Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
bioRxiv ; 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746217

RESUMEN

Serine/threonine kinase AKT isoforms play a well-established role in cell metabolism and growth. Most pancreatic adenocarcinoma (PDAC) harbors activation mutations of KRAS, which activates the PI3K/AKT signaling pathway. However, AKT inhibitors are not effective in the treatment of pancreatic cancer. To better understand the role of AKT signaling in mutant-KRAS pancreatic tumors, this study utilizes proteolysis-targeting chimeras (PROTACs) and CRISPR-Cas9-genome editing to investigate AKT proteins. PROTAC down-regulation of AKT proteins markedly slowed the growth of three pancreatic tumor cell lines harboring mutant KRAS. In contrast, inhibition of AKT kinase activity alone had very little effect on the growth of these cell lines. Concurrent genetic deletion of all AKT isoforms (AKT1, AKT2, and AKT3) in the KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cell line also dramatically slowed its growth in vitro and when orthotopically implanted in syngeneic mice. Surprisingly, insulin-like growth factor-1 (IGF-1), but not epidermal growth factor (EGF), restored KPC cell growth in serum-deprived conditions and the IGF-1 growth stimulation effect was AKT dependent. RNA-seq analysis of AKT1/2/3-deficient KPC cells suggested that reduced cholesterol synthesis may be responsible for the decreased response to IGF-1 stimulation. These results indicate that the presence of all three AKT isoforms supports pancreatic tumor cell growth and pharmacological degradation of AKT proteins may be more effective than AKT catalytic inhibitors for treating pancreatic cancer.

2.
J Ethnopharmacol ; 321: 117202, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37742878

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ginkgo biloba L. is a well-known and highly regarded resource in Chinese traditional medicine due to its effectiveness and safety. Ginkgo Folium, the leaf of Ginkgo biloba L., contains biologically active constituents with diverse pharmacological activities. Recent studies have shown promising antitumor effects of the bioactive constituents found in Ginkgo Folium against various types of cancer cells, highlighting its potential as a natural source of antitumor agents. Further research is needed to elucidate the underlying mechanisms and optimize its therapeutic potential. AIM OF THE REVIEW: To provide a detailed understanding of the pharmacological activities of Ginkgo Folium and its potential therapeutic benefits for cancer patients. MATERIALS AND METHODS: In this study, we conducted a thorough and systematic search of multiple online databases, including PubMed, Web of Science, Medline, using relevant keywords such as "Ginkgo Folium," "flavonoids," "terpenoids," "Ginkgo Folium extracts," and "antitumor" to cover a broad range of studies that could inform our review. Additionally, we followed a rigorous selection process to ensure that the studies included in our review met the predetermined inclusion criteria. RESULTS: The active constituents of Ginkgo Folium primarily consist of flavonoids and terpenoids, with quercetin, kaempferol, isorhamnetin, ginkgolides, and bilobalide being the major compounds. These active constituents exert their antitumor effects through crucial biological events such as apoptosis, cell cycle arrest, autophagy, and inhibition of invasion and metastasis via modulating diverse signaling pathways. During the process of apoptosis, active constituents primarily exert their effects by modulating the caspase-8 mediated death receptor pathway and caspase-9 mediated mitochondrial pathway via regulating specific signaling pathways. Furthermore, by modulating multiple signaling pathways, active constituents effectively induce G1, G0/G1, G2, and G2/M phase arrest. Among these, the pathways associated with G2/M phase arrest are particularly extensive, with the cyclin-dependent kinases (CDKs) being most involved. Moreover, active constituents primarily mediate autophagy by modulating certain inflammatory factors and stressors, facilitating the fusion stage between autophagosomes and lysosomes. Additionally, through the modulation of specific chemokines and matrix metalloproteinases, active constituents effectively inhibit the processes of epithelial-mesenchymal transition (EMT) and angiogenesis, exerting a significant impact on cellular invasion and migration. Synergistic effects are observed among the active constituents, particularly quercetin and kaempferol. CONCLUSION: Active components derived from Ginkgo Folium demonstrate a comprehensive antitumor effect across various levels and pathways, presenting compelling evidence for their potential in new drug development. However, in order to facilitate their broad and adaptable clinical application, further extensive experimental investigations are required to thoroughly explore their efficacy, safety, and underlying mechanisms of action.


Asunto(s)
Ginkgo biloba , Quercetina , Humanos , Quercetina/farmacología , Quempferoles , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Flavonoides
3.
bioRxiv ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-37546948

RESUMEN

Most human pancreatic ductal adenocarcinoma (PDAC) are not infiltrated with cytotoxic T cells and are highly resistant to immunotherapy. Over 90% of PDAC have oncogenic KRAS mutations, and phosphoinositide 3-kinases (PI3Ks) are direct effectors of KRAS. Our previous study demonstrated that ablation of Pik3ca in KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cells induced host T cells to infiltrate and completely eliminate the tumors in a syngeneic orthotopic implantation mouse model. Now, we show that implantation of Pik3ca-/- KPC (named αKO) cancer cells induces clonal expansion of cytotoxic T cells infiltrating the pancreatic tumors. To identify potential molecules that can regulate the activity of these anti-tumor T cells, we conducted an in vivo genome-wide gene-deletion screen using αKO cells implanted in the mouse pancreas. The result shows that deletion of propionyl-CoA carboxylase subunit B gene (Pccb) in αKO cells (named p-αKO) leads to immune evasion, tumor progression and death of host mice. Surprisingly, p-αKO tumors are still infiltrated with clonally expanded CD8+ T cells but they are inactive against tumor cells. However, blockade of PD-L1/PD1 interaction reactivated these clonally expanded T cells infiltrating p-αKO tumors, leading to slower tumor progression and improve survival of host mice. These results indicate that Pccb can modulate the activity of cytotoxic T cells infiltrating some pancreatic cancers and this understanding may lead to improvement in immunotherapy for this difficult-to-treat cancer.

4.
J Adv Res ; 2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37689240

RESUMEN

INTRODUCTION: ß-Elemene (ß-ELE), derived from Curcuma wenyujin, has anticancer effect on non-small cell lung cancer (NSCLC). However, the potential target and detail mechanism were still not clear. TFEB is the master regulator of lysosome biogenesis. Ferroptosis, a promising strategy for cancer therapy could be triggered via suppression on glutathione peroxidase 4 (GPX4). Weather TFEB-mediated lysosome degradation contributes to GPX4 decline and how ß-ELE modulates on this process are not clear. OBJECTIVES: To observe the action of ß-ELE on TFEB, and the role of TFEB-mediated GPX4 degradation in ß-ELE induced ferroptosis. METHODS: Surface plasmon resonance (SPR) and molecular docking were applied to observe the binding affinity of ß-ELE on TFEB. Activation of TFEB and lysosome were observed by immunofluorescence, western blot, flow cytometry and qPCR. Ferroptosis induced by ß-ELE was observed via lipid ROS, a labile iron pool (LIP) assay and western blot. A549TFEB KO cells were established via CRISPR/Cas9. The regulation of TFEB on GPX4 and ferroptosis was observed in ß-ELE treated A549WT and A549TFEB KO cells, which was further studied in orthotopic NOD/SCID mouse model. RESULTS: ß-ELE can bind to TFEB, notably activate TFEB, lysosome and transcriptional increase on downstream gene GLA, MCOLN1, SLC26A11 involved in lysosome activity in EGFR wild-type NSCLC cells. ß-ELE increased GPX4 ubiquitination and lysosomal localization, with the increase on lysosome degradation of GPX4. Furthermore, ß-ELE induced ferroptosis, which could be promoted by TFEB overexpression or compromised by TFEB knockout. Genetic knockout or inactivation of TFEB compromised ß-ELE induced lysosome degradation of GPX4, which was further demonstrated in orthotopic NSCLC NOD/SCID mice model. CONCLUSION: This study firstly demonstrated that TFEB promoted GPX4 lysosome degradation contributes to ß-ELE induced ferroptosis in EGFR wild-type NSCLC, which gives a clue that TFEB mediated GPX4 degradation would be a novel strategy for ferroptosis induction and NSCLC therapy.

5.
Cancers (Basel) ; 15(14)2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37509352

RESUMEN

Dendritic cells (DCs) are professional antigen-presenting cells that play a crucial role in activating naive T cells through presenting antigen information, thereby influencing immunity and anti-cancer responses. Fascin, a 55-kDa actin-bundling protein, is highly expressed in mature DCs and serves as a marker protein for their identification. However, the precise role of fascin in intratumoral DCs remains poorly understood. In this review, we aim to summarize the role of fascin in both normal and intratumoral DCs. In normal DCs, fascin promotes immune effects through facilitating DC maturation and migration. Through targeting intratumoral DCs, fascin inhibitors enhance anti-tumor immune activity. These roles of fascin in different DC populations offer valuable insights for future research in immunotherapy and strategies aimed at improving cancer treatments.

6.
J Mol Med (Berl) ; 101(8): 917-929, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37328669

RESUMEN

Transcription factor EB, a member of the microphthalmia-associated transcription factor (MiTF/TFE) family, is a master regulator of autophagy, lysosome biogenesis, and TAMs. Metastasis is one of the main reasons for the failure of tumor therapy. Studies on the relationship between TFEB and tumor metastasis are contradictory. On the positive side, TFEB mainly affects tumor cell metastasis via five aspects, including autophagy, epithelial-mesenchymal transition (EMT), lysosomal biogenesis, lipid metabolism, and oncogenic signaling pathways; on the negative side, TFEB mainly affects tumor cell metastasis in two aspects, including tumor-associated macrophages (TAMs) and EMT. In this review, we described the detailed mechanism of TFEB-mediated regulation of metastasis. In addition, we also described the activation and inactivation of TFEB in several aspects, including the mTORC1 and Rag GTPase systems, ERK2, and AKT. However, the exact process by which TFEB regulates tumor metastasis remains unclear in some pathways, which requires further studies.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal , Lisosomas/metabolismo , Fosforilación
7.
World J Stem Cells ; 14(7): 473-489, 2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-36157529

RESUMEN

With advances in the fields of regenerative medicine, cell-free therapy has received increased attention. Exosomes have a variety of endogenous properties that provide stability for molecular transport across biological barriers to cells, as a form of cell-to-cell communication that regulates function and phenotype. In addition, exosomes are an important component of paracrine signaling in stem-cell-based therapy and can be used as a stand-alone therapy or as a drug delivery system. The remarkable potential of exosomes has paved the pathway for cell-free treatment in bone regeneration. Exosomes are enriched in distinct noncoding RNAs (ncRNAs), including microRNAs, long ncRNAs and circular RNAs. Different ncRNAs have multiple functions. Altered expression of ncRNA in exosomes is associated with the regenerative potential and development of various diseases, such as femoral head osteonecrosis, myocardial infarction, and cancer. Although there is increasing evidence that exosome-derived ncRNAs (exo-ncRNAs) have the potential for bone regeneration, the detailed mechanisms are not fully understood. Here, we review the biogenesis of exo-ncRNA and the effects of ncRNAs on angiogenesis and osteoblast- and osteoclast-related pathways in different diseases. However, there are still many unsolved problems and challenges in the clinical application of ncRNA; for instance, production, storage, targeted delivery and therapeutic potency assessment. Advancements in exo-ncRNA methods and design will promote the development of therapeutics, revolutionizing the present landscape.

8.
Shanghai Kou Qiang Yi Xue ; 30(5): 477-480, 2021 Oct.
Artículo en Chino | MEDLINE | ID: mdl-34888598

RESUMEN

PURPOSE: To investigate the effect of paired related homeobox1 (PRRX1) on autophagy of salivary adenoidcystic carcinoma (SACC) cells. METHODS: PRRX1-overexpressed lentiviral vectors and their negative control lentiviral vectors were used to transfect SACC cells. The transfection effect was detected by Western blot. Autophagosome was observed under transmission electron microscopy(TEM). The level of autophagy markers (LC3-II/Beclin1) was detected by immunofluorescence and Western blot. Statistical analysis was performed with SPSS 17.0 software package. RESULTS: PRRX1 protein level increased, autophagosome number decreased, and autophagy marker level decreased in the PRRX1 overexpressed group, compared to the control (P<0.05). CONCLUSIONS: PRRX1 inhibits the autophagy of SACC cells.


Asunto(s)
Carcinoma Adenoide Quístico , Neoplasias de las Glándulas Salivales , Autofagia , Beclina-1 , Carcinoma Adenoide Quístico/genética , Línea Celular Tumoral , Proteínas de Homeodominio , Humanos , Neoplasias de las Glándulas Salivales/genética
9.
J Thromb Haemost ; 18(12): 3359-3370, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32920974

RESUMEN

OBJECTIVE: Cardiovascular complications are the leading cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). The acquired kinase mutation JAK2V617F plays a central role in these disorders. Mechanisms responsible for cardiovascular dysfunction in MPNs are not fully understood, limiting the effectiveness of current treatment. Vascular endothelial cells (ECs) carrying the JAK2V617F mutation can be detected in patients with MPNs. The goal of this study was to test the hypothesis that the JAK2V617F mutation alters endothelial function to promote cardiovascular complications in patients with MPNs. APPROACH AND RESULTS: We employed murine models of MPN in which the JAK2V617F mutation is expressed in specific cell lineages. When JAK2V617F is expressed in both blood cells and vascular ECs, the mice developed MPN and spontaneous, age-related dilated cardiomyopathy with an increased risk of sudden death as well as a prothrombotic and vasculopathy phenotype on histology evaluation. In contrast, despite having significantly higher leukocyte and platelet counts than controls, mice with JAK2V617F-mutant blood cells alone did not demonstrate any cardiac dysfunction, suggesting that JAK2V617F-mutant ECs are required for this cardiovascular disease phenotype. Furthermore, we demonstrated that the JAK2V617F mutation promotes a pro-adhesive, pro-inflammatory, and vasculopathy EC phenotype, and mutant ECs respond to flow shear differently than wild-type ECs. CONCLUSIONS: These findings suggest that the JAK2V617F mutation can alter vascular endothelial function to promote cardiovascular complications in MPNs. Therefore, targeting the MPN vasculature represents a promising new therapeutic strategy for patients with MPNs.


Asunto(s)
Cardiomiopatías , Trastornos Mieloproliferativos , Neoplasias , Trombosis , Animales , Modelos Animales de Enfermedad , Células Endoteliales , Humanos , Janus Quinasa 2/genética , Ratones , Mutación , Trastornos Mieloproliferativos/genética , Trombosis/genética
10.
J Cell Mol Med ; 24(19): 11465-11476, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32820613

RESUMEN

Fatty acid synthase (FASN) has been shown to be selectively up-regulated in cancer cells to drive the development of cancer. However, the role and associated mechanism of FASN in regulating the malignant progression of salivary adenoid cystic carcinoma (SACC) still remains unclear. In this study, we demonstrated that FASN inhibition attenuated invasion, metastasis and EMT of SACC cells as well as the expression ofPRRX1, ZEB1, Twist, Slug and Snail, among which the level of PRRX1 changed the most obviously. Overexpression of PRRX1 restored migration and invasion in FASN knockdown cells, indicating that PRRX1 is an important downstream target of FASN signalling. Levels of cyclin D1 and c-Myc, targets of Wnt/ß-catenin pathway, were significantly decreased by FASN silencing and restored by PRRX1 overexpression. In addition, FASN expression was positively associated with metastasis and poor prognosis of SACC patients as well as with the expression of PRRX1, cyclin D1 and c-Myc in SACC tissues. Our findings revealed that FASN in SACC progression may induce EMT in a PRRX1/Wnt/ß-catenin dependent manner.


Asunto(s)
Carcinoma Adenoide Quístico/patología , Transición Epitelial-Mesenquimal , Ácido Graso Sintasas/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias de las Glándulas Salivales/patología , Vía de Señalización Wnt , Animales , Apoptosis/genética , Carcinoma Adenoide Quístico/genética , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Neoplasias de las Glándulas Salivales/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cell Prolif ; 53(1): e12705, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31657086

RESUMEN

OBJECTIVES: Increasing evidences demonstrate a close correlation between epithelial-to-mesenchymal transition (EMT) induction and cancer lipid metabolism. However, the molecular mechanisms have not been clarified. MATERIALS AND METHODS: In our study, the relative expression level of PRRX1 was detected, its relationship with free fatty acid (FFA) and PPARG2 was analysed in 85 SACC tissues and 15 salivary glands from the benign salivary tumours. We also compared the FFAs composition and levels in these SACC cells. PPARG2 was detected in PRRX1-induced FFAs treatment as well as Src and MMP-9 were detected in FFAs treatment-induced invasion and migration of SACC cells, and ChIP test was performed to identify the target interactions. RESULTS: Our data showed that overexpression of PRRX1 induced EMT and facilitated the invasion and migration of SACC cells, and PRRX1 expression was closely associated with high FFAs level and poor prognosis of SACC patients. Furthermore, PRRX1 silence led to the increase of PPARG2 and the reduction of FFAs level and the migration and invasion of SACC cells. And inhibition of PPARG2 rescued FFAs level and migration and invasion capabilities of SACC cells. Free fatty acids treatment induced an increase of Stat5-DNA binding activity via Src- and MMP-9-dependent pathway. CONCLUSIONS: Collectively, our findings showed that the PRRX1/PPARG2/FFAs signalling in SACC was important for accelerating tumour metastasis through the induction of EMT and the metabolic reprogramming of FFAs.


Asunto(s)
Carcinoma Adenoide Quístico/metabolismo , Reprogramación Celular , Transición Epitelial-Mesenquimal , Ácidos Grasos/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de las Glándulas Salivales/metabolismo , Animales , Carcinoma Adenoide Quístico/genética , Carcinoma Adenoide Quístico/patología , Línea Celular Tumoral , Ácidos Grasos/genética , Proteínas de Homeodominio/genética , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/patología
12.
Chem Biol Interact ; 315: 108869, 2020 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-31682803

RESUMEN

Spermatogenic dysfunction is one of the major secondary complications of male diabetes. Salidroside (SAL) is the important active ingredients isolated from Herba Cistanche, which exhibits numerous pharmacological activities such as antioxidant, anti-diabetic, and anti-inflammatory effects. The present study was designed to determine whether SAL contributes to the recovery from spermatogenic dysfunction in streptozotocin (STZ) induced type-1 diabetic mice. SAL (25, 50, or 100 mg/kg) and Clomiphene citrate (CC, 5 mg/kg) were orally administered to male type-1 diabetic mice for 10 weeks. Testis tissues were collected for histopathological and biochemical analysis. Moreover, reproductive organ weight, sperm parameters, and testicular cell DNA damage were estimated. The results revealed that SAL significantly improved the weight of the reproductive organs, sperm parameters and testicular morphology to different degrees in type-1 diabetic mice. Furthermore, reactive oxygen species (ROS) and malondialdehyde (MDA) levels were significantly reduced, and the activities of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH), markedly increased in the testicular tissue after SAL treatment. In addition, our data also showed a marked downregulation the fluorescence expressions of p38 MAPK phosphorylation and upregulation the protein expressions of ZO-1, Occludin, Claudin-11 and N-cadherin after SAL administration (100 mg/kg) compared with the type-1 diabetic group. In conclusion, these results demonstrated that SAL exerts protective effects on type-1 diabetes-induced male spermatogenic dysfunction, which is likely mediated by inhibiting oxidative stress-mediated blood testis barrier damage.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Glucósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fenoles/farmacología , Sustancias Protectoras/farmacología , Espermatogénesis/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Barrera Hematotesticular/metabolismo , Catalasa/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Glutatión/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Recuento de Espermatozoides/métodos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Estreptozocina/farmacología , Superóxido Dismutasa/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo
13.
Cell Rep ; 29(5): 1287-1298.e6, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665640

RESUMEN

Glutamine is thought to play an important role in cancer cells by being deaminated via glutaminolysis to α-ketoglutarate (aKG) to fuel the tricarboxylic acid (TCA) cycle. Supporting this notion, aKG supplementation can restore growth/survival of glutamine-deprived cells. However, pancreatic cancers are often poorly vascularized and limited in glutamine supply, in alignment with recent concerns on the significance of glutaminolysis in pancreatic cancer. Here, we show that aKG-mediated rescue of glutamine-deprived pancreatic ductal carcinoma (PDAC) cells requires glutamate ammonia ligase (GLUL), the enzyme responsible for de novo glutamine synthesis. GLUL-deficient PDAC cells are capable of the TCA cycle but defective in aKG-coupled glutamine biosynthesis and subsequent nitrogen anabolic processes. Importantly, GLUL expression is elevated in pancreatic cancer patient samples and in mouse PDAC models. GLUL ablation suppresses the development of KrasG12D-driven murine PDAC. Therefore, GLUL-mediated glutamine biosynthesis couples the TCA cycle with nitrogen anabolism and plays a critical role in PDAC.


Asunto(s)
Carbono/metabolismo , Glutamina/metabolismo , Nitrógeno/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Eliminación de Gen , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Humanos , Ácidos Cetoglutáricos/metabolismo , Masculino , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología
14.
Biomed Pharmacother ; 120: 109474, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31585299

RESUMEN

BACKGROUND: Blood-testis barrier (BTB) impairments is one of the major secondary complications of diabetes. Betaine (BET) is the important active ingredients isolated from Lycium barbarum, which exhibits numerous pharmacological activities such as antioxidant, anti-diabetic, and anti-inflammatory effects. This study aimed to establish whether BET contributes to the recovery from BTB dysfunction in streptozotocin (STZ) induced diabetic mice. METHODS: BET (200, 400, 800 mg/kg) was orally administered to diabetic mice for 8 weeks. Testis tissues were collected for histopathological and biochemical analysis, the reproductive organ weight was estimated. Antioxidant enzyme activity and BTB associated protein expressions were determined with their corresponding assay kits and western blot analysis. The results revealed that BET significantly improved the weight of the reproductive organs and testicular morphology in diabetic mice. Furthermore, reactive oxygen species (ROS) and malondialdehyde (MDA) levels were significantly reduced, and the activities of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH), markedly increased in the testicular tissue after SAL treatment. In addition, our data also showed a marked down-regulated the expressions of p38 MAPK phosphorylation and up-regulation the protein expressions of ZO-1, Occludin, Claudin-11, N-cadherin, and Connexin-43 after BET administration compared with the diabetic group. In conclusion, these results demonstrated that BET exerts protective effects on diabetes-induced BTB dysfunction, which may be through regulating oxidative stress-mediated p38 MAPK pathways.


Asunto(s)
Betaína/farmacología , Barrera Hematotesticular/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antioxidantes/metabolismo , Barrera Hematotesticular/metabolismo , Catalasa/metabolismo , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Glutatión/metabolismo , Lycium/química , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos ICR , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo
15.
Oncotarget ; 10(42): 4307-4320, 2019 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-31303964

RESUMEN

Cell migration-inducing protein (CEMIP) and binding immunoglobulin protein (BiP) are upregulated in human cancers, where they drive cancer progression and metastasis. It has been shown that CEMIP resides in the endoplasmic reticulum (ER) where it interacts with BiP to induce cell migration, but the relationship between the two proteins was previously unknown. Here we show that CEMIP mediates activation of the BiP promoter and upregulates BiP transcript and protein levels in breast cancer cell lines. Moreover, CEMIP overexpression confers protective adaptations to cancer cells under hypoxic conditions, by decreasing apoptosis, activating autophagy, and increasing glucose uptake, to facilitate tumor growth. We demonstrate that BiP signals downstream of CEMIP, modulating cellular resistance to hypoxia. Reducing BiP in CEMIP-expressing cells sensitized cells to hypoxia treatment, decreased glucose uptake, and resulted in tumor regression in vivo. Our study provides insights into the link between CEMIP and BiP expression and the pro-survival role they play in hypoxia. Better understanding of the mechanisms behind cancer cell adaptations to harsh tumor environments could lead to development of improved cancer treatments.

16.
J Gen Physiol ; 151(8): 1051-1058, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31217223

RESUMEN

Heart rate in physiological conditions is set by the sinoatrial node (SN), the primary cardiac pacing tissue. Phosphoinositide 3-kinase (PI3K) signaling is a major regulatory pathway in all normal cells, and its dysregulation is prominent in diabetes, cancer, and heart failure. Here, we show that inhibition of PI3K slows the pacing rate of the SN in situ and in vitro and reduces the early slope of diastolic depolarization. Furthermore, inhibition of PI3K causes a negative shift in the voltage dependence of activation of the pacemaker current, I F, while addition of its second messenger, phosphatidylinositol 3,4,5-trisphosphate, induces a positive shift. These shifts in the activation of I F are independent of, and larger than, those induced by the autonomic nervous system. These results suggest that PI3K is an important regulator of heart rate, and perturbations in this signaling pathway may contribute to the development of arrhythmias.


Asunto(s)
Frecuencia Cardíaca , Fosfatidilinositol 3-Quinasas/metabolismo , Sistemas de Mensajero Secundario , Nodo Sinoatrial/fisiología , Potenciales de Acción , Animales , Relojes Biológicos , Células Cultivadas , Perros , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatos de Fosfatidilinositol/metabolismo , Conejos , Nodo Sinoatrial/metabolismo
17.
J Clin Invest ; 129(8): 3264-3276, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112530

RESUMEN

The presence of tumor-infiltrating T cells is associated with favorable patient outcomes, yet most pancreatic cancers are immunologically silent and resistant to currently available immunotherapies. Here we show using a syngeneic orthotopic implantation model of pancreatic cancer that Pik3ca regulates tumor immunogenicity. Genetic silencing of Pik3ca in KrasG12D/Trp53R172H-driven pancreatic tumors resulted in infiltration of T cells, complete tumor regression, and 100% survival of immunocompetent host mice. By contrast, Pik3ca-null tumors implanted in T cell-deficient mice progressed and killed all of the animals. Adoptive transfer of tumor antigen-experienced T cells eliminated Pik3ca-null tumors in immunodeficient mice. Loss of PIK3CA or inhibition of its effector, AKT, increased the expression of MHC Class I and CD80 on tumor cells. These changes contributed to the increased susceptibility of Pik3ca-null tumors to T cell surveillance. Our results indicate that tumor cell PIK3CA-AKT signaling limits T cell recognition and clearance of pancreatic cancer cells. Strategies that target this pathway may yield an effective immunotherapy for this cancer.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Pancreáticas/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Noqueados , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/genética , Linfocitos T/patología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Biomed Pharmacother ; 110: 561-570, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30537673

RESUMEN

Spermatogenic dysfunction is a common complication in men with diabetes and is the most important manifestation of diabetes-related male reproduction damage. Astragalin (AG) is one of the main flavonoids from Cuscuta chinensis, which has rich pharmacological activities. This study aimed to establish whether AG may contribute to the recovery from spermatogenic dysfunction. AG (3.3, 10 and 30 mg/kg) and Clomiphene (5 mg/kg) were orally administered to streptozotocin-induced diabetic male mice for 8 weeks. After the experiments performed, reproductive organs, sperm parameters and histomorphological changes were analysed. Antioxidant and anti-inflammatory capacity were estimated in testicular tissues. The results revealed that AG significantly improved the reproductive organs, sperm parameters and testicular morphology to different degrees in diabetic mice. Nitric oxide (NO) and malondialdehyde (MDA) levels were significantly reduced, and the activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT), markedly increased in the testicular tissue after AG was administered. Interestingly, AG also downregulated the protein expressions of tumour necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) in testes. In conclusion, AG is a potential beneficial agent to protect diabetic-induced spermatogenic dysfunction in male mice by increasing antioxidant enzymes activities and inhibiting inflammation.


Asunto(s)
Antioxidantes/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Mediadores de Inflamación/metabolismo , Quempferoles/uso terapéutico , Espermatogénesis/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Quempferoles/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Distribución Aleatoria , Espermatogénesis/fisiología , Resultado del Tratamiento
19.
Chem Biol Interact ; 297: 119-129, 2019 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-30365938

RESUMEN

Diabetes-associated sexual dysfunction and fertility impairments are major secondary complications in diabetic patients and animal models. Natural herbs are important sources of therapeutic agents for diabetic complications. This study investigated the effect of vitexin on male sexual dysfunction and fertility impairments in streptozotocin (STZ)-induced diabetic mice. Diabetes was induced by intraperitoneal injection of 45 mg/kg STZ for 5 consecutive days in mice. Vitexin (10, 20 or 40 mg/kg) and Sildenafil citrate (SC, 5 mg/kg) were administered daily for 62 days after the induction of diabetes. The parameters of sexual behavior and fertility were analyzed. The reproductive organ weight, sperm motility, and viability of the treated mice were examined. Testicular histopathological alterations were detected by hematoxylin and eosin (H&E) staining. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate serum hormonal levels. Results showed that 40 mg/kg vitexin significantly improved the sexual behavior and fertility levels compared with the diabetic group. Moreover, vitexin (20 or 40 mg/kg) significantly increased reproductive organ weight and improved testicular pathological structure damage. Meanwhile, sperm analysis demonstrated that vitexin significantly restored sperm quality in a dose-dependent manner. Furthermore, ELISA data showed that vitexin significantly increased the serum testosterone (T), follicular-stimulating hormone (FSH), and luteinizing hormone (LH) levels but decreased the gonadotropin-releasing hormone (GnRH) level to different degrees. These findings suggest that vitexin ameliorates sexual dysfunction and fertility impairments in male diabetic mice possibly by modulating the hypothalamus-pituitary-gonadal axis.


Asunto(s)
Apigenina/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Fertilidad/efectos de los fármacos , Gónadas/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipófisis/efectos de los fármacos , Disfunciones Sexuales Fisiológicas/tratamiento farmacológico , Animales , Apigenina/administración & dosificación , Apigenina/química , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Ratones , Estructura Molecular , Disfunciones Sexuales Fisiológicas/inducido químicamente , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Estreptozocina/administración & dosificación , Relación Estructura-Actividad
20.
Cell Tissue Res ; 374(3): 653-666, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30073544

RESUMEN

Testicular dysfunction is one of the serious secondary complications in diabetes. Lycium barbarum polysaccharide (LBP) has long been considered to possess a wide range of beneficial properties including antiaging, anticancer and reproductive-enhancing. Abnormal autophagy was reported to play a significant role in accelerating diabetic reproductive injury. However, the autophagy regulation mechanism of LBP on diabetic testicular dysfunction is incompletely understood. We investigate the protective effects of LBP on diabetic testicular dysfunction and its underlying mechanism with different approaches. Protective effects of LBP (40 mg/kg) on testicular functions were assessed through the use of sperm parameters, testosterone levels and hematoxylin and eosin staining. Antioxidant capacity and serum malondialdehyde levels were determined using assay kits. Immune intensity of Beclin-1 and LC3I in testes was detected by immunofluorescence staining. Western blot analysis was used to detect expressions of p-PI3K, Akt, p-Akt, Beclin-1, LC3I and LC3II proteins. Q-PCR was used to evaluate Beclin-1 and LC3I mRNA expressions in testis. Administration of LBP (40 mg/kg) considerably recovered testicular function, obviously improved testicular histopathologic structure and significantly increased antioxidant enzyme activities. Immunofluorescence staining showed that immune intensity of Beclin-1 and LC3I significantly decreased in the LBP 40 mg/kg group. The results of Q-PCR and western blot analysis showed that LBP 40 mg/kg significantly downregulated Beclin-1 and LC3I protein expressions upregulated p-PI3K and p-Akt protein expressions and decreased Beclin-1 and LC3I mRNA expressions compared with diabetic mice. In conclusion, inhibition of PI3K/Akt pathway-mediated testicular excessive autophagy may be a target for protective effects of LBP on diabetic testicular dysfunction.


Asunto(s)
Autofagia , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/fisiopatología , Medicamentos Herbarios Chinos/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testículo/patología , Testículo/fisiopatología , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Medicamentos Herbarios Chinos/farmacología , Masculino , Ratones Endogámicos ICR , Proteínas Asociadas a Microtúbulos/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Testículo/efectos de los fármacos , Testosterona/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...