1 to K17 is not clear in this context. Moreover, the expression of other keratins such as K5, K6, K7 and K16 is not reported. Therefore, the aim of our study was to assay the expression of K5, K6, K7, K14, K16, K17 and Lef-1 in postnatal developing teeth, and clarify the corresponding immunophenotypes of the JE and ERM. Upper jaws of Wistar rats aged from postnatal (PN) day 3.5 to PN21 were used and processed for immunohistochemistry. K5 and K14 were intensely expressed in inner enamel epithelium (IEE), reduced enamel epithelium (REE), ERM and JE. There was no staining for K16 in the tissue, except for strong staining in the oral epithelium. Specifically, at PN3.5 and PN7, K17 was initially strongly expressed and then negative in the IEE. At PN16 and PN21, both REE and ERM were strongly stained for K17, whereas K17 was negative in the JE. In addition, K6, K7 and Lef-1 were not detected in any tissue investigated. REE and ERM have an identical keratin expression pattern before eruption, while JE differs from ERM in the expression of K17 after eruption. The expression of K17 does not coincide with that of Lef-1. These data indicate that JE has a unique phenotype different from ERM, which is of odontogenic origin.


Assuntos
Inserção Epitelial , Descanso , Ratos , Animais , Inserção Epitelial/metabolismo , Ratos Wistar , Epitélio/metabolismo , Imuno-Histoquímica , Queratinas/metabolismo
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167161, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38599260

RESUMO

The tongue epithelium is maintained by a proliferative basal layer. This layer contains long-lived stem cells (SCs), which produce progeny cells that move up to the surface as they differentiate. B-lymphoma Mo-MLV insertion region 1 (BMI1), a protein in mammalian Polycomb Repressive Complex 1 (PRC1) and a biomarker of oral squamous cell carcinoma, is expressed in almost all basal epithelial SCs of the tongue, and single, Bmi1-labelled SCs give rise to cells in all epithelial layers. We previously developed a transgenic mouse model (KrTB) containing a doxycycline- (dox) controlled, Tet-responsive element system to selectively overexpress Bmi1 in the tongue basal epithelial SCs. Here, we used this model to assess BMI1 actions in tongue epithelia. Genome-wide transcriptomics revealed increased levels of transcripts involved in the cellular response to hypoxia in Bmi1-overexpressing (KrTB+DOX) oral epithelia even though these mice were not subjected to hypoxia conditions. Ectopic Bmi1 expression in tongue epithelia increased the levels of hypoxia inducible factor-1 alpha (HIF1α) and HIF1α targets linked to metabolic reprogramming during hypoxia. We used chromatin immunoprecipitation (ChIP) to demonstrate that Bmi1 associates with the promoters of HIF1A and HIF1A-activator RELA (p65) in tongue epithelia. We also detected increased SC proliferation and oxidative stress in Bmi1-overexpressing tongue epithelia. Finally, using a human oral keratinocyte line (OKF6-TERT1R), we showed that ectopic BMI1 overexpression decreases the oxygen consumption rate while increasing the extracellular acidification rate, indicative of elevated glycolysis. Thus, our data demonstrate that high BMI1 expression drives hypoxic signaling, including metabolic reprogramming, in normal oral cavity epithelia.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos Transgênicos , Complexo Repressor Polycomb 1 , Transdução de Sinais , Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 1/genética , Animais , Camundongos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Humanos , Língua/metabolismo , Língua/patologia , Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , Hipóxia Celular , Epitélio/metabolismo , Boca/metabolismo , Boca/patologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Neoplasias Bucais/genética , Proteínas Proto-Oncogênicas
20.
FEBS Lett ; 598(11): 1335-1353, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38485451

RESUMO

Our epithelium represents a battle ground against a variety of insults including pathogens and danger signals. It encodes multiple sensors that detect and respond to such insults, playing an essential role in maintaining and defending tissue homeostasis. One key set of defense mechanisms is our inflammasomes which drive innate immune responses including, sensing and responding to pathogen attack, through the secretion of pro-inflammatory cytokines and cell death. Identification of physiologically relevant triggers for inflammasomes has greatly influenced our ability to decipher the mechanisms behind inflammasome activation. Furthermore, identification of patient mutations within inflammasome components implicates their involvement in a range of epithelial diseases. This review will focus on exploring the roles of inflammasomes in epithelial immunity and cover: the diversity and differential expression of inflammasome sensors amongst our epithelial barriers, their ability to sense local infection and damage and the contribution of the inflammasomes to epithelial homeostasis and disease.


Assuntos
Imunidade Inata , Inflamassomos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Humanos , Animais , Epitélio/imunologia , Epitélio/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Homeostase/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...