Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 578(7795): 437-443, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025032

RESUMEN

LGR5 marks resident adult epithelial stem cells at the gland base in the mouse pyloric stomach1, but the identity of the equivalent human stem cell population remains unknown owing to a lack of surface markers that facilitate its prospective isolation and validation. In mouse models of intestinal cancer, LGR5+ intestinal stem cells are major sources of cancer following hyperactivation of the WNT pathway2. However, the contribution of pyloric LGR5+ stem cells to gastric cancer following dysregulation of the WNT pathway-a frequent event in gastric cancer in humans3-is unknown. Here we use comparative profiling of LGR5+ stem cell populations along the mouse gastrointestinal tract to identify, and then functionally validate, the membrane protein AQP5 as a marker that enriches for mouse and human adult pyloric stem cells. We show that stem cells within the AQP5+ compartment are a source of WNT-driven, invasive gastric cancer in vivo, using newly generated Aqp5-creERT2 mouse models. Additionally, tumour-resident AQP5+ cells can selectively initiate organoid growth in vitro, which indicates that this population contains potential cancer stem cells. In humans, AQP5 is frequently expressed in primary intestinal and diffuse subtypes of gastric cancer (and in metastases of these subtypes), and often displays altered cellular localization compared with healthy tissue. These newly identified markers and mouse models will be an invaluable resource for deciphering the early formation of gastric cancer, and for isolating and characterizing human-stomach stem cells as a prerequisite for harnessing the regenerative-medicine potential of these cells in the clinic.


Asunto(s)
Acuaporina 5/metabolismo , Carcinogénesis/patología , Células Madre Neoplásicas/patología , Neoplasias Gástricas/patología , Estómago/patología , Animales , Biomarcadores/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Píloro/patología , Receptores Acoplados a Proteínas G/metabolismo , Vía de Señalización Wnt
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35046017

RESUMEN

Alveolar macrophages (AMs) are critical for lung immune defense and homeostasis. They are orchestrators of chronic obstructive pulmonary disease (COPD), with their number significantly increased and functions altered in COPD. However, it is unclear how AM number and function are controlled in a healthy lung and if changes in AMs without environmental assault are sufficient to trigger lung inflammation and COPD. We report here that absence of isthmin 1 (ISM1) in mice (Ism1-/- ) leads to increase in both AM number and functional heterogeneity, with enduring lung inflammation, progressive emphysema, and significant lung function decline, phenotypes similar to human COPD. We reveal that ISM1 is a lung resident anti-inflammatory protein that selectively triggers the apoptosis of AMs that harbor high levels of its receptor cell-surface GRP78 (csGRP78). csGRP78 is present at a heterogeneous level in the AMs of a healthy lung, but csGRP78high AMs are expanded in Ism1-/- mice, cigarette smoke (CS)-induced COPD mice, and human COPD lung, making these cells the prime targets of ISM1-mediated apoptosis. We show that csGRP78high AMs mostly express MMP-12, hence proinflammatory. Intratracheal delivery of recombinant ISM1 (rISM1) depleted csGRP78high AMs in both Ism1-/- and CS-induced COPD mice, blocked emphysema development, and preserved lung function. Consistently, ISM1 expression in human lungs positively correlates with AM apoptosis, suggesting similar function of ISM1-csGRP78 in human lungs. Our findings reveal that AM apoptosis regulation is an important physiological mechanism for maintaining lung homeostasis and demonstrate the potential of pulmonary-delivered rISM1 to target csGRP78 as a therapeutic strategy for COPD.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/patología , Macrófagos Alveolares/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Apoptosis/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico/fisiología , Femenino , Homeostasis , Inflamación , Péptidos y Proteínas de Señalización Intercelular/fisiología , Pulmón/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Fagocitosis/fisiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/metabolismo , Humo/efectos adversos , Fumar/efectos adversos , Nicotiana/efectos adversos
3.
Gastric Cancer ; 27(2): 263-274, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38221567

RESUMEN

BACKGROUND: Mucosal gastric atrophy and intestinal metaplasia (IM) increase the risk for the development of gastric cancer (GC) as they represent a field for development of dysplasia and intestinal-type gastric adenocarcinoma. METHODS: We have investigated the expression of two dysplasia markers, CEACAM5 and TROP2, in human antral IM and gastric tumors to assess their potential as molecular markers. RESULTS: In the normal antral mucosa, weak CEACAM5 and TROP2 expression was only observed in the foveolar epithelium, while inflamed antrum exhibited increased expression of both markers. Complete IM exhibited weak CEACAM5 expression at the apical surface, but no basolateral TROP2 expression. On the other hand, incomplete IM demonstrated high levels of both CEACAM5 and TROP2 expression. Notably, incomplete IM with dysplastic morphology (dysplastic incomplete IM) exhibited higher levels of CEACAM5 and TROP2 expression compared to incomplete IM without dysplastic features (simple incomplete IM). In addition, dysplastic incomplete IM showed diminished SOX2 and elevated CDX2 expression compared to simple incomplete IM. CEACAM5 and TROP2 positivity in incomplete IM was similar to that of gastric adenomas and GC. Significant association was found between CEACAM5 and TROP2 positivity and histology of GC. CONCLUSIONS: These findings support the concept that incomplete IM is more likely associated with GC development. Overall, our study provides evidence of the heterogeneity of gastric IM and the distinct expression profiles of CEACAM5 and TROP2 in dysplastic incomplete IM. Our findings support the potential use of CEACAM5 and TROP2 as molecular markers for identifying individuals with a higher risk of GC development in the context of incomplete IM.


Asunto(s)
Lesiones Precancerosas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Mucosa Gástrica/patología , Lesiones Precancerosas/patología , Metaplasia , Antígeno Carcinoembrionario , Proteínas Ligadas a GPI/metabolismo
4.
Molecules ; 29(3)2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38338420

RESUMEN

Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are enzymes that break down and reduce the level of the neurotransmitter acetylcholine (ACh). This can cause a variety of cognitive and neurological problems, including Alzheimer's disease. Taxifolin is a natural phytochemical generally found in yew tree bark and has significant pharmacological properties, such as being anti-cancer, anti-inflammatory, and antioxidant. The binding affinity and inhibitory potency of taxifolin to these enzymes were evaluated through molecular docking and molecular dynamics simulations followed by the MMPBSA approach, and the results were significant. Taxifolin's affinity for binding to the AChE-taxifolin complex was -8.85 kcal/mol, with an inhibition constant of 326.70 nM. It was observed to interact through hydrogen bonds. In contrast, the BChE-taxifolin complex binding energy was observed to be -7.42 kcal/mol, and it was significantly nearly equal to the standard inhibitor donepezil. The molecular dynamics and simulation signified the observed interactions of taxifolin with the studied enzymes. The MMPBSA total free energy of binding for AChE-taxifolin was -24.34 kcal/mol, while BChE-taxifolin was -16.14 kcal/mol. The present research suggests that taxifolin has a strong ability to bind and inhibit AChE and BChE and could be used to manage neuron-associated problems; however, further research is required to explore taxifolin's neurological therapeutic potential using animal models of Alzheimer's disease.


Asunto(s)
Acetilcolinesterasa , Enfermedad de Alzheimer , Quercetina/análogos & derivados , Animales , Acetilcolinesterasa/metabolismo , Butirilcolinesterasa/química , Enfermedad de Alzheimer/tratamiento farmacológico , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Inhibidores de la Colinesterasa/química , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
5.
Gut ; 72(2): 226-241, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35817555

RESUMEN

OBJECTIVE: Gastric cancer (GC) comprises multiple molecular subtypes. Recent studies have highlighted mesenchymal-subtype GC (Mes-GC) as a clinically aggressive subtype with few treatment options. Combining multiple studies, we derived and applied a consensus Mes-GC classifier to define the Mes-GC enhancer landscape revealing disease vulnerabilities. DESIGN: Transcriptomic profiles of ~1000 primary GCs and cell lines were analysed to derive a consensus Mes-GC classifier. Clinical and genomic associations were performed across >1200 patients with GC. Genome-wide epigenomic profiles (H3K27ac, H3K4me1 and assay for transposase-accessible chromatin with sequencing (ATAC-seq)) of 49 primary GCs and GC cell lines were generated to identify Mes-GC-specific enhancer landscapes. Upstream regulators and downstream targets of Mes-GC enhancers were interrogated using chromatin immunoprecipitation followed by sequencing (ChIP-seq), RNA sequencing, CRISPR/Cas9 editing, functional assays and pharmacological inhibition. RESULTS: We identified and validated a 993-gene cancer-cell intrinsic Mes-GC classifier applicable to retrospective cohorts or prospective single samples. Multicohort analysis of Mes-GCs confirmed associations with poor patient survival, therapy resistance and few targetable genomic alterations. Analysis of enhancer profiles revealed a distinctive Mes-GC epigenomic landscape, with TEAD1 as a master regulator of Mes-GC enhancers and Mes-GCs exhibiting preferential sensitivity to TEAD1 pharmacological inhibition. Analysis of Mes-GC super-enhancers also highlighted NUAK1 kinase as a downstream target, with synergistic effects observed between NUAK1 inhibition and cisplatin treatment. CONCLUSION: Our results establish a consensus Mes-GC classifier applicable to multiple transcriptomic scenarios. Mes-GCs exhibit a distinct epigenomic landscape, and TEAD1 inhibition and combinatorial NUAK1 inhibition/cisplatin may represent potential targetable options.


Asunto(s)
Elementos de Facilitación Genéticos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Gástricas , Humanos , Cisplatino/metabolismo , Cisplatino/uso terapéutico , Estudios Prospectivos , Proteínas Quinasas/genética , Proteínas Represoras , Estudios Retrospectivos , Neoplasias Gástricas/genética
6.
Gut ; 71(5): 854-863, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33975867

RESUMEN

OBJECTIVE: To investigate the incidence of gastric cancer (GC) attributed to gastric intestinal metaplasia (IM), and validate the Operative Link on Gastric Intestinal Metaplasia (OLGIM) for targeted endoscopic surveillance in regions with low-intermediate incidence of GC. METHODS: A prospective, longitudinal and multicentre study was carried out in Singapore. The study participants comprised 2980 patients undergoing screening gastroscopy with standardised gastric mucosal sampling, from January 2004 and December 2010, with scheduled surveillance endoscopies at year 3 and 5. Participants were also matched against the National Registry of Diseases Office for missed diagnoses of early gastric neoplasia (EGN). RESULTS: There were 21 participants diagnosed with EGN. IM was a significant risk factor for EGN (adjusted-HR 5.36; 95% CI 1.51 to 19.0; p<0.01). The age-adjusted EGN incidence rates for patients with and without IM were 133.9 and 12.5 per 100 000 person-years. Participants with OLGIM stages III-IV were at greatest risk (adjusted-HR 20.7; 95% CI 5.04 to 85.6; p<0.01). More than half of the EGNs (n=4/7) attributed to baseline OLGIM III-IV developed within 2 years (range: 12.7-44.8 months). Serum trefoil factor 3 distinguishes (Area Under the Receiver Operating Characteristics 0.749) patients with OLGIM III-IV if they are negative for H. pylori. Participants with OLGIM II were also at significant risk of EGN (adjusted-HR 7.34; 95% CI 1.60 to 33.7; p=0.02). A significant smoking history further increases the risk of EGN among patients with OLGIM stages II-IV. CONCLUSIONS: We suggest a risk-stratified approach and recommend that high-risk patients (OLGIM III-IV) have endoscopic surveillance in 2 years, intermediate-risk patients (OLGIM II) in 5 years.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Lesiones Precancerosas , Neoplasias Gástricas , Gastroscopía , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/epidemiología , Humanos , Metaplasia , Lesiones Precancerosas/epidemiología , Estudios Prospectivos , Factores de Riesgo , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/etiología
7.
Gastroenterology ; 161(6): 1907-1923.e26, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34391772

RESUMEN

BACKGROUND & AIMS: Metaplasia and dysplasia in the corpus are reportedly derived from de-differentiation of chief cells. However, the cellular origin of metaplasia and cancer remained uncertain. Therefore, we investigated whether pepsinogen C (PGC) transcript-expressing cells represent the cellular origin of metaplasia and cancer using a novel Pgc-specific CreERT2 recombinase mouse model. METHODS: We generated a Pgc-mCherry-IRES-CreERT2 (Pgc-CreERT2) knock-in mouse model. Pgc-CreERT2/+ and Rosa-EYFP mice were crossed to generate Pgc-CreERT2/Rosa-EYFP (Pgc-CreERT2/YFP) mice. Gastric tissues were collected, followed by lineage-tracing experiments and histologic and immunofluorescence staining. We further established Pgc-CreERT2;KrasG12D/+ mice and investigated whether PGC transcript-expressing cells are responsible for the precancerous state in gastric glands. To investigate cancer development from PGC transcript-expressing cells with activated Kras, inactivated Apc, and Trp53 signaling pathways, we crossed Pgc-CreERT2/+ mice with conditional KrasG12D, Apcflox, Trp53flox mice. RESULTS: Expectedly, mCherry mainly labeled chief cells in the Pgc-CreERT2 mice. However, mCherry was also detected throughout the neck cell and isthmal stem/progenitor regions, albeit at lower levels. In the Pgc-CreERT2;KrasG12D/+ mice, PGC transcript-expressing cells with KrasG12D/+ mutation presented pseudopyloric metaplasia. The early induction of proliferation at the isthmus may reflect the ability of isthmal progenitors to react rapidly to Pgc-driven KrasG12D/+ oncogenic mutation. Furthermore, Pgc-CreERT2;KrasG12D/+;Apcflox/flox mice presented intramucosal dysplasia/carcinoma and Pgc-CreERT2;KrasG12D/+;Apcflox/flox;Trp53flox/flox mice presented invasive and metastatic gastric carcinoma. CONCLUSIONS: The Pgc-CreERT2 knock-in mouse is an invaluable tool to study the effects of successive oncogenic activation in the mouse corpus. Time-course observations can be made regarding the responses of isthmal and chief cells to oncogenic insults. We can observe stomach-specific tumorigenesis from the beginning to metastatic development.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/genética , Células Principales Gástricas/enzimología , Integrasas/genética , Pepsinógeno C/genética , Lesiones Precancerosas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Gástricas/genética , Activación Transcripcional , Animales , Desdiferenciación Celular , Linaje de la Célula , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Principales Gástricas/patología , Regulación Neoplásica de la Expresión Génica , Genes APC , Predisposición Genética a la Enfermedad , Integrasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Metaplasia , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Pepsinógeno C/metabolismo , Fenotipo , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína Fluorescente Roja
8.
FASEB J ; 35(2): e21290, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33475198

RESUMEN

Dysregulation of signaling pathways is responsible for many human diseases. The lack of understanding of the molecular etiology of gastric cancer (GC) poses a substantial challenge to the development of effective cancer therapy. To better understand the molecular mechanisms underlying the pathogenesis of GC, which will facilitate the identification and development of effective therapeutic approaches to improve patient outcomes, mass spectrometry-based phosphoproteomics analysis was performed to map the global molecular changes in GC. A total of 530 proteins with altered phosphorylation levels were detected across a panel of 15 normal and GC cell lines. WW domain-binding protein 2 (WBP2) was validated to be upregulated in a subset of GC cell lines. WBP2 is overexpressed in 61% cases of GC compared to non-cancer tissues and high WBP2 expression correlates with poor clinical outcomes. WBP2 was found to be required for GC cell migration but is dispensable for cell growth and proliferation. WBP2 knockdown increased p-LATS2 with a concomitant increase in p-YAP, resulting in the cytoplasmic retention of YAP and ultimately the inhibition of YAP/TEAD activity and downregulation of TEAD target genes--CTGF and CYR61. Importantly, the loss of LATS2 reversed the activation of Hippo pathway caused by WBP2 knockdown, indicating that WBP2 acts through LATS2 to exert its function on the Hippo pathway. Moreover, WBP2 interacted with LATS2 to inhibit its phosphorylation and activity. In conclusion, our study established a pivotal role for WBP2 in the promotion of GC cell migration via a novel mechanism that inactivates the Hippo pathway transducer LATS2.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Gástricas/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Humanos , Immunoblotting , Inmunohistoquímica , Proteínas Serina-Treonina Quinasas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Neoplasias Gástricas/genética , Transactivadores/genética , Proteínas Supresoras de Tumor/genética
9.
Gut ; 70(10): 1833-1846, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33293280

RESUMEN

OBJECTIVE: Tissue stem cells are central regulators of organ homoeostasis. We looked for a protein that is exclusively expressed and functionally involved in stem cell activity in rapidly proliferating isthmus stem cells in the stomach corpus. DESIGN: We uncovered the specific expression of Iqgap3 in proliferating isthmus stem cells through immunofluorescence and in situ hybridisation. We performed lineage tracing and transcriptomic analysis of Iqgap3 +isthmus stem cells with the Iqgap3-2A-tdTomato mouse model. Depletion of Iqgap3 revealed its functional importance in maintenance and proliferation of stem cells. We further studied Iqgap3 expression and the associated gene expression changes during tissue repair after tamoxifen-induced damage. Immunohistochemistry revealed elevated expression of Iqgap3 in proliferating regions of gastric tumours from patient samples. RESULTS: Iqgap3 is a highly specific marker of proliferating isthmus stem cells during homoeostasis. Iqgap3+isthmus stem cells give rise to major cell types of the corpus unit. Iqgap3 expression is essential for the maintenance of stem potential. The Ras pathway is a critical partner of Iqgap3 in promoting strong proliferation in isthmus stem cells. The robust induction of Iqgap3 expression following tissue damage indicates an active role for Iqgap3 in tissue regeneration. CONCLUSION: IQGAP3 is a major regulator of stomach epithelial tissue homoeostasis and repair. The upregulation of IQGAP3 in gastric cancer suggests that IQGAP3 plays an important role in cancer cell proliferation.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Mucosa Gástrica/citología , Homeostasis/fisiología , Células Madre/citología , Neoplasias Gástricas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neoplasias Gástricas/tratamiento farmacológico , Tamoxifeno/toxicidad
10.
Cell Tissue Res ; 383(2): 617-644, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33201351

RESUMEN

Injuries to the peripheral nervous system remain a large-scale clinical problem. These injuries often lead to loss of motor and/or sensory function that significantly affects patients' quality of life. The current neurosurgical approach for peripheral nerve repair involves autologous nerve transplantation, which often leads to clinical complications. The most pressing need is to increase the regenerative capacity of existing tubular constructs in the repair of large nerve gaps through development of tissue-engineered approaches that can surpass the performance of autografts. To fully realize the clinical potential of nerve conduit technology, there is a need to reconsider design strategies, biomaterial selection, fabrication techniques and the various potential modifications to optimize a conduit microenvironment that can best mimic the natural process of regeneration. In recent years, a significant progress has been made in the designing and functionality of bioengineered nerve conduits to bridge long peripheral nerve gaps in various animal models. However, translation of this work from lab to commercial scale has not been achieve. The current review summarizes recent advances in the development of tissue engineered nerve guidance conduits (NGCs) with regard to choice of material, novel fabrication methods, surface modifications and regenerative cues such as stem cells and growth factors to improve regeneration performance. Also, the current clinical potential and future perspectives to achieve therapeutic benefits of NGCs will be discussed in context of peripheral nerve regeneration.


Asunto(s)
Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/fisiopatología , Ingeniería de Tejidos , Andamios del Tejido/química , Investigación Biomédica Traslacional , Animales , Materiales Biocompatibles/farmacología , Humanos
11.
Gut ; 69(10): 1738-1749, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31937549

RESUMEN

OBJECTIVE: Intestinal metaplasia (IM) is a premalignant stage that poses a greater risk for subsequent gastric cancer (GC). However, factors regulating IM to GC progression remain unclear. Previously, activated DNA damage response (DDR) signalling factors were shown to engage tumour-suppressive networks in premalignant lesions. Here, we interrogate the relationship of DDR signalling to mutational accumulation in IM lesions. DESIGN: IM biopsies were procured from the gastric cancer epidemiology programme, an endoscopic surveillance programme where biopsies have been subjected to (epi)genomic characterisation. IM samples were classified as genome-stable or genome-unstable based on their mutational burden/somatic copy-number alteration (CNA) profiles. Samples were probed for DDR signalling and cell proliferation, using the markers γH2AX and MCM2, respectively. The expression of the gastric stem cell marker, CD44v9, was also assessed. Tissue microarrays representing the GC progression spectrum were included. RESULTS: MCM2-positivity increased during GC progression, while γH2AX-positivity showed modest increase from normal to gastritis and IM stages, with further increase in GC. γH2AX levels correlated with the extent of chronic inflammation. Interestingly, genome-stable IM lesions had higher γH2AX levels underscoring a protective anti-cancer role for DDR signalling. In contrast, genome-unstable IM lesions with higher mutational burden/CNAs had lower γH2AX levels, elevated CD44v9 expression and modest promoter hypermethylation of DNA repair genes WRN, MLH1 and RAD52. CONCLUSIONS: Our data suggest that IM lesions with active DDR will likely experience a longer latency at the premalignant state until additional hits that override DDR signalling clonally expand and promote progression. These observations provide insights on the factors governing IM progression.


Asunto(s)
Mucosa Gástrica/patología , Histonas/genética , Componente 2 del Complejo de Mantenimiento de Minicromosoma/genética , Homólogo 1 de la Proteína MutL/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Neoplasias Gástricas , Helicasa del Síndrome de Werner/genética , Biopsia/métodos , Daño del ADN/genética , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/análisis , Masculino , Metaplasia/genética , Metaplasia/patología , Persona de Mediana Edad , Mutación , Factores Protectores , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
12.
Mod Pathol ; 33(10): 2075-2086, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32269290

RESUMEN

Spasmolytic polypeptide-expressing metaplasia (SPEM) and pyloric gland adenoma (PGA) in the stomach are metaplastic and neoplastic lesions, respectively, in which gastric body glands are replaced by pyloric glands. The aim of this study was to evaluate the genomic profile of SPEM and compare it with intestinal-type gastric cancer (GC) and PGA. Thirteen gastrectomies showing PGA with or without dysplasia, GC and SPEM were retrospectively selected. MUC5AC, MUC6, gastrin, and TFF2 IHC were performed. Lesions were subjected to laser capture microdissection followed by DNA extraction. Forty-three DNA samples were extracted from PGA without cytological dysplasia, PGA with low-grade and high-grade dysplasia and pyloric gland adenocarcinoma, GC, SPEM, and adjacent normal tissue from the body of the stomach and were subjected to exome sequencing for 49 genes that are commonly dysregulated in GC. Sanger sequencing was performed for confirmation. Twenty nonsynonymous mutations were identified in SPEM, and none of these were frameshifts or indels. PGA with or without cytological dysplasia showed a significantly higher number of mutations compared with SPEM. As cytological dysplasia increased from no dysplasia to dysplasia in PGA, the percentage of frameshift mutations, indels, and missense variations increased. Further missense or frameshift mutations were observed in the KRAS, APC, TP53, and CTNNB1 genes in the PGA group. In GC, mutations were observed in the TP53 gene (p.Arg248Gln). Missense mutations in the MUC5AC, KRAS, BRAF, and EZH2 genes were common between SPEM and GC. SPEM showed fewer genomic variations than GC and PGA, and was genomically distinct from the pyloric epithelium in PGA. Stepwise progression of PGA from PGA without dysplasia to PGA with dysplasia/adenocarcinoma was associated an increase in mutations. SPEM appears to be more genomically similar to GC than PGA.


Asunto(s)
Adenoma/genética , Mucosa Gástrica/patología , Lesiones Precancerosas/genética , Gastropatías/genética , Neoplasias Gástricas/genética , Adenoma/patología , Humanos , Captura por Microdisección con Láser , Metaplasia/genética , Metaplasia/patología , Mutación , Lesiones Precancerosas/patología , Estudios Retrospectivos , Singapur , Estómago/patología , Gastropatías/patología , Neoplasias Gástricas/patología
13.
Mol Biol Rep ; 47(10): 8007-8021, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32888123

RESUMEN

Osteoarthritis (OA) involves gradual destruction of articular cartilagemanifested by pain, stiffness of joints, and impaired movement especially in knees and hips. Non-vascularity of this tissue hinders its self-regenerative capacity and thus, the application of reparative or restorative modalities becomes imperative in OA treatment. In recent years, stem cell-based therapies have been explored as potential modalities for addressing OA complications. While mesenchymal stem cells (MSCs) hold immense promise, the recapitulation of native articular cartilage usingMSCs remains elusive. In this review, we have highlighted the chondrogenic potential of MSCs, factors guiding in vitro chondrogenic differentiation, biomaterials available for cartilage repair, their current market status, and the outcomes of major clinical trials. Our search on ClinicalTrials.gov using terms "stem cell" and "osteoarthritis" yielded 83 results. An analysis of the 29 trials that have been completed revealed differences in source of MSCs (bone marrow, adipose tissue, umbilical cord etc.), cell type (autologous or allogenic), and dose administered. Moreover, only 02 out of 29 studies have reported the use of matrix for cartilage repair. From future perspective, aconsensus on choice of cells, differentiation inducers, biomaterials, and clinical settings might pave a way for concocting robust strategies to improve the clinical applicability of biomimetic neocartilage constructs.


Asunto(s)
Cartílago Articular/metabolismo , Diferenciación Celular , Condrogénesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osteoartritis , Animales , Cartílago Articular/patología , Humanos , Células Madre Mesenquimatosas/patología , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/terapia
14.
Gastroenterology ; 152(1): 218-231.e14, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27670082

RESUMEN

BACKGROUND & AIMS: Little is known about the mechanisms of gastric carcinogenesis, partly because it has been a challenge to identify characterize gastric stem cells. Runx genes regulate development and their products are transcription factors associated with cancer development. A Runx1 enhancer element, eR1, is a marker of hematopoietic stem cells. We studied expression from eR1 in the stomach and the roles of gastric stem cells in gastric carcinogenesis in transgenic mice. METHODS: We used in situ hybridization and immunofluorescence analyses to study expression of Runx1 in gastric tissues from C57BL/6 (control) mice. We then created mice that expressed enhanced green fluorescent protein (EGFP) or CreERT2 under the control of eR1 (eR1-CreERT2;Rosa-Lox-Stop-Lox [LSL]-tdTomato, eR1-CreERT2;Rosa-LSL-EYFP mice). Gastric tissues were collected and lineage-tracing experiments were performed. Gastric organoids were cultured from eR1-CreERT2(5-2);Rosa-LSL-tdTomato mice and immunofluorescence analyses were performed. We investigated the effects of expressing oncogenic mutations in stem cells under control of eR1 using eR1-CreERT2;LSL-KrasG12D/+ mice; gastric tissues were collected and analyzed by histology and immunofluorescence. RESULTS: Most proliferation occurred in the isthmus; 86% of proliferating cells were RUNX1-positive and 76% were MUC5AC-positive. In eR1-EGFP mice, EGFP signals were detected mainly in the upper part of the gastric unit, and 83% of EGFP-positive cells were located in the isthmus/pit region. We found that eR1 marked undifferentiated stem cells in the isthmus and a smaller number of terminally differentiated chief cells at the base. eR1 also marked cells in the pyloric gland in the antrum. Lineage-tracing experiments demonstrated that stem cells in the isthmus and antrum continuously gave rise to mature cells to maintain the gastric unit. eR1-positive cells in the isthmus and pyloric gland generated organoid cultures in vitro. In eR1-CreERT2;LSL-Kras G12D/+ mice, MUC5AC-positive cells rapidly differentiated from stem cells in the isthmus, resulting in distinct metaplastic lesions similar to that observed in human gastric atrophy. CONCLUSIONS: Using lineage-tracing experiments in mice, we found that a Runx1 enhancer element, eR1, promotes its expression in the isthmus stem cells of stomach corpus as well as pyloric gland in the antrum. We were able to use eR1 to express oncogenic mutations in gastric stem cells, proving a new model for studies of gastric carcinogenesis.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Elementos de Facilitación Genéticos/genética , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Antro Pilórico/patología , ARN Mensajero/metabolismo , Células Madre/metabolismo , Células Madre/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Animales , Carcinogénesis , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Antígeno Ki-67/metabolismo , Metaplasia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mucina 5AC/metabolismo , Antro Pilórico/metabolismo , Técnicas de Cultivo de Tejidos
16.
N Engl J Med ; 368(24): 2266-76, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23758232

RESUMEN

BACKGROUND: Hepatocellular carcinoma is the third leading cause of cancer-related deaths worldwide. In the heterogeneous group of hepatocellular carcinomas, those with characteristics of embryonic stem-cell and progenitor-cell gene expression are associated with the worst prognosis. The oncofetal gene SALL4, a marker of a subtype of hepatocellular carcinoma with progenitor-like features, is associated with a poor prognosis and is a potential target for treatment. METHODS: We screened specimens obtained from patients with primary hepatocellular carcinoma for the expression of SALL4 and carried out a clinicopathological analysis. Loss-of-function studies were then performed to evaluate the role of SALL4 in hepatocarcinogenesis and its potential as a molecular target for therapy. To assess the therapeutic effects of a peptide that targets SALL4, we used in vitro functional and in vivo xenograft assays. RESULTS: SALL4 is an oncofetal protein that is expressed in the human fetal liver and silenced in the adult liver, but it is reexpressed in a subgroup of patients who have hepatocellular carcinoma and an unfavorable prognosis. Gene-expression analysis showed the enrichment of progenitor-like gene signatures with overexpression of proliferative and metastatic genes in SALL4-positive hepatocellular carcinomas. Loss-of-function studies confirmed the critical role of SALL4 in cell survival and tumorigenicity. Blocking SALL4-corepressor interactions released suppression of PTEN (the phosphatase and tensin homologue protein) and inhibited tumor formation in xenograft models in vivo. CONCLUSIONS: SALL4 is a marker for a progenitor subclass of hepatocellular carcinoma with an aggressive phenotype. The absence of SALL4 expression in the healthy adult liver enhances the potential of SALL4 as a treatment target in hepatocellular carcinoma. (Funded by the Singapore National Medical Research Council and others.).


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Factores de Transcripción/genética , Adulto , Animales , Carcinoma Hepatocelular/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Redes y Vías Metabólicas/fisiología , Ratones , Ratones Endogámicos , Fosfohidrolasa PTEN/metabolismo , Pronóstico , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
17.
Hepatology ; 61(3): 965-78, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25363290

RESUMEN

UNLABELLED: CCAAT enhancer binding protein α (C/EBPα) plays an essential role in cellular differentiation, growth, and energy metabolism. Here, we investigate the correlation between C/EBPα and hepatocellular carcinoma (HCC) patient outcomes and how C/EBPα protects cells against energy starvation. Expression of C/EBPα protein was increased in the majority of HCCs examined (191 pairs) compared with adjacent nontumor liver tissues in HCC tissue microarrays. Its upregulation was correlated significantly with poorer overall patient survival in both Kaplan-Meier survival (P=0.017) and multivariate Cox regression (P=0.028) analyses. Stable C/EBPα-silenced cells failed to establish xenograft tumors in nude mice due to extensive necrosis, consistent with increased necrosis in human C/EBPα-deficient HCC nodules. Expression of C/EBPα protected HCC cells in vitro from glucose and glutamine starvation-induced cell death through autophagy-involved lipid catabolism. Firstly, C/EBPα promoted lipid catabolism during starvation, while inhibition of fatty acid beta-oxidation significantly sensitized cell death. Secondly, autophagy was activated in C/EBPα-expressing cells, and the inhibition of autophagy by ATG7 knockdown or chloroquine treatment attenuated lipid catabolism and subsequently sensitized cell death. Finally, we identified TMEM166 as a key player in C/EBPα-mediated autophagy induction and protection against starvation. CONCLUSION: The C/EBPα gene is important in that it links HCC carcinogenesis to autophagy-mediated lipid metabolism and resistance to energy starvation; its expression in HCC predicts poorer patient prognosis.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Adulto , Anciano , Animales , Autofagia , Carcinoma Hepatocelular/metabolismo , Muerte Celular , Línea Celular Tumoral , Humanos , Metabolismo de los Lípidos , Neoplasias Hepáticas/metabolismo , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales
18.
J Basic Microbiol ; 55(1): 82-90, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24277414

RESUMEN

To investigate the biocontrol mechanism of two antagonistic Bacillus strains (Bacillus subtilis MB14 and Bacillus amyloliquefaciens MB101), three in vitro antagonism assays were screened and the results were concluded that both strains inhibited Rhizoctonia solani growth in a similar manner by dual culture assay, but the maximum percent of inhibition only resulted with MB101 by volatile and diffusible metabolite assays. Moreover, cell free supernatant (CFS) of MB101 also showed significant (p > 0.05) growth inhibition as compared to MB14, when 10 and 20% CFS mix with the growth medium of R. solani. After in vitro-validation, both strains were evaluated under greenhouse and the results concluded that strain MB101 had significant biocontrol potential as compared to MB14. Strain MB101 was enhanced the plant height, biomass and chlorophyll content of tomato plant through a higher degree of root colonization. In field trials, strain MB101 showed higher lessening in root rot symptoms with significant fruit yield as compare to strain MB14 and infected control. Next to the field study, the presence of four antibiotic genes (srfAA, fenD, ituC, and bmyB) also concluded the antifungal nature of both Bacillus strains. Phylogenetic analysis of protein sequences revealed a close relatedness of three genes (srfAA, fenD, and ituC) with earlier reported sequences of B. subtilis and B. amyloliquefaciens. However, bmyB showed heterogeneity in among both strains (MB14 and MB101) and it may be concluded that higher degree of antagonism, root colonization and different antibiotic producing genes may play an important role in biocontrol mechanism of strain MB101.


Asunto(s)
Antibiosis , Bacillus/fisiología , Agentes de Control Biológico , Enfermedades de las Plantas/prevención & control , Rhizoctonia/crecimiento & desarrollo , Solanum lycopersicum/crecimiento & desarrollo , Solanum lycopersicum/microbiología , Antibiosis/genética , Antifúngicos/química , Antifúngicos/metabolismo , Bacillus/genética , Bacillus/ultraestructura , Bacillus subtilis/genética , Bacillus subtilis/fisiología , Bacillus subtilis/ultraestructura , Proteínas Bacterianas/genética , Genes Bacterianos , Solanum lycopersicum/fisiología , Péptido Sintasas/genética , Filogenia , Raíces de Plantas/microbiología , Reacción en Cadena de la Polimerasa , Rhizoctonia/fisiología , Rhizoctonia/ultraestructura , Análisis de Secuencia de Proteína , Microbiología del Suelo
19.
J Basic Microbiol ; 54(6): 585-97, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23686438

RESUMEN

Plant protection through siderophore producing rhizobacteria (SPR) has emerged as a sustainable approach for crop health management. In present study, 220 bacteria isolated from tomato rhizosphere were screened for in vitro antagonistic activity against Rhizoctonia solani AG-4. Nine potent antagonistic strains viz., Alcaligenes sp. (MUN1, MB21, and MPF37), Enterobacter sp. (MPM1), Pseudomonas sp. (M10A and MB65), P. aeruginosa (MPF14 and MB123) and P. fluorescens (MPF47) were identified on the basis of physiological characters and 16S rDNA sequencing. These strains were able to produce hydrolytic enzymes, hydrogen cyanide, indole acetic acid, although, only few strains were able to solubilize phosphate. Two strains (MB123 and MPF47) showed significant disease reduction in glasshouse conditions were further evaluated under field conditions using three different application methods. Application of P. fluorescens (MPF47) in nursery as soil mix + seedling root treatments prior to transplantation resulted in significant disease reduction compared to control. Total chlorophyll and available iron were significantly higher in the MPF47 treated plants in contrast to infected control. In conclusion, siderophore producing bacteria MPF47 have strong biocontrol abilities and its application as soil mix + seedling root treatments provided strong shield to plant roots against R. solani and could be used for effective bio-management of pathogen.


Asunto(s)
Antibiosis , Bacterias/metabolismo , Rhizoctonia/crecimiento & desarrollo , Sideróforos/metabolismo , Microbiología del Suelo , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Técnicas de Tipificación Bacteriana , Análisis por Conglomerados , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Concentración de Iones de Hidrógeno , Solanum lycopersicum , Datos de Secuencia Molecular , Control Biológico de Vectores , Filogenia , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/prevención & control , ARN Ribosómico 16S/genética , Rizosfera , Análisis de Secuencia de ADN
20.
Surg Endosc ; 27(12): 4649-55, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23892761

RESUMEN

OBJECTIVE: We prospectively compared the diagnostic performance of autofluorescence imaging (AFI), magnifying narrow band imaging (mNBI), and probe-based confocal laser endomicroscopy (pCLE) with white light endoscopy (WLE) for the diagnosis of gastric intestinal metaplasia (GIM), using histology as the "gold standard." DESIGN: Chinese >50 years old with history of GIM were prospectively recruited. All subjects underwent WLE, followed by AFI and NBI, and finally pCLE. Patients were randomized to undergo either AFI before NBI or vice versa. In each patient, a minimum of six sites (antrum lesser and greater curve, body lesser and greater curve, incisura, cardia, and any lesion) were each examined by WLE, AFI, NBI, and pCLE. The diagnoses were made real-time. Biopsies for histology were taken from all examined sites. pCLE videos also were reviewed off-site. Analysis was performed per-site. RESULTS: A total of 125 sites in 20 patients were examined. For diagnosing GIM, real-time pCLE had better sensitivity (90.9 vs. 37.9%, p < 0.001) and accuracy (88.0 vs. 64.8%, p < 0.001) compared with WLE. Sensitivity (90.9 vs. 68.2%, p = 0.001), specificity (84.7 vs. 69.5%, p = 0.042), and accuracy (88 vs. 68.8%, p < 0.001) of real-time pCLE were better than AFI. Sensitivity, specificity, and accuracy of real-time pCLE and mNBI for diagnosing GIM were similar. Off-site pCLE had significantly better accuracy for diagnosing GIM compared to WLE, AFI, and mNBI. Off-site pCLE had superior specificity (94.9 vs. 84.7%, p = 0.031) and accuracy (95.2 vs. 88.0%, p = 0.012) compared with real-time pCLE. CONCLUSIONS: pCLE was superior to AFI and WLE for diagnosing GIM. Off-site review improved performance of pCLE.


Asunto(s)
Endoscopía Gastrointestinal/métodos , Mucosa Gástrica/patología , Microscopía Confocal/métodos , Imagen de Banda Estrecha/métodos , Neoplasias Gástricas/diagnóstico , Biopsia , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Metaplasia , Persona de Mediana Edad , Estudios Prospectivos , Curva ROC , Reproducibilidad de los Resultados , Grabación en Video
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA