Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.419
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39273316

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that is almost entirely resistant to conventional chemotherapy and radiation therapy. A significant factor in this resistance appears to be the dense desmoplastic stroma, which contains various cancer-associated fibroblast (CAF) populations. However, our understanding of the communication between tumor cells and CAFs that contributes to this aggressive malignancy is still developing. Recently, we used an advanced three-dimensional heterospecies, heterospheroid co-culture model to investigate the signaling between human pancreatic tumor Panc1 cells and mouse pancreatic stellate cells (mPSCs) through global expression profiling. Upon discovering that CCN1 was significantly upregulated in Panc1 cells during co-culture, we decided to explore the role of CCN1 using CRISPR-Cas9 knockout technology. Panc1 cells lacking CCN1 showed reduced differentiation and decreased sensitivity to gemcitabine, primarily due to lower expression of genes involved in gemcitabine transport and metabolism. Additionally, we observed that stimulation with TGF-ß1 and lysophosphatidic acid increased CCN1 expression in Panc1 cells and induced a shift in mPSCs towards a more myofibroblastic CAF-like phenotype.


Asunto(s)
Técnicas de Cocultivo , Proteína 61 Rica en Cisteína , Desoxicitidina , Gemcitabina , Neoplasias Pancreáticas , Células Estrelladas Pancreáticas , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Proteína 61 Rica en Cisteína/metabolismo , Proteína 61 Rica en Cisteína/genética , Humanos , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Ratones , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos
2.
Int J Mol Sci ; 25(15)2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39125841

RESUMEN

Glioblastoma multiforme is the most common and fatal brain tumor among human cancers. Ceramide (Cer) and Sphingosine 1-phosphate (S1P) have emerged as bioeffector molecules that control several biological processes involved in both cancer development and resistance. Cer acts as a tumor suppressor, inhibiting cancer progression, promoting apoptosis, enhancing immunotherapy and sensitizing cells to chemotherapy. In contrast, S1P functions as an onco-promoter molecule, increasing proliferation, survival, invasiveness, and resistance to drug-induced apoptosis. The pro-survival PI3K/Akt pathway is a recognized downstream target of S1P, and we have previously demonstrated that in glioma cells it also improves Cer transport and metabolism towards complex sphingolipids in glioma cells. Here, we first examined the possibility that, in T98G glioma cells, S1P may regulate Cer metabolism through PI3K/Akt signaling. Our research showed that exogenous S1P increases the rate of vesicular trafficking of Cer from the endoplasmic reticulum (ER) to the Golgi apparatus through S1P receptor-mediated activation of the PI3K/Akt pathway. Interestingly, the effect of S1P results in cell protection against toxicity arising from Cer accumulation in the ER, highlighting the role of S1P as a survival factor to escape from the Cer-generating cell death response.


Asunto(s)
Supervivencia Celular , Ceramidas , Retículo Endoplásmico , Glioma , Aparato de Golgi , Lisofosfolípidos , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Esfingosina , Humanos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Ceramidas/metabolismo , Ceramidas/farmacología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Aparato de Golgi/metabolismo , Aparato de Golgi/efectos de los fármacos , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma/metabolismo , Glioma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Apoptosis/efectos de los fármacos
3.
Biomolecules ; 14(6)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38927035

RESUMEN

Lysophosphatidic acid (LPA) is a well-documented pro-oncogenic factor in different cancers, but relatively little is known on its biological activity in neuroblastoma. The LPA effects and the participation of the tyrosine kinase receptor anaplastic lymphoma kinase (ALK) in LPA mitogenic signaling were studied in human neuroblastoma cell lines. We used light microscopy and [3H]-thymidine incorporation to determine cell proliferation, Western blot to study intracellular signaling, and pharmacological and molecular tools to examine the role of ALK. We found that LPA stimulated the growth of human neuroblastoma cells, as indicated by the enhanced cell number, clonogenic activity, and DNA synthesis. These effects were curtailed by the selective ALK inhibitors NPV-TAE684 and alectinib. In a panel of human neuroblastoma cell lines harboring different ALK genomic status, the ALK inhibitors suppressed LPA-induced phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2), which are major regulators of cell proliferation. ALK depletion by siRNA treatment attenuated LPA-induced ERK1/2 activation. LPA enhanced ALK phosphorylation and potentiated ALK activation by the ALK ligand FAM150B. LPA enhanced the inhibitory phosphorylation of the tumor suppressor FoxO3a, and this response was impaired by the ALK inhibitors. These results indicate that LPA stimulates mitogenesis of human neuroblastoma cells through a crosstalk with ALK.


Asunto(s)
Quinasa de Linfoma Anaplásico , Proliferación Celular , Lisofosfolípidos , Neuroblastoma , Transducción de Señal , Humanos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Quinasa de Linfoma Anaplásico/metabolismo , Quinasa de Linfoma Anaplásico/genética , Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Carbazoles/farmacología , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos
4.
J Gene Med ; 26(6): e3708, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38837511

RESUMEN

BACKGROUND: Lysophosphatidic acid (LPA) is a small bioactive lipid which acts as a potent regulator in various tumor progressions through six G-protein-coupled receptors (LPA1-LPA6). Our previous study demonstrated that the LPA-producing enzyme, autotaxin (ATX), was upregulated in esophageal squamous cell carcinoma (ESCC) and ATX high expression levels indicated a poor prognosis. Esophageal squamous cell carcinoma is a type of malignant tumor which originates from epithelial cells. Its progression can be affected by the interaction between cancer cells and normal cells. However, the impact of LPA on the interaction between esophageal epithelial cells and cancer cells in the development of ESCC remains uncertain. METHODS: MTS and Edu assays were performed to determine ESCC cell proliferation in culture medium (CM) derived from LPA-stimulated esophageal epithelial cells (Het-1a). A wound healing assay, transwell migration and an invasion assay were performed to assess the metastatic ability of ESCC cells. Cytokine array analysis was conducted to detect the differentially secreted cytokines in CM. The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were utilized to uncover the pathways and cytokines that are influenced by LPA in ESCC. Immunohistochemical staining was employed to measure the expression of ATX and CCL2 in early-stage ESCC. Quantitative real-time PCR, western blot, enzyme-linked immunosorbent assay and an antibody neutralization assay were employed to measure the mechanism of LPA-mediated communication between epithelial cells and cancer cells. RESULTS: Functional experiments showed that exposing ESCC cancer cells to CM from LPA-treated Het-1a results in promoting proliferation, migration, invasion and epithelial-mesenchymal transition processes. Using cytokine array analysis, we discovered that LPA triggers the release of multiple cytokines from epithelial cells. After screening of the TCGA and GEO databases, CCL2 was identified and found to be correlated with ATX expression in ESCC. Furthermore, CCL2 levels in both mRNA expression and secretion were observed to be upregulated in epithelial cells upon stimulation with LPA. Blocking CCL2 effectively reduced the pro-migration influence of CM derived from LPA-treated Het-1a. Mechanism studies have demonstrated that LPA activated the NF-κB signaling pathway through LPA1/3, ultimately causing an increase in CCL2 expression and secretion in Het-1a. CONCLUSIONS: Our findings, taken together, demonstrate that CM from LPA-treated esophageal epithelial cells plays a significant role in promoting the progression of ESCC, with CCL2 acting as the primary regulator.


Asunto(s)
Movimiento Celular , Proliferación Celular , Quimiocina CCL2 , Células Epiteliales , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Regulación Neoplásica de la Expresión Génica , Lisofosfolípidos , Humanos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Progresión de la Enfermedad , Transducción de Señal/efectos de los fármacos , Esófago/metabolismo , Esófago/patología , Esófago/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos
5.
J Surg Res ; 300: 25-32, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38795670

RESUMEN

INTRODUCTION: Previous literature suggests that sphingolipids may impact systemic coagulation and platelet aggregation, thus modulating the risks of thrombotic events. The goal of this investigation was to evaluate the role of serum sphingolipids on intrinsic platelet function to assess whether pharmacologic manipulation of sphingolipid metabolites would impact platelet aggregability. METHODS: C57BL/6J mice were injected with either normal saline, 1 mg/kg FTY720 (synthetic sphingosine-1-phosphate [S1P] receptor analog), or 5 mg/kg SLM6031434 (sphingosine kinase two inhibitor). Mice were sacrificed at 6 h and whole blood (WB) was collected for impedance aggregometry assessing platelet responsiveness to arachidonic acid or adenosine diphosphate. Ex vivo studies utilized WB or platelet-rich plasma that was pretreated with S1P, FTY720, amitriptyline, or d-sphingosine then analyzed by aggregability and flow cytometry for platelet and platelet-derived microvesicle characteristics. RESULTS: FTY720 and SLM6031434 pretreated induced similar arachidonic acid and adenosine diphosphate-mediated platelet aggregation as controls. Ex vivo WB and platelet-rich plasma treatment with S1P, FTY720, amitriptyline and d-sphingosine did not impact platelet aggregation. The percentages of CD41+, CD62P+ and CD41+/ceramide+, CD62P+/ceramide + platelets, and platelet-derived microvesicle were not significantly different between amitriptyline-treated and normal saline-treated cohorts. CONCLUSIONS: Sphingolipid modulating agents, such as FTY720, SLM6031434, S1P, amitriptyline, ceramide, and d-sphingosine do not appear to independently impact platelet aggregation in murine models.


Asunto(s)
Plaquetas , Clorhidrato de Fingolimod , Ratones Endogámicos C57BL , Agregación Plaquetaria , Esfingolípidos , Esfingosina , Animales , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Esfingosina/análogos & derivados , Esfingosina/sangre , Ratones , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Esfingolípidos/sangre , Esfingolípidos/metabolismo , Masculino , Lisofosfolípidos/farmacología , Lisofosfolípidos/sangre , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Ácido Araquidónico/farmacología , Amitriptilina/farmacología , Adenosina Difosfato/farmacología
6.
Pathol Res Pract ; 257: 155293, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615508

RESUMEN

Lysophosphatidic acid (LPA) binds to its specific G protein-coupled LPA receptors (LPA1 to LPA6), resulting in the activation of various cellular functions. LPA receptor-mediated signaling facilitates tumor progression in human malignancies. In the present study, we investigated whether LPA receptor-mediated signaling contributes to cellular responses to X-ray irradiation in osteosarcoma MG-63 cells. After X-ray irradiation (2, 4 and 8 Gy), LPAR2 and LPAR3 expression levels in MG-63 cells were significantly elevated in a dose-dependent manner, but no change of LPAR1 expression level was observed. The cell growth activities of MG-63 cells irradiated with X-rays (2, 4 and 8 Gy) were reduced by LPA. Conversely, LPA3 agonist (2 S)-OMPT enhanced the cell growth activities of X-ray irradiated MG-63 cells. The cell movement of MG-63 cells exposed to X-ray irradiation (8 Gy) was inhibited by (2 S)OMPT. In cell survival assay, (2 S)-OMPT suppressed the cell survival to cisplatin (CDDP) of MG-63 cells irradiated with X-rays (8 Gy). The cell survival to CDDP of X-ray irradiated cells was elevated by LPA3 knockdown. Moreover, we evaluated the effects of LPA2 on the cell survival to CDDP of MG-63 cells exposed to X-ray irradiation (8 Gy). The cell survival to CDDP of X-ray irradiated cells was increased by LPA2 agonist GRI-977143 and reduced by LPA2 knockdown. These results suggest that LPA receptor-signaling participates in the modulation of cellular functions induced by X-ray irradiation in osteosarcoma cells.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Receptores del Ácido Lisofosfatídico , Humanos , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/radioterapia , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores del Ácido Lisofosfatídico/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Rayos X
7.
Biol Chem ; 405(4): 267-281, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38081222

RESUMEN

Celastrol (Cel) shows potent antitumor activity in various experimental models. This study examined the relationship between Cel's antivascular and antitumor effects and sphingolipids. CCK-8 assay, transwell assay, Matrigel, PCR-array/RT-PCR/western blotting/immunohistochemistry assay, ELISA and HE staining were used to detect cell proliferation, migration and invasion, adhesion and angiogenesis, mRNA and protein expression, S1P production and tumor morphology. The results showed that Cel could inhibit proliferation, migration or invasion, adhesion and angiogenesis of human umbilical vein endothelial cells (HUVECs) and MDA-MB-231 cells by downregulating the expression of degenerative spermatocyte homolog 1 (DEGS1). Transfection experiments showed that downregulation of DEGS1 inhibited the above processes and sphingosine-1-phosphate (S1P) production of HUVECs and MDA-MB-231 cells, while upregulation of DEGS1 had the opposite effects. Coculture experiments showed that HUVECs could promote proliferation, migration and invasion of MDA-MB-231 cells through S1P/sphingosine-1-phosphate receptor (S1PR) signaling pathway, while Cel inhibited these processes in MDA-MB-231 cells induced by HUVECs. Animal experiments showed that Cel could inhibit tumor growth in nude mice. Western blotting, immunohistochemistry and ELISA assay showed that Cel downregulated the expression of DEGS1, CD146, S1PR1-3 and S1P production. These data confirm that DEGS1/S1P signaling pathway may be related to the antivascular and antitumor effects of cel.


Asunto(s)
Fenómenos Biológicos , Triterpenos Pentacíclicos , Receptores de Lisoesfingolípidos , Esfingosina/análogos & derivados , Ratones , Animales , Humanos , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Células MDA-MB-231 , Angiogénesis , Ratones Desnudos , Transducción de Señal , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Esfingosina/farmacología , Esfingosina/metabolismo , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo
8.
Sci Rep ; 13(1): 19716, 2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37953311

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by an increase in hepatic lipid accumulation due to impaired lipid metabolism. Although a correlation was found between NAFLD and sphingosine-1-phosphate (S1P), the role of the sphingolipid remains controversial. The aim of this study was to investigate any involvement of S1P in steatosis using its analog FTY720P and HepG2 cells. Lipid accumulation was induced by incubating the cells in a mixture of oleic and palmitic acid, and was quantified using Oil Red O. The involvement of signaling mediators was studied using pharmacological inhibitors and western blot analysis. FTY720P increased lipid accumulation, but this increase wasn't maintained in the presence of inhibitors of S1PR3, Gq, SREBP, mTOR, PI3K, and PPARγ indicating their involvement in the process. The results revealed that FTY720P binds to S1PR3 which activates sequentially Gq, PI3K, and mTOR leading to an increase in SREBP expression and PPARγ activation. It was concluded that in presence of a high level of fatty acids, lipid accumulation is increased in hepatocytes by the exogenously added FTY720P.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Células Hep G2 , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , PPAR gamma/metabolismo , Hígado/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Metabolismo de los Lípidos , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
9.
Int J Mol Sci ; 24(22)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-38003272

RESUMEN

Mechanical ventilation (MV) is a life-supporting strategy employed in the Intensive Care Unit (ICU). However, MV-associated mechanical stress exacerbates existing lung inflammation in ICU patients, resulting in limited improvement in mortality and a condition known as Ventilator-Induced Lung Injury (VILI). Sphingosine-1-phosphate (S1P) is a circulating bioactive lipid that maintains endothelial integrity primarily through S1P receptor 1 (S1PR1). During VILI, mechanical stress upregulates endothelial S1PR3 levels. Unlike S1PR1, S1PR3 mediates endothelial barrier disruption through Rho-dependent pathways. However, the specific impact of elevated S1PR3 on lung endothelial function, apart from Rho activation, remains poorly understood. In this study, we investigated the effects of S1PR3 in endothelial pathobiology during VILI using an S1PR3 overexpression adenovirus. S1PR3 overexpression caused cytoskeleton rearrangement, formation of paracellular gaps, and a modified endothelial response towards S1P. It resulted in a shift from S1PR1-dependent barrier enhancement to S1PR3-dependent barrier disruption. Moreover, S1PR3 overexpression induced an ADAM10-dependent cleavage of Vascular Endothelial (VE)-cadherin, which hindered endothelial barrier recovery. S1PR3-induced cleavage of VE-cadherin was at least partially regulated by S1PR3-mediated NFκB activation. Additionally, we employed an S1PR3 inhibitor TY-52156 in a murine model of VILI. TY-52156 effectively attenuated VILI-induced increases in bronchoalveolar lavage cell counts and protein concentration, suppressed the release of pro-inflammatory cytokines, and inhibited lung inflammation as assessed via a histological evaluation. These findings confirm that mechanical stress associated with VILI increases S1PR3 levels, thereby altering the pulmonary endothelial response towards S1P and impairing barrier recovery. Inhibiting S1PR3 is validated as an effective therapeutic strategy for VILI.


Asunto(s)
Neumonía , Lesión Pulmonar Inducida por Ventilación Mecánica , Humanos , Ratones , Animales , Receptores de Esfingosina-1-Fosfato , Cadherinas , Esfingosina/farmacología , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lisofosfolípidos/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Proteína ADAM10 , Proteínas de la Membrana , Secretasas de la Proteína Precursora del Amiloide
10.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37870076

RESUMEN

Improvement of nutrient utilization to promote growth performance is always pursued in poultry. In this study, a total of 360 1-d-old male ducklings was randomly assigned to 3 treatments in terms of diet treatment groups. Three treatments were as follows: basal diet (Con group) or basal diet supplemented with 300 mg/kg multi-enzymes (ENZ group) or 500 mg/kg lysophospholipids (LPL group). On day 42, ducks were slaughtered for samplings. The results revealed that supplementary LPL improved the body weight (BW) at day 14 and average daily gain (ADG) during days 1 to 14 and improved the feed conversion rate (FCR) for the overall period (P < 0.05) by improving nutrient utilization of dry matter and ether extract (P < 0.05) compared with the Con group. Dietary ENZ improved the FCR from days 15-42 and 1-42, and nitrogen utilization (P < 0.05) compared with the Con group. Jejunal villus height and villus height/crypt depth ratio were higher (P < 0.05) in the LPL group and tended to be higher (P < 0.1) in the ENZ group compared to the Con group. Supplementation with either LPL or ENZ reduced interleukin-1ß concentration in jejunal mucus (P < 0.05). Both LPL and ENZ enhanced serum total superoxide dismutase activity (P < 0.05), whereas only supplementation with LPL elevated total antioxidant capacity (P < 0.05). In terms of cecal microbiota, microbial richness tended to be reduced by LPL, with low observed-OTUs and Chao1 (0.05 < P < 0.1). Supplementation with ENZ led to higher abundances of cellulolytic bacteria such as Fibrobacterota, [Eubacterium]_xylanophilum_group, and Bifidobacterium. Overall, both LPL and ENZ improved FCR, which may be relevant to ameliorative intestinal health, overall antioxidant ability, and cecal microbiome.


Well known in the industry, enhancing nutrient utilization in meat ducks is a vital sustainability tactic to manage production costs. This is especially relevant because meat ducks require more feed, and grain prices are on the rise. Lysophospholipids (LPL) have been confirmed to effectively emulsify fat, which boosts fat utilization. Additionally, multi-exogenous enzymes (ENZ) play a significant role in nutrient breakdown. Our feeding experiment on Cherry Vallery male ducks demonstrated that a dietary supplement of LPL at 500 mg/kg improves the body weight, average daily gain, and feed conversion rate during the starter period. It also elevates the feed conversion rate over the entire period, enhances ether extract utilization, and positively impacts jejunal morphology development in the finishing phase. However, LPL negatively affects the α-diversity of cecal flora. On the other hand, supplementing with 300 mg/kg ENZ improves the feed conversion rate throughout the period, increases nitrogen utilization in the finisher phase, diminishes interleukin-1ß levels in the jejunum, elevates superoxide dismutase in the serum, and promotes the prevalence of cellulolytic bacteria. In summary, feed supplemented with 500 mg/kg LPL and 300 mg/kg ENZ aids in reducing the FCR of meat ducks.


Asunto(s)
Antioxidantes , Microbioma Gastrointestinal , Masculino , Animales , Antioxidantes/farmacología , Patos , Suplementos Dietéticos/análisis , Dieta/veterinaria , Lisofosfolípidos/farmacología , Alimentación Animal/análisis
11.
Biochem Biophys Res Commun ; 681: 186-193, 2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37783116

RESUMEN

Primary cilia are essential cellular antennae that transmit external signals into intracellular responses. These sensory organelles perform crucial tasks in triggering intracellular signaling pathways, including those initiated by G protein-coupled receptors (GPCRs). Given the involvement of GPCRs in serum-induced signaling, we investigated the contribution of ciliary proteins in mitogen perception and cell proliferation. We found that depletion of cilia via IFT88 silencing impaired cell growth and repressed YAP activation against serum and its mitogenic constituents, namely lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). To identify the key player of serum mitogen signaling, a mutant cell line library with 30 ablated individual ciliary proteins was established and screened based on YAP dephosphorylation and target gene induction. While 9 of them had altered signaling, ablation of IFT38 or IFT144 led to a particularly robust repression of YAP activation upon LPA and S1P. The deficiency of IFT38 and IFT144 attenuated cell proliferation, as corroborated in either 2-dimensional cultures or tumor spheroids. In subcutaneous skin melanoma patients, expression of IFT38 and IFT144 was associated with unfavorable outcomes in overall survival. In conclusion, our study demonstrates the involvement of ciliary proteins in mitogen signaling and identifies the regulatory roles of IFT38 and IFT144 in serum-mediated Hippo pathway signaling and cellular growth.


Asunto(s)
Mitógenos , Transducción de Señal , Humanos , Línea Celular , Proliferación Celular , Lisofosfolípidos/farmacología , Receptores Acoplados a Proteínas G/metabolismo
12.
Cells ; 12(17)2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37681929

RESUMEN

Lysophosphatidic acid (LPA) signalling is essential for maintaining germ cell viability during mouse spermatogenesis; however, its role in human spermatozoa is unknown. We previously demonstrated that peroxiredoxin 6 (PRDX6) calcium-independent phospholipase A2 (iPLA2) releases lysophospholipids such as LPA or arachidonic acid (AA) and that inhibiting PRDX6 iPLA2 activity impairs sperm cell viability. The exogenous addition of LPA bypassed the inhibition of PRDX6 iPLA2 activity and maintained the active phosphoinositide 3-kinase (PI3K)/AKT pathway. Here, we aimed to study PI3K/AKT pathway regulation via LPA signalling and protein kinases in maintaining sperm viability. The localization of LPARs in human spermatozoa was determined using immunocytochemistry, and P-PI3K and P-AKT substrate phosphorylations via immunoblotting. Sperm viability was determined using the hypo-osmotic swelling test. LPAR1, 3, 5 and 6 were located on the sperm plasma membrane. The inhibition of LPAR1-3 with Ki16425 promoted the impairment of sperm viability and decreased the phosphorylation of PI3K AKT substrates. Inhibitors of PKC, receptor-type PTK and PLC impaired sperm viability and the PI3K/AKT pathway. Adding 1-oleoyl-2-acetyl-snglycerol (OAG), a cell-permeable analog of diacylglycerol (DAG), prevented the loss of sperm viability and maintained the phosphorylation of PI3K. In conclusion, human sperm viability is supported by LPAR signalling and regulated by PLC, PKC and RT-PTK by maintaining phosphorylation levels of PI3K and AKT substrates.


Asunto(s)
Lisofosfolípidos , Fosfatidilinositol 3-Quinasas , Humanos , Masculino , Lisofosfolípidos/farmacología , Peroxiredoxina VI , Proteínas Proto-Oncogénicas c-akt , Semen
13.
Eur J Pharmacol ; 954: 175893, 2023 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-37392830

RESUMEN

GPR82 is an orphan G protein-coupled receptor (GPCR) that has been implicated in lipid storage in mouse adipocytes. However, the intracellular signaling as well as the specific ligands of GPR82 remain unknown. GPR82 is closely related to GPR34, a GPCR for the bioactive lipid molecule lysophosphatidylserine. In this study, we screened a lipid library using GPR82-transfected cells to search for ligands that act on GPR82. By measuring cyclic adenosine monophosphate levels, we found that GPR82 is an apparently constitutively active GPCR that leads to Gi protein activation. In addition, edelfosine (1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine), an artificial lysophospholipid with a cationic head group that exerts antitumor activity, inhibited the Gi protein activation by GPR82. Two endogenous lysophospholipids with cationic head groups, lysophosphatidylcholine (1-oleoyl-sn-glycero-3-phosphocholine) and lysophosphatidylethanolamine (1-oleoyl-sn-glycero-3-phosphoethanolamine), also exhibited GPR82 inhibitory activity, albeit weaker than edelfosine. Förster resonance energy transfer imaging analysis consistently demonstrated that Gi protein-coupled GPR82 has an apparent constitutive activity that is edelfosine-sensitive. Consistent data were obtained from GPR82-mediated binding analysis of guanosine-5'-O-(3-thiotriphosphate) to cell membranes. Furthermore, in GPR82-transfected cells, edelfosine inhibited insulin-induced extracellular signal-regulated kinase activation, like compounds that function as inverse agonists at other GPCRs. Therefore, edelfosine is likely to act as an inverse agonist of GPR82. Finally, GPR82 expression inhibited adipocyte lipolysis, which was abrogated by edelfosine. Our findings suggested that the cationic lysophospholipids edelfosine, lysophosphatidylcholine and lysophosphatidylethanolamine are novel inverse agonists for Gi-coupled GPR82, which is apparently constitutively active, and has the potential to exert lipolytic effects through GPR82.


Asunto(s)
Agonismo Inverso de Drogas , Lisofosfatidilcolinas , Animales , Ratones , Ligandos , Fosforilcolina , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo
14.
Biochem Biophys Res Commun ; 666: 154-161, 2023 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-37187093

RESUMEN

Cannabinoid receptor 2 (CB2) and lysophosphatidic acid receptor 5 (LPA5) are both classified as G-protein coupled receptors (GPCRs) activated by bioactive lipids and are highly expressed in colon cancer cells. However, crosstalk between two receptors and its potential effects on cancer cell physiology have not been fully elucidated. In the present study, the results of bioluminescence resonance energy transfer analysis showed that, among the LPA receptors, CB2 strongly and specifically interacted with LPA5. Both receptors were co-localized in the plasma membrane in the absence of agonists, and the receptors were co-internalized upon activation of either receptor alone or both receptors together. We further investigated the effects of expression of both receptors on cell proliferation and migration, and the molecular mechanisms underlying these effects in HCT116 colon cancer cells. Co-expression of receptors significantly increased cell proliferation and migration by increasing Akt phosphorylation and tumor progression-related gene expression, whereas no such effect was seen upon expression of either receptor alone. These results suggest the possibility of physical and functional crosstalk between CB2 and LPA5.


Asunto(s)
Neoplasias del Colon , Receptores del Ácido Lisofosfatídico , Humanos , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/patología , Lisofosfolípidos/farmacología , Receptores de Cannabinoides , Receptores del Ácido Lisofosfatídico/metabolismo , Células HCT116
15.
J Bioenerg Biomembr ; 55(3): 169-177, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37219648

RESUMEN

The tumor microenvironment (TME) consists of various cell types, including fibroblasts. The TME plays a central role in the promotion of tumor progression. In the present study, we investigated whether lysophosphatidic acid (LPA) receptor-mediated signaling regulates cellular functions by the TME of pancreatic cancer PANC-1 cells. To obtain fibroblast 3T3 cell supernatants, 3T3 cells were cultured in 5% charcoal stripped FCS-DMEM for 48 h. LPAR2 and LPAR3 expression levels were elevated in PANC-1 cells cultured in 3T3 cell supernatants. While PANC-1 cell motility was decreased by 3T3 cell supernatants, the cell survival to cisplatin (CDDP) of PANC-1 cells was markedly enhanced. Moreover, the cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants was increased by GRI-977,143 (LPA2 agonist) and (2 S)-OMPT (LPA3 agonist). Since hypoxia is caused by the restriction of adequate vascular networks to deliver oxygen into solid tumors, PANC-1 cells were cultured in 3T3 cell supernatants at 1% O2 conditions. The cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants at 1% O2 was significantly elevated, correlating with LPAR2 and LPAR3 expressions. These results suggest that LPA signaling via LPA2 and LPA3 is involved in the promotion of malignant properties by the TME in PANC-1 cells.


Asunto(s)
Neoplasias Pancreáticas , Receptores del Ácido Lisofosfatídico , Ratones , Animales , Humanos , Receptores del Ácido Lisofosfatídico/metabolismo , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Cisplatino/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Movimiento Celular , Hipoxia/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
16.
J Biochem ; 174(3): 253-266, 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37098187

RESUMEN

Sphingosine 1-phosphate (S1P) is one of the lipid mediators involved in diverse physiological functions. S1P circulates in blood and lymph bound to carrier proteins. Three S1P carrier proteins have been reported, albumin, apolipoprotein M (ApoM) and apolipoprotein A4 (ApoA4). The carrier-bound S1P exerts its functions via specific S1P receptors (S1PR1-5) on target cells. Previous studies showed several differences in physiological functions between albumin-bound S1P and ApoM-bound S1P. However, molecular mechanisms underlying the carrier-dependent differences have not been clarified. In addition, ApoA4 is a recently identified S1P carrier protein, and its functional differences from albumin and ApoM have not been addressed. Here, we compared the three carrier proteins in the processes of S1P degradation, release from S1P-producing cells and receptor activation. ApoM retained S1P more stable than albumin and ApoA4 in the cell culture medium when compared in the equimolar amounts. ApoM facilitated theS1P release from endothelial cells most efficiently. Furthermore, ApoM-bound S1P showed a tendency to induce prolonged activation of Akt via S1PR1 and S1PR3. These results suggest that the carrier-dependent functional differences of S1P are partly ascribed to the differences in the S1P stability, S1P-releasing efficiency and signaling duration.


Asunto(s)
Lisofosfolípidos , Proteínas Proto-Oncogénicas c-akt , Humanos , Apolipoproteínas M/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Lisofosfolípidos/farmacología , Esfingosina/farmacología , Proteínas Portadoras/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Albúminas/metabolismo
17.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37047288

RESUMEN

Mast cells (MCs) are the main participants in the control of immune reactions associated with inflammation, allergies, defense against pathogens, and tumor growth. Bioactive lipids are lipophilic compounds able to modulate MC activation. Here, we explored some of the effects of the bioactive lipid lysophosphatidylinositol (LPI) on MCs. Utilizing murine bone marrow-derived mast cells (BMMCs), we found that LPI did not cause degranulation, but slightly increased FcεRI-dependent ß-hexosaminidase release. However, LPI induced strong chemotaxis together with changes in LIM kinase (LIMK) and cofilin phosphorylation. LPI also promoted modifications to actin cytoskeleton dynamics that were detected by an increase in cell size and interruptions in the continuity of the cortical actin ring. The chemotaxis and cortical actin ring changes were dependent on GPR55 receptor activation, since the specific agonist O1602 mimicked the effects of LPI and the selective antagonist ML193 prevented them. The LPI and O1602-dependent stimulation of BMMC also led to VEGF, TNF, IL-1α, and IL-1ß mRNA accumulation, but, in contrast with chemotaxis-related processes, the effects on cytokine transcription were dependent on GPR55 and cannabinoid (CB) 2 receptors, since they were sensitive to ML193 and to the specific CB2 receptor antagonist AM630. Remarkably, GPR55-dependent BMMC chemotaxis was observed towards conditioned media from distinct mouse and human cancer cells. Our data suggest that LPI induces the chemotaxis of MCs and leads to cytokine production in MC in vitro with the differential participation of GPR55 and CB2 receptors. These effects could play a significant role in the recruitment of MCs to tumors and the production of MC-derived pro-angiogenic factors in the tumor microenvironment.


Asunto(s)
Receptor Cannabinoide CB2 , Receptores Acoplados a Proteínas G , Ratones , Humanos , Animales , Receptores Acoplados a Proteínas G/genética , Receptor Cannabinoide CB2/genética , Quimiotaxis , Mastocitos , Citocinas , Actinas , Receptores de Cannabinoides/genética , Lisofosfolípidos/farmacología , Lisofosfolípidos/fisiología
18.
J Reprod Dev ; 69(3): 163-169, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37045747

RESUMEN

Lysophosphatidic acid (LPA) has been implicated in the uterine endometrial functions of implantation and decidualization; however, not much is known about its myometrial contractile function. Herein we characterized the uterotonic effects of LPA in non-pregnant (estrus) and peri-parturient rats in vitro. LPA dose-dependently (0.01-10 µM) stimulated the amplitude and integral, but not the frequency, of the uterine strip contraction of estrous rats. The stimulatory effect of LPA was enhanced 1 day before parturition but was lost 1 day postpartum. LPA did not cause the de novo synthesis of prostaglandin (PG) F2α but stimulated contractions cooperatively with the PG. LPA-induced contractions were significantly inhibited by an LPA1/2/3 antagonist in the uteri of estrous rats but not in term rats. This study characterized the uterotonic effect of a natural LPA that occurs at physiological concentrations, changes with reproductive states, and is independent of mediation by the newly synthesized PG.


Asunto(s)
Contracción Uterina , Útero , Embarazo , Femenino , Ratas , Animales , Lisofosfolípidos/farmacología , Endometrio , Receptores del Ácido Lisofosfatídico
19.
Biochem Biophys Res Commun ; 661: 21-27, 2023 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-37084489

RESUMEN

In tumor microenvironment, cancer cells can adapt to low conditions of nutrients and oxygen. Lysophosphatidic acid (LPA) receptor-mediated signaling is involved in the promotion of malignant properties in cancer cells. In the present study, to examine the roles of LPA receptors in the regulation of cell motility and survival to cisplatin (CDDP) of pancreatic cancer PANC-1 cells under glucose-deprived and hypoxic conditions, cells were cultured in 4500 mg/L high glucose (HG)-DMEM, 500 mg/L middle glucose (MG)-DMEM and 100 mg/L low glucose (LG)-DMEM at 21% and 1% O2. The expression levels of LPAR1 and LPAR2 genes in cells cultured in MG-DMEM and LG-DMEM were significantly elevated, compared with HG-DMEM cells. The cell motility and survival rate to CDDP of cells cultured in MG-DMEM and LG-DMEM were significantly lower than those of cells cultured in HG-DMEM. The cell survival to CDDP was enhanced by LPA1 knockdown and suppressed by LPA2 knockdown. Under hypoxic conditions (1% O2), LPAR1, LPAR2 and LPAR3 expressions were markedly higher in cells cultured in MG-DMEM and LG-DMEM than in cells cultured in HG-DMEM. The cell survival rates to CDDP of cells cultured in MG-DMEM and LG-DMEM were elevated in comparison with HG-DMEM. The cell survival to CDDP was reduced by LPA3 knockdown. These results suggest that LPA receptor-mediated signaling is involved in the regulation of malignant properties of PANC-1 cells under glucose-deprived and hypoxic conditions.


Asunto(s)
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Receptores del Ácido Lisofosfatídico/metabolismo , Antineoplásicos/farmacología , Cisplatino/farmacología , Neoplasias Pancreáticas/patología , Movimiento Celular , Lisofosfolípidos/farmacología , Microambiente Tumoral , Neoplasias Pancreáticas
20.
FASEB J ; 37(3): e22787, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36723905

RESUMEN

Sphingosine kinase 1 (SK1) converts the pro-death lipid sphingosine to the pro-survival sphingosine-1-phosphate (S1P) and is upregulated in several cancers. DNA damaging agents, such as the chemotherapeutic doxorubicin (Dox), have been shown to degrade SK1 protein in cancer cells, a process dependent on wild-type p53. As mutations in p53 are very common across several types of cancer, we evaluated the effects of Dox on SK1 in p53 mutant cancer cells. In the p53 mutant breast cancer cell line MDA-MB-231, we show that Dox treatment significantly increases SK1 protein and S1P. Using MDA-MB-231 cells with CRISPR-mediated knockout of SK1 or the selective SK1 inhibitor PF-543, we implicated SK1 in both Dox-induced migration and in a newly uncovered proangiogenic program induced by Dox. Mechanistically, inhibition of SK1 suppressed the induction of the cytokine BMP4 and of the EMT transcription factor Snail in response to Dox. Interestingly, induction of BMP4 by SK1 increased Snail levels following Dox treatment by stabilizing Snail protein. Furthermore, we found that SK1 was required for Dox-induced p38 MAP kinase phosphorylation and that active p38 MAPK in turn upregulated BMP4 and Snail, positioning p38 downstream of SK1 and upstream of BMP4/Snail. Modulating production of S1P by inhibition of de novo sphingolipid synthesis or knockdown of the S1P-degrading enzyme S1P lyase identified S1P as the sphingolipid activator of p38 in this model. This work establishes a novel angiogenic pathway in response to a commonly utilized chemotherapeutic and highlights the potential of SK1 as a secondary drug target for patients with p53 mutant cancer.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Regulación hacia Arriba , Proteína p53 Supresora de Tumor/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingolípidos , Doxorrubicina/farmacología , Esfingosina/farmacología , Esfingosina/metabolismo , Lisofosfolípidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA