Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.115
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 28(10): e18397, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38766687

RESUMEN

Malignant insulinoma is an extremely rare type of functioning pancreatic neuroendocrine tumour with a high degree of malignancy and a high incidence of metastasis. However, it is still unclear how malignant insulinomas develop and metastasize. Serum amyloid P component (SAP), a member of the pentraxin protein family, is an acute-phase protein secreted by liver cells. The role of SAP in insulinoma and the related mechanism are still unknown. To determine the effect of SAP on insulinoma, we crossed Rip1-Tag2 mice, which spontaneously develop insulinoma, and SAP knockout (KO) mice to generate Rip1-Tag2;SAP-/- mice. We found that SAP deletion significantly promoted the growth, invasion and metastasis of malignant insulinoma through C-X-C motif chemokine ligand 12 (CXCL12) secreted by cancer-associated fibroblasts (CAFs). Further study showed that SAP deletion promoted CXCL12 secretion by CAFs through the CXCR4/p38/ERK signalling pathway. These findings reveal a novel role and mechanism of SAP in malignant insulinoma and provide direct evidence that SAP may be a therapeutic agent for this disease.


Asunto(s)
Quimiocina CXCL12 , Insulinoma , Sistema de Señalización de MAP Quinasas , Ratones Noqueados , Receptores CXCR4 , Animales , Insulinoma/metabolismo , Insulinoma/patología , Insulinoma/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Eliminación de Gen , Progresión de la Enfermedad , Humanos , Línea Celular Tumoral , Proliferación Celular
2.
Pancreatology ; 23(5): 543-549, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37236853

RESUMEN

BACKGROUND/OBJECTIVES: Insulinomas are rare, functioning pancreatic neuroendocrine neoplasms (pNEN), whose gold standard therapy is surgical resection. Endoscopic ultrasound-guided radiofrequency ablation (EUS-RFA) is a recent technique that has emerged as a minimally invasive therapeutic option for patients with pancreatic lesions not eligible for surgery. In this study, we aimed to describe a series of patients with unresectable pancreatic insulinoma treated with EUS-RFA. METHODS: This is a single-center, retrospective study including all consecutive patients with functioning pancreatic insulinoma undergoing EUS-RFA for surgical unfitness or surgery refusal, between March 2017 and September 2021. Technical success (i.e., complete mass ablation), adverse event rate and severity, clinical and radiologic outcomes (i.e., symptom remission with a normal concentration of blood glucose, and the presence of intralesional necrosis), and post-procedural follow-up were assessed. RESULTS: A total of 10 patients (mean age: 67.1 ± 10.1years; F:M 7:3) were included. The mean size of insulinoma was 11.9 ± 3.3 mm. Technical success and clinical remission were achieved in 100% of patients. Only one (10%) patient was successfully treated with two RFA sessions. Two procedure-related early adverse events occurred, including two (20%) cases of mild abdominal pain. No major complications were observed. The complete radiologic response within 3 months after EUS-RFA was observed in all patients (100%). After a median follow-up of 19.5 (range12-59) months, symptom remission and persistent euglycemia were assessed in all the patients. CONCLUSIONS: Data from this case series suggest that EUS-RFA is a feasible and safe therapeutic approach for pancreatic insulinomas in patients unwilling or unable to undergo surgery with medium-term efficacy.


Asunto(s)
Insulinoma , Neoplasias Pancreáticas , Ablación por Radiofrecuencia , Humanos , Persona de Mediana Edad , Anciano , Insulinoma/diagnóstico por imagen , Insulinoma/cirugía , Insulinoma/patología , Estudios Retrospectivos , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/cirugía , Neoplasias Pancreáticas/patología , Ablación por Radiofrecuencia/métodos , Endosonografía/métodos , Ultrasonografía Intervencional
3.
Neuroendocrinology ; 113(6): 641-656, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36758529

RESUMEN

INTRODUCTION: 5-Iodotubercidin, a type of purine derivative, has attracted increasing attention in tumor chemotherapy because of its potential as an antitumor agent in recent years. In this study, we confirmed the effects on apoptosis in insulinoma cell lines induced by 5-iodotubercidin and tried to illuminate the underlying mechanisms. METHODS: We used 5-iodotubercidin in the treatment of insulinoma cells and the cell proliferation was examined using CCK-8 assay, colony-forming assays, and insulinoma animal models. Cell apoptosis was examined using TUNEL assays and Western blotting. Cellular DNA damage was shown by comet assay and immunofluorescence. The expression of apoptosis-regulating proteins and DNA damage biomarker was investigated by Western blotting. Subcutaneous inoculation of the insulinoma cells into nude mice was to measure blood glucose, insulin levels, and tumor growth. ATM siRNA and p53 siRNA were used as loss-of-function targets to evaluate 5-iodotubercidin treatment. RESULTS: 5-Iodotubercidin inhibited the proliferation of insulinoma cells and induced DNA damage and cell apoptosis. Moreover, 5-iodotubercidin induced ATM and p53 activated. In vivo, 5-iodotubercidin inhibited the growth of Ins-1 and Min-6 cells xenografts in nude mice. CONCLUSION: 5-Iodotubercidin induces DNA damage leading to insulinoma cells apoptosis by activating ATM/p53 pathway. Therefore, this is a potential strategy for treating insulinoma.


Asunto(s)
Insulinoma , Neoplasias Pancreáticas , Ratones , Animales , Humanos , Insulinoma/tratamiento farmacológico , Insulinoma/metabolismo , Insulinoma/patología , Ratones Desnudos , Proteína p53 Supresora de Tumor/genética , Apoptosis , Proliferación Celular , Neoplasias Pancreáticas/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Línea Celular Tumoral
4.
FASEB J ; 35(5): e21515, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811688

RESUMEN

The conserved endoplasmic reticulum (ER) membrane protein TRAPα (translocon-associated protein, also known as signal sequence receptor 1, SSR1) has been reported to play a critical but unclear role in insulin biosynthesis. TRAPα/SSR1 is one component of a four-protein complex including TRAPß/SSR2, TRAPγ/SSR3, and TRAPδ/SSR4. The TRAP complex topologically has a small exposure on the cytosolic side of the ER via its TRAPγ/SSR3 subunit, whereas TRAPß/SSR2 and TRAPδ/SSR4 function along with TRAPα/SSR1 largely on the luminal side of the ER membrane. Here, we have examined pancreatic ß-cells with deficient expression of either TRAPß/SSR2 or TRAPδ/SSR4, which does not perturb mRNA expression levels of other TRAP subunits, or insulin mRNA. However, deficient protein expression of TRAPß/SSR2 and, to a lesser degree, TRAPδ/SSR4, diminishes the protein levels of other TRAP subunits, concomitant with deficient steady-state levels of proinsulin and insulin. Deficient TRAPß/SSR2 or TRAPδ/SSR4 is not associated with any apparent defect of exocytotic mechanism but rather by a decreased abundance of the proinsulin and insulin that accompanies glucose-stimulated secretion. Amino acid pulse labeling directly establishes that much of the steady-state deficiency of intracellular proinsulin can be accounted for by diminished proinsulin biosynthesis, observed in a pulse-labeling as short as 5 minutes. The proinsulin and insulin levels in TRAPß/SSR2 or TRAPδ/SSR4 null mutant ß-cells are notably recovered upon re-expression of the missing TRAP subunit, accompanying a rebound of proinsulin biosynthesis. Remarkably, overexpression of TRAPα/SSR1 can also suppress defects in ß-cells with diminished expression of TRAPß/SSR2, strongly suggesting that TRAPß/SSR2 is needed to support TRAPα/SSR1 function.


Asunto(s)
Proteínas de Unión al Calcio/deficiencia , Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Insulina/biosíntesis , Insulinoma/patología , Glicoproteínas de Membrana/deficiencia , Proinsulina/biosíntesis , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores de Péptidos/deficiencia , Animales , Células Cultivadas , Células Secretoras de Insulina/citología , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas
5.
Molecules ; 27(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35408679

RESUMEN

Noninvasive targeted visualization of pancreatic beta cells or islets is becoming the focus of molecular imaging application in diabetes and islet transplantation studies. In this study, we aimed to produce the beta-cell-targeted peptide for molecular imaging of islet. We used phage display libraries to screen a beta-cell-targeted peptide, LNTPLKS, which was tagged with fluorescein isothiocyanate (FITC). This peptide was validated for targeting beta-cell with in vitro and in vivo studies. Immunocytochemistry (ICC) and fluorescence-activated cell sorting (FACS) analysis were used to validate the target specificity of the peptide. FITC-LNTPLKS displayed much higher fluorescence in beta cells vs. control cells in ICC. This discrimination was consistently observed using primary rodent islet. FACS analysis showed right shift of peak point in beta cells compared to control cells. The specific bind to in situ islet was verified by in vitro experiments using rodent and human pancreatic slices. The peptide also showed high affinity of islet grafts under the renal capsule. In the insulinoma animal model, we could find FITC-LNTPLKS accumulated specifically to the tumor, thus indicating a potential clinical application of molecular imaging of insulinoma. In conclusion, LNTPLKS showed a specific probe for beta-cells, which might be further utilized in targeted imaging/monitoring beta cells and theragnosis for beta-cells-related disease (diabetes, insulinoma, etc.).


Asunto(s)
Células Secretoras de Insulina , Insulinoma , Islotes Pancreáticos , Neoplasias Pancreáticas , Animales , Fluoresceína-5-Isotiocianato/química , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Insulinoma/patología , Imagen Molecular/métodos , Neoplasias Pancreáticas/metabolismo , Péptidos/química
6.
Med Lav ; 113(1): e2022007, 2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35226655

RESUMEN

A 50-year-old man who worked as a helicopter rescue pilot transiently lost consciousness while piloting a helicopter rescue. In the diagnostic process, all tests for the main possible differential diagnoses of loss of consciousness (cardiological and neurological) were performed and yielded normal results. Blood chemistry tests revealed recurrent fasting hypoglycemia and Chromogranin A was at the upper limit of normal. Fine needle aspiration guided by endoscopic ultrasonography was used to diagnose insulinoma-type neuroendocrine tumor of the pancreas. According to the Italian policies, the occupational physician aims to maintain professional skills without neglecting flight safety. A careful analysis of the relationship between the characteristics of the state of health of the aviator and his specific work needs was carried out, and he was given the opportunity to continue working as a rescue pilot thanks to medical therapies associated with organizational interventions in the workplace.


Asunto(s)
Insulinoma , Neoplasias Pancreáticas , Pilotos , Aeronaves , Estado de Conciencia , Humanos , Insulinoma/complicaciones , Insulinoma/diagnóstico , Insulinoma/patología , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patología , Inconsciencia/complicaciones
7.
Histochem Cell Biol ; 156(1): 69-73, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33743067

RESUMEN

Diabetes mellitus is a metabolic disorder projected to afflict 700 million people globally by 2045. Fundamental to the progression of diabetes is an insufficient supply of insulin to meet metabolic demand. The MIN6-K8 cell line is a mouse insulinoma model of pancreatic ß-cells frequently used to study the mechanisms of insulin secretion. Here, we evaluated the effects of short-term exposure to dimethyl sulfoxide (DMSO), a polar aprotic solvent commonly used in drug screening, on physiological characteristics of MIN6-K8 cells. Short-term exposure of MIN6-K8 cells to DMSO enhanced glucose-induced and tolbutamide-stimulated insulin secretion without significant effects on basal secretion or potassium responsiveness. Calcium influx was enhanced during glucose and tolbutamide treatments, suggesting that DMSO's mechanism of action is upstream of calcium-dependent insulin granule exocytosis. Based on these studies, investigators should use caution when conducting experiments with DMSO in the MIN6-K8 cell line and should report all DMSO concentrations when used as a solvent.


Asunto(s)
Dimetilsulfóxido/farmacología , Insulina/metabolismo , Insulinoma/metabolismo , Animales , Células Cultivadas , Insulinoma/patología , Ratones
8.
Mol Cell Biochem ; 476(11): 3879-3887, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34129156

RESUMEN

Citrus flavonoids particularly quercetin which is abundant in grapefruit, onion, green tea, berries etc. are known to have a protective effect on oxidative stress. Pancreatic ß cells which synthesize and secrete insulin are prone to oxidative stress induced damage because of low cellular antioxidant enzymes. To delineate the effects of quercetin on pancreatic ß cells we evaluated the protective effect of quercetin on TC6 insulinoma cells subjected to oxidative stress induced by tert-butyl-hydrogen-peroxide (TBHP). Quercetin was found to reduce TBHP induced apoptosis and trigger insulin secretion in response to glucose, in a dose-dependent manner. Quercetin treatment increased mitochondrial biogenesis, caused hypertrophy in pancreatic ß cells and activated mTOR signaling with a transient change in mitochondrial membrane potential and AMP/ATP. Activation of mTOR signaling resulted in enhanced insulin secretion in TC6 cells.


Asunto(s)
Flavonoides/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/tratamiento farmacológico , Estrés Oxidativo/fisiología , Quercetina/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Insulinoma/patología , Ratones , Transducción de Señal , terc-Butilhidroperóxido/farmacología
9.
Surg Endosc ; 35(1): 486-492, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32959183

RESUMEN

BACKGROUND: and study aims Pancreatic neuroendocrine tumors (pNETs) can be difficult to detect intra-operatively. The aim of this paper is to evaluate the safety and efficacy of preoperative endoscopic ultrasound guided fine needle tattooing (EUS-FNT) to facilitate intra-operative detection of pNETs. PATIENTS AND METHODS: Sixteen patients with pNETs (8 insulinoma and 8 non-functional pancreatic neuroendocrine tumors) underwent EUS-FNT. The procedure was carried out using the conventional curvilinear EUS. Tattooing was performed by intralesional injection of 1-2 mL of Spot® ink (Spot®, GI Supply, Comp Hill, PA, US) using a standard 22 gauge EUS-FNA needle. RESULTS: All identified pNETs could be tattooed in one session. The procedure was well tolerated in all patients without any complication. The time interval between tattooing and surgery was between 1 and 565 days (mean of 52 days). Nine patients underwent open and seven laparoscopic surgery. The tattooed lesions could be recognized in all but one patient. In one patient, a small hematoma secondary to the EUS-FNT was observed. Pathological examination of the resection specimen showed local R0 resection in all cases, and no interference with the specimen evaluation was encountered. CONCLUSIONS: Our results suggest that EUS-guided FNT is a safe and useful method to mark preoperatively small (≤ 2 cm) pNETs.


Asunto(s)
Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico/métodos , Tumores Neuroendocrinos/diagnóstico por imagen , Neoplasias Pancreáticas/diagnóstico por imagen , Tatuaje/métodos , Adulto , Anciano , Femenino , Humanos , Inyecciones Intralesiones , Insulinoma/diagnóstico por imagen , Insulinoma/patología , Insulinoma/cirugía , Laparoscopía , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/cirugía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Cuidados Preoperatorios , Estudios Retrospectivos , Factores de Tiempo
10.
Proc Natl Acad Sci U S A ; 115(5): 1027-1032, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339498

RESUMEN

The ß-cell-enriched MAFA transcription factor plays a central role in regulating glucose-stimulated insulin secretion while also demonstrating oncogenic transformation potential in vitro. No disease-causing MAFA variants have been previously described. We investigated a large pedigree with autosomal dominant inheritance of diabetes mellitus or insulinomatosis, an adult-onset condition of recurrent hyperinsulinemic hypoglycemia caused by multiple insulin-secreting neuroendocrine tumors of the pancreas. Using exome sequencing, we identified a missense MAFA mutation (p.Ser64Phe, c.191C>T) segregating with both phenotypes of insulinomatosis and diabetes. This mutation was also found in a second unrelated family with the same clinical phenotype, while no germline or somatic MAFA mutations were identified in nine patients with sporadic insulinomatosis. In the two families, insulinomatosis presented more frequently in females (eight females/two males) and diabetes more often in males (12 males/four females). Four patients from the index family, including two homozygotes, had a history of congenital cataract and/or glaucoma. The p.Ser64Phe mutation was found to impair phosphorylation within the transactivation domain of MAFA and profoundly increased MAFA protein stability under both high and low glucose concentrations in ß-cell lines. In addition, the transactivation potential of p.Ser64Phe MAFA in ß-cell lines was enhanced compared with wild-type MAFA. In summary, the p.Ser64Phe missense MAFA mutation leads to familial insulinomatosis or diabetes by impacting MAFA protein stability and transactivation ability. The human phenotypes associated with the p.Ser64Phe MAFA missense mutation reflect both the oncogenic capacity of MAFA and its key role in islet ß-cell activity.


Asunto(s)
Diabetes Mellitus/genética , Hiperinsulinismo/genética , Insulinoma/genética , Factores de Transcripción Maf de Gran Tamaño/genética , Proteínas Mutantes/genética , Mutación Missense , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Femenino , Genes Dominantes , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulinoma/metabolismo , Insulinoma/patología , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Proteínas Mutantes/metabolismo , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Linaje , Estabilidad Proteica , Activación Transcripcional , Secuenciación del Exoma
11.
J Postgrad Med ; 67(3): 164-167, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34414928

RESUMEN

Insulinoma is a rare neuroendocrine tumor originating from hypersecreting beta-cells of islets of Langerhans in the pancreas. We report a case of 72-year-old male, with chronic alcohol abuse, presenting with atypical features like refractory recurrent secondary generalized seizures and behavioral disturbances with increased irritability, initially mistreated as alcohol withdrawal. Detailed history, particularly the relationship of the symptoms with food intake, made us think of other causes of seizures. Fasting biochemical investigations and localizing studies helped clinch the diagnosis. The tumor was localized with the help of endoscopic ultrasonography and whole-body Ga68-DOTANOC PET-CT. The patient was treated conservatively with diazoxide and is doing well on follow-up. The present case report emphasizes the importance of detailed clinical history, more so in atypically presenting cases of refractory seizures. Insulinoma can be medically managed despite surgery being the gold standard curative treatment.


Asunto(s)
Antihipertensivos/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Diazóxido/uso terapéutico , Insulinoma/diagnóstico por imagen , Neoplasias Pancreáticas/patología , Anciano , Alcoholismo , Complicaciones de la Diabetes , Diabetes Mellitus Tipo 2/sangre , Endosonografía , Humanos , Insulinoma/patología , Insulinoma/cirugía , Masculino , Compuestos Organometálicos/metabolismo , Neoplasias Pancreáticas/cirugía , Tomografía Computarizada por Tomografía de Emisión de Positrones , Cintigrafía , Convulsiones/etiología , Imagen de Cuerpo Entero
12.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884483

RESUMEN

Cells for therapeutic use are often preserved at +4 °C, and the storage period is generally limited to 2-3 days. Here, we report that the survival rate (%) of mammalian cells is improved to 10-20 days when they are preserved with a subzero supercooled solution containing the antifreeze protein (AFP), for which an ability to stabilize both supercooled water and cell membrane integrity has been postulated. We chose adherent rat insulinoma (RIN-5F) cells as the preservation target, which were immersed into -5 °C-, -2 °C-, or +4 °C-chilled "unfrozen" solution of Euro-Collins or University of Washington (UW) containing the AFP sample obtained from insect or fish. Our results show that the survival rate of the cells preserved with the solution containing insect AFP was always higher than that of the fish AFP solution. A combination of the -5 °C-supercooling and insect AFP gave the best preservation result, namely, UW solution containing insect AFP kept 53% of the cells alive, even after 20 days of preservation at -5 °C. The insect AFP locates highly organized ice-like waters on its molecular surface. Such waters may bind to semiclathrate waters constructing both embryonic ice crystals and a membrane-water interface in the supercooled solution, thereby protecting the cells from damage due to chilling.


Asunto(s)
Proteínas Anticongelantes/administración & dosificación , Criopreservación/métodos , Crioprotectores/administración & dosificación , Hipotermia/tratamiento farmacológico , Proteínas de Insectos/administración & dosificación , Insulinoma/patología , Animales , Supervivencia Celular , Hielo , Insectos , Neoplasias Pancreáticas/patología , Ratas , Células Tumorales Cultivadas
13.
Semin Cell Dev Biol ; 73: 57-63, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28779980

RESUMEN

A wide range of cellular activities including protein folding and cell secretion, such as neurotransmission or insulin release, are all governed by intracellular pH homeostasis, underscoring the importance of pH on critical life processes. Nano- scale pH measurements of cells and biomolecules therefore hold great promise in understanding a plethora of cellular functions, in addition to disease detection and therapy. In the current study, a novel approach using cadmium telluride quantum dots (CdTeQDs) as pH sensors, combined with fluorescent imaging, spectrofluorimetry, atomic force microscopy (AFM), and Western blot analysis, enabled the study of intracellular pH dynamics at 1 milli-pH sensitivity and 80nm pixel resolution, during insulin secretion. Additionally, the pH-dependent interaction between membrane fusion proteins, also called the soluble N-ethylmaleimide-sensitive factor activating protein receptor (SNARE), was determined. Glucose stimulation of CdTeQD-loaded insulin secreting Min-6 mouse insulinoma cell line demonstrated the initial (5-6min) intracellular acidification reflected as a loss in QD fluorescence, followed by alkalization and a return to resting pH in 10min. Analysis of the SNARE complex in insulin secreting Min-6 cells demonstrated an initial gain followed by loss of complexed SNAREs in 10min. Stabilization of the SNARE complex at low intracellular pH is further supported by results from studies utilizing both native and AFM measurements of liposome-reconstituted recombinant neuronal SNAREs, providing a molecular understanding of the role of pH during cell secretion.


Asunto(s)
Fluorescencia , Insulinoma/metabolismo , Insulinoma/patología , Fusión de Membrana , Microscopía de Fuerza Atómica , Imagen Óptica , Animales , Concentración de Iones de Hidrógeno , Simulación de Dinámica Molecular
14.
Carcinogenesis ; 41(3): 326-333, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31155641

RESUMEN

Insulinoma-associated protein 1 (INSM1), a zinc finger transcriptional factor, is proven to be deregulated in several types of cancers. However, comprehension of the molecular mechanism of INSM1-mediated tumor progression remains poor. Here, we show that the radioresistant nasopharyngeal carcinoma (NPC) patients have higher expressions of INSM1 that correlated with poor prognosis. Genetic manipulation of INSM1 expression sufficiently controls the response of NPC cells to irradiation (IR). Mechanistically, cells exposed to IR, increased intracellular INSM1 competitively disrupts the interaction of cyclin D1 and CDK4 resulting in cell survival by the cyclin D1-dependent DNA repair machinery. Moreover, knockdown of INSM1 sensitives NPC cells to IR in vivo and protects xenograft mice from mortality. Taken together, these results indicate that INSM1 modulates NPC to radiotherapy by controlling cyclin D1-dependent DNA repair machinery that could be manipulated as a novel molecular target for NPC therapy.


Asunto(s)
Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/genética , Insulinoma/radioterapia , Carcinoma Nasofaríngeo/radioterapia , Proteínas Represoras/genética , Animales , Biopsia , Línea Celular Tumoral , Reparación del ADN/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Xenoinjertos , Humanos , Insulinoma/genética , Insulinoma/patología , Masculino , Ratones , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patología , Tolerancia a Radiación/genética
15.
J Biol Chem ; 294(45): 16992-17006, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31591269

RESUMEN

Zinc transporter-8 (ZnT8) primarily functions as a zinc-sequestrating transporter in the insulin-secretory granules (ISGs) of pancreatic ß-cells. Loss-of-function mutations in ZnT8 are associated with protection against type-2 diabetes (T2D), but the protective mechanism is unclear. Here, we developed an in-cell ZnT8 assay to track endogenous ZnT8 responses to metabolic and inflammatory stresses applied to human insulinoma EndoC-ßH1 cells. Unexpectedly, high glucose and free fatty acids did not alter cellular ZnT8 levels, but proinflammatory cytokines acutely, reversibly, and gradually down-regulated ZnT8. Approximately 50% of the cellular ZnT8 was localized to the endoplasmic reticulum (ER), which was the primary target of the cytokine-mediated ZnT8 down-regulation. Transcriptome profiling of cytokine-exposed ß-cells revealed an adaptive unfolded protein response (UPR) including a marked immunoproteasome activation that coordinately degraded ZnT8 and insulin over a 1,000-fold cytokine concentration range. RNAi-mediated ZnT8 knockdown protected cells against cytokine cytotoxicity, whereas inhibiting immunoproteasomes blocked cytokine-induced ZnT8 degradation and triggered a transition of the adaptive UPR to cell apoptosis. Hence, cytokine-induced down-regulation of the ER ZnT8 level promotes adaptive UPR, acting as a protective mechanism that decongests the ER burden of ZnT8 to protect ß-cells from proapoptotic UPR during chronic low-grade inflammation.


Asunto(s)
Regulación hacia Abajo , Estrés del Retículo Endoplásmico/genética , Insulinoma/patología , Islotes Pancreáticos/metabolismo , Neoplasias Pancreáticas/patología , Transportador 8 de Zinc/genética , Línea Celular Tumoral , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/genética , Inflamación/patología , Insulina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas , Transportador 8 de Zinc/deficiencia
16.
J Cell Biochem ; 121(1): 840-855, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31452250

RESUMEN

Pancreatic stellate cells (PSCs) secrete various factors, which can influence the ß-cell function. The identification of stellate cell infiltration into the islets in pancreatic diseases suggests possible existence of cross-talk between these cells. To elucidate the influence of PSCs on ß-cell function, mouse PSCs were cocultured with Min6 cells using the Transwell inserts. Glucose-stimulated insulin secretion from Min6 cells in response to PSCs was quantified by enzyme-linked immunosorbent assay and insulin gene expression was measured by quantitative polymerase chain reaction. Upon cytometric identification of IL6 in PSC culture supernatants, Min6 cells were cultured with IL6 to assess its influence on the insulin secretion and gene expression. PLC-IP3 pathway inhibitors were added in the cocultures, to determine the influence of PSC-secreted IL6 on Glucose-stimulated insulin secretion from Min6 cells. Increased insulin secretion with a concomitant decrease in total insulin content was noticed in PSC-cocultured Min6 cells. Although increased GSIS was noted from IL6-treated Min6 cells, no change in the total insulin content was noted. Coculture of Min6 cells with PSCs or their exposure to IL6 did not alter either the expression of ß-cell-specific genes or that of miRNA-375. PSC-cocultured Min6 cells, in the presence of PLC-IP3 pathway inhibitors (U73122, Neomycin, and Xestospongin C), did not revoke the observed increase in GSIS. In conclusion, the obtained results indicate that augmented insulin secretion from Min6 cells in response to PSC secretions is independent of IL6-mediated PLC-IP3 pathway.


Asunto(s)
Glucosa/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Interleucina-6/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/patología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Células Estrelladas Pancreáticas/citología , Células Estrelladas Pancreáticas/efectos de los fármacos , Edulcorantes/farmacología
17.
FASEB J ; 33(1): 88-100, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29957057

RESUMEN

Zinc finger BED domain containing protein 6 ( Zbed6) has evolved from a domesticated DNA transposon and encodes a transcription factor unique to placental mammals. The aim of the present study was to investigate further the role of ZBED6 in insulin-producing cells, using mouse MIN6 cells, and to evaluate the effects of Zbed6 knockdown on basal ß-cell functions, such as morphology, transcriptional regulation, insulin content, and release. Zbed6-silenced cells and controls were characterized with a range of methods, including RNA sequencing, chromatin immunoprecipitation sequencing, insulin content and release, subplasma membrane Ca2+ measurements, cAMP determination, and morphologic studies. More than 700 genes showed differential expression in response to Zbed6 knockdown, which was paralleled by increased capacity to generate cAMP, as well as by augmented subplasmalemmal calcium concentration and insulin secretion in response to glucose stimulation. We identified >4000 putative ZBED6-binding sites in the MIN6 genome, with an enrichment of ZBED6 sites at upregulated genes, such as the ß-cell transcription factors v-maf musculoaponeurotic fibrosarcoma oncogene homolog A and Nk6 homeobox 1. We also observed altered morphology/growth patterns, as indicated by increased cell clustering, and in the appearance of axon-like Neurofilament, medium polypeptide and tubulin ß 3, class III-positive protrusions. We conclude that ZBED6 acts as a transcriptional regulator in MIN6 cells and that its activity suppresses insulin production, cell aggregation, and neuronal-like differentiation.-Wang, X., Jiang, L., Wallerman, O., Younis, S., Yu, Q., Klaesson, A., Tengholm, A., Welsh, N., Andersson, L. ZBED6 negatively regulates insulin production, neuronal differentiation, and cell aggregation in MIN6 cells.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/patología , Insulina/metabolismo , Insulinoma/patología , Neuronas/patología , Neoplasias Pancreáticas/patología , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Adhesión Celular , Agregación Celular , Regulación de la Expresión Génica , Silenciador del Gen , Glucosa/administración & dosificación , Secuenciación de Nucleótidos de Alto Rendimiento , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Ratones , Neuronas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transcripción Genética , Células Tumorales Cultivadas
18.
Pharmacol Res ; 159: 104983, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32504838

RESUMEN

A hallmark of diabetes mellitus is the inability of pancreatic ß-cells to secrete sufficient amounts of insulin for maintaining normoglycemia. The formation of smaller islets may underlie the development of a diabetic phenotype, as a decreased ß-cell mass will produce an insufficient amount of insulin. For a pharmacological intervention it is crucial to identify the proteins determining ß-cell mass. Here, we identified the ternary complex factor (TCF) Elk-1 as a regulator of the size of pancreatic islets. Elk-1 mediates, together with a dimer of the serum-response factor (SRF), serum response element-regulated gene transcription. Elk-1 is activated in glucose-treated pancreatic ß-cells but the biological functions of this protein in ß-cells are so far unknown. Elk-1 and homologous TCF proteins are expressed in islets and insulinoma cells. Gene targeting experiments revealed that the TCF proteins show redundant activities. To solve the problem of functional redundancy of these homologous proteins, we generated conditional transgenic mice expressing a dominant-negative mutant of Elk-1 in pancreatic ß-cells. The mutant competes with the wild-type TCFs for DNA and SRF-binding. Expression of the Elk-1 mutant in pancreatic ß-cells resulted in the generation of significantly smaller islets and increased caspase-3 activity, indicating that apoptosis was responsible for the reduction of the pancreatic islet size. Glucose tolerance tests revealed that transgenic mice expressing the dominant-negative mutant of Elk-1 in pancreatic ß-cells displayed impaired glucose tolerance. Thus, we show here for the first time that TCF controls important functions of pancreatic ß-cells in vivo. Elk-1 may be considered as a new therapeutic target for the treatment of diabetes.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Línea Celular Tumoral , Tamaño de la Célula , Homeostasis , Células Secretoras de Insulina/patología , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Ratones Transgénicos , Mutación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/genética
19.
J Biochem Mol Toxicol ; 34(7): e22488, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32128977

RESUMEN

Insulinomas originate from pancreatic ß cells and it is the most widely known tumor. Indomethacin is a nonsteroidal anti-inflammatory drug, which is used for blocking the production of some natural substances that cause inflammation and decrease pain. In this study, I aimed to investigate the effects of indomethacin on rat insulinoma INS-1 cells. The relationship between cell death and insulin metabolism was determined with the administration of indomethacin. The cell viability by WST-1; the apoptosis and necrosis levels by ELISA kits; malondialdehyde levels by spectrophotometer; and beclin, intracellular insulin, insulin secretion, KCa3.1, insulin receptor (IR), glucose transporter type 2 (GLUT2), activating transcription factor 2 (ATF2), Elk1, c-Jun, Akt and phosphorylated ATF2, Elk1, c-Jun, Akt, intracellular betacellulin and betacellulin secretion levels by Western blot analysis investigated. The Ins1, Ins2, IR, GLUT2, ATF2, Elk1, c-Jun, Akt, and Betacellulin gene expression levels were determined by the real-time quantitative reverse transcription-polymerase chain reaction method. Apoptotic cell death was observed with the administration of indomethacin. The insulin secretion and Ins1, Ins2 gene expression levels decreased. The insulin receptor and GLUT2 levels increased, while KCa3.1 (KCNN4) levels decreased with the administration of indomethacin to insulinoma INS-1 cells. A decrease was observed in the total c-Jun, phosphorylated ATF2, Elk1, c-Jun, and Akt levels. Betacellulin secretion levels increased. In insulinoma INS-1 cells, apoptotic cell death occurred in the following manner: (i) indomethacin might decrease insulin secretion by reducing KCa3.1, (ii) might inactivate the JNK/ERK pathway with the inactivity of transcription factors.


Asunto(s)
Apoptosis/efectos de los fármacos , Indometacina/farmacología , Secreción de Insulina/efectos de los fármacos , Insulina/biosíntesis , Insulinoma/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/patología , Neoplasias Pancreáticas/patología , Ratas
20.
Exp Cell Res ; 382(1): 111467, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31202710

RESUMEN

Hyperglycemia and hyperlipidemia (glycolipotoxicity)-triggered islet ß-cell dysfunction is known to drive the progression of obesity-related type 2 diabetes, however the underlying mechanisms have not been clearly elucidated. The current study aimed to investigate the role of mitogen-activated protein kinase phosphatase 5 (MKP-5) in islet cells under glucolipotoxic conditions. Using gene overexpression and knockdown approaches, we demonstrated that MKP-5 could alleviate glucolipotoxicity-induced apoptosis via the endoplasmic reticulum (ER) stress and mitochondrial apoptosis pathways owing to the altered regulation of caspase family members and ER stress-related molecules in MIN6 and primary islet cells. Overexpression of MKP-5 reversed the glucose and palmitic acid (GP)-induced impairment of insulin secretion as well as the abnormal decreases in the expression of islet functional genes, thereby maintaining the normal insulin secretory functionality, whereas the absence of MKP-5 aggravated islet cell dysfunction. In parallel, the production of ROS and increased inflammation-associated genes in response to GP were also reduced upon MKP-5 overexpression. Further, inhibition of JNK or P38 MAPK pathways resisted to glucolipotoxicity observed in MKP-5 knockdown MIN6 cells. These findings indicate that MKP-5 is an important mediator for glucolipotoxicity-induced islet cell dysfunction and apoptosis, with JNK and P38 as the critical downstream pathways.


Asunto(s)
Apoptosis/fisiología , Fosfatasas de Especificidad Dual/fisiología , Estrés del Retículo Endoplásmico/fisiología , Glucosa/toxicidad , Islotes Pancreáticos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/fisiología , Palmitatos/toxicidad , Animales , Línea Celular Tumoral , Dieta Alta en Grasa/efectos adversos , Fosfatasas de Especificidad Dual/genética , Técnicas de Silenciamiento del Gen , Humanos , Insulina/metabolismo , Insulinoma/patología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Neoplasias Pancreáticas/patología , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA