Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Comput Math Methods Med ; 2022: 5367753, 2022.
Article in English | MEDLINE | ID: mdl-36238480

ABSTRACT

Acute myocardial infarction (AMI) is the most severe form of coronary heart disease caused by ischemia and hypoxia. The study is aimed at investigating the role of neuropeptides and the mechanism of electroacupuncture (EA) in acute myocardial infarction (AMI) treatment. Compared with the normal population, a significant increase in substance P (SP) was observed in the serum of patients with AMI. PGI2 expression was increased in the SP-treated AMI mouse model, and TXA2 expression was decreased. And PI3K pathway-related genes, including Pik3ca, Akt, and Mtor, were upregulated in myocardial tissue of SP-treated AMI patients. Human cardiomyocyte cell lines (HCM) treated with SP increased mRNA and protein expression of PI3K pathway-related genes (Pik3ca, Pik3cb, Akt, and Mtor). Compared to MI control and EA-treated MI rat models, Myd88, MTOR, Akt1, Sp, and Irak1 were differentially expressed, consistent with in vivo and in vitro studies. EA treatment significantly enriched PI3K/AKT signaling pathway genes within MI-associated differentially expressed genes (DEGs) according to Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, it was confirmed by molecular docking analysis that PIK3CA, AKT1, and mTOR form stable dockings with neuropeptide SP. PI3K/AKT pathway activity may be affected directly or indirectly by EA via SP, which corrects the PGI2/TXA2 metabolic imbalance in AMI. MI treatment is now better understood as a result of this finding.


Subject(s)
Electroacupuncture , Myocardial Infarction , Animals , Class I Phosphatidylinositol 3-Kinases/metabolism , Computational Biology , Homeostasis , Humans , Mice , Molecular Docking Simulation , Myeloid Differentiation Factor 88/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger , Rats , Receptors, Epoprostenol/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Substance P/genetics , Substance P/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Phytomedicine ; 106: 154400, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36049428

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Paeoniflorin (PF) was found to exhibit renal protection from diabetic kidney disease (DKD) in previous trials, but its specific mechanism remains to be elucidated. AIM OF THE STUDY: This study furtherly explored the specific mechanism of PF in protect podocyte injury in DKD. MATERIALS AND METHODS: We observed the effects of PF on renal tissue and podocytes in DKD by constructing the vitro and vivo models after measuring the pharmacokinetic characteristics of PF. Target proteins of PF were found through target prediction, and verified by molecular docking, CESTA, and SPR, and then furtherly explored the downstream regulation mechanism related to podocyte autophagy and apoptosis by network prediction and co-immunoprecipitation. Finally, by using the target protein inhibitor in vivo and knocking down the target protein gene in vitro, it was verified that PF played a role in regulating autophagy and apoptosis through the target protein in diabetic nephropathy. RESULTS: This study found that in STZ-induced mice model, PF could improve the renal biochemical and pathological damage and podocyte injure (p < 0.05), upregulate autophagy activity (p < 0.05), but inhibit apoptosis (p < 0.01). Vascular endothelial growth factor receptor 2 (VEGFR2), predicted as the target of PF, directly bind with PF reflected by molecular docking and surface plasmon resonance detection. Animal studies demonstrated that VEGFR2 inhibitors have a protective effect similar to that of PF on DKD. Network prediction and co-immunoprecipitation further confirmed that VEGFR2 was able to bind PIK3CA to regulate PI3K-AKT signaling pathway. Furthermore, PF downregulated the phosphorylation of PI3K and AKT (p < 0.05). In vitro, similarly to autophagy inhibitors, PF was also found to improve podocyte markers (p < 0.05) and autophagy activity (p < 0.05), decrease caspase 3 protein (p < 0.05) and further inhibited VEGFR2-PI3K-AKT activity (p < 0.05). Finally, the results of VEGFR2 knockdown were similar to the effect of PF in HG-stimulated podocytes. CONCLUSION: In conclusion, PF restores autophagy and inhibits apoptosis by targeting the VEGFR2-mediated PI3K-AKT pathway to improve renal injury in DKD, that provided a theoretical basis for PF treatment in DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Animals , Apoptosis , Autophagy , Caspase 3/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/therapeutic use , Diabetic Nephropathies/metabolism , Glucosides , Mice , Molecular Docking Simulation , Monoterpenes , Phosphatidylinositol 3-Kinases/metabolism , Podocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Elife ; 112022 07 05.
Article in English | MEDLINE | ID: mdl-35787784

ABSTRACT

Background: Lymphatic malformations (LMs) often pose treatment challenges due to a large size or a critical location that could lead to disfigurement, and there are no standardized treatment approaches for either refractory or unresectable cases. Methods: We examined the genomic landscape of a patient cohort of LMs (n = 30 cases) that underwent comprehensive genomic profiling using a large-panel next-generation sequencing assay. Immunohistochemical analyses were completed in parallel. Results: These LMs had low mutational burden with hotspot PIK3CA mutations (n = 20) and NRAS (n = 5) mutations being most frequent, and mutually exclusive. All LM cases with Kaposi sarcoma-like (kaposiform) histology had NRAS mutations. One index patient presented with subacute abdominal pain and was diagnosed with a large retroperitoneal LM harboring a somatic PIK3CA gain-of-function mutation (H1047R). The patient achieved a rapid and durable radiologic complete response, as defined in RECIST1.1, to the PI3Kα inhibitor alpelisib within the context of a personalized N-of-1 clinical trial (NCT03941782). In translational correlative studies, canonical PI3Kα pathway activation was confirmed by immunohistochemistry and human LM-derived lymphatic endothelial cells carrying an allele with an activating mutation at the same locus were sensitive to alpelisib treatment in vitro, which was demonstrated by a concentration-dependent drop in measurable impedance, an assessment of cell status. Conclusions: Our findings establish that LM patients with conventional or kaposiform histology have distinct, yet targetable, driver mutations. Funding: R.P. and W.A. are supported by awards from the Levy-Longenbaugh Fund. S.G. is supported by awards from the Hugs for Brady Foundation. This work has been funded in part by the NCI Cancer Center Support Grants (CCSG; P30) to the University of Arizona Cancer Center (CA023074), the University of New Mexico Comprehensive Cancer Center (CA118100), and the Rutgers Cancer Institute of New Jersey (CA072720). B.K.M. was supported by National Science Foundation via Graduate Research Fellowship DGE-1143953. Clinical trial number: NCT03941782.


Subject(s)
Antineoplastic Agents , Class I Phosphatidylinositol 3-Kinases , GTP Phosphohydrolases , Lymphangioma , Lymphatic Abnormalities , Membrane Proteins , Thiazoles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , GTP Phosphohydrolases/genetics , Genomics , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Lymphangioma/drug therapy , Lymphangioma/genetics , Lymphatic Abnormalities/drug therapy , Lymphatic Abnormalities/genetics , Membrane Proteins/genetics , Mutation , Sequence Analysis, DNA , Thiazoles/pharmacology , Thiazoles/therapeutic use
4.
J Clin Lab Anal ; 36(6): e24444, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35435290

ABSTRACT

BACKGROUND AND OBJECTIVE: Aberrant gene expression and abnormal signaling pathways often occur in patients with colorectal cancer, in which mutations in B-Raf Proto-Oncogene (BRAF), KRAS Proto-Oncogene (KRAS), and Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha (PIK3CA) are quite common. In this study, the relationship between BRAF, KRAS, and PIK3CA mutations and clinicopathologic features and prognosis of colorectal cancer patients was investigated. METHODS: One hundred and fifty patients with colorectal cancer admitted to Affiliated people's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine were collected and grouped according to the mutation patterns of BRAF, KRAS, and PIK3CA. The association between BRAF, KRAS, and PIK3CA mutations and pathological factors (age, sex, etc.) was analyzed using the Chi-square test. Subsequently, survival analysis was performed to screen the impact factors of overall survival time by Kaplan-Meier (K-M) curve, and Cox regression model was established for the selected factors. RESULTS: BRAF, KRAS, and PIK3CA mutations were not associated with age, sex, and alcoholism. K-M curve and log-rank test results demonstrated that among the factors included in this study, overall survival rate of colorectal cancer patients was only associated with mutation factors. The prognosis of KRAS+/PIK3CA-/BRAF-mutant and KRAS-/PIK3CA-/BRAF+mutant patients was better than that of KRAS+/PIK3CA+/BRAF-mutant patients. CONCLUSION: The mutant patterns of BRAF, KRAS, and PIK3CA were not related to the general and clinicopathological features of patients. The mutant pattern could be used as an independent prognostic factor for colorectal cancer.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins p21(ras) , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
5.
Bioengineered ; 13(1): 1575-1589, 2022 01.
Article in English | MEDLINE | ID: mdl-35012428

ABSTRACT

The present study attempts to explore the effective components, action targets, and potential mechanism of nightshade for colon cancer treatment. The relationship network diagram of 'traditional Chinese medicine - component - target - disease' was firstly constructed by employing network pharmacology. Experiments were conducted in vivo and in vitro to verify the influence of quercetin, the core effective component of nightshade, on colon cancer. Meanwhile, the regulatory effects of quercetin on core targets and main signaling pathways were determined. Based on the network diagram of 'traditional Chinese medicine - component - target - disease' and KEGG analysis, quercetin might exhibit certain effects on colon cancer treatment by regulating the biological behavior of core targets related to cell apoptosis in tumors including PIK3R1, PIK3CA, Akt1, and Akt2. Furthermore, quercetin has been demonstrated in vitro experiments to suppress the proliferation and migration of colon cancer cells whereas promote their apoptosis in a dose-dependent fashion. In vivo experiments indicate that quercetin had an antitumor effect on human colon cancer SW480 cells in nude mice bearing tumors. Furthermore, PIK3CA could bind to quercetin directly, which is validated by immunocoprecipitation. Therefore, the activation of PI3K/AKT phosphorylation was inhibited by quercetin and moreover the expressions of apoptotic proteins caspase-3 and Bcl2-Associated X protein (BAX) were up-regulated. In conclusion, the potential mechanism of nightshade lies in the activation of the PI3K/AKT signaling pathway inhibited by quercetin, thus promoting apoptosis of colon cancer cells for colon cancer treatment.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Colonic Neoplasms/drug therapy , Quercetin/administration & dosage , Solanum/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Network Pharmacology , Phosphorylation/drug effects , Plant Extracts/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 42: 128046, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33865969

ABSTRACT

PI3K-δ mediates key immune cell signaling pathways and is a target of interest for treatment of oncological and immunological disorders. Here we describe the discovery and optimization of a novel series of PI3K-δ selective inhibitors. We first identified hits containing an isoindolinone scaffold using a combined ligand- and receptor-based virtual screening workflow, and then improved potency and selectivity guided by structural data and modeling. Careful optimization of molecular properties led to compounds with improved permeability and pharmacokinetic profile, and high potency in a whole blood assay.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Discovery , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phthalimides/pharmacology , Class I Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phthalimides/chemical synthesis , Phthalimides/chemistry , Structure-Activity Relationship
7.
Bioorg Med Chem ; 29: 115863, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33199203

ABSTRACT

PI3Kα is an attractive target for PIK3CA mutated malignant tumor and searching for lead compounds with novel scaffold is important for the development of PI3Kα inhibitors. Therefore, the strategy of docking-based virtual screening was performed to discovery potent inhibitors. The 4L2Y_A PI3Kα crystal structure was used as the model protein receptor due to its high docking reliability. After the multistep virtual screening protocol and biological evaluation, three hits were picked up and further similarity searching led to more potent 2-(5-(quinolin-6-yl)-1,3,4-oxadiazol-2-yl)acetamide derivatives ES-25 and ES-27. In addition, the primary SAR of these novel derivatives was discussed, which provide a basis for the further structural modification.


Subject(s)
Acetamides/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Discovery , Molecular Docking Simulation , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Acetamides/chemical synthesis , Acetamides/chemistry , Class I Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/chemistry , Structure-Activity Relationship
8.
J Anat ; 238(3): 743-750, 2021 03.
Article in English | MEDLINE | ID: mdl-33094520

ABSTRACT

The anatomy of the hypothalamus includes many nuclei and a complex network of neurocircuits. In this context, some hypothalamic nuclei reside closer to the blood-brain barrier, allowing communication with the peripheral organs through some molecules, such as leptin. Leptin is considered the main adipokine for energy homeostasis control. Furthermore, leptin signalling in the hypothalamus can communicate with insulin signalling through the activation of phosphoinositide 3-kinase (PI3k). Previous data suggest that isoforms of PI3k are necessary to mediate insulin action in the hypothalamus. However, obese animals show impairment in the central signalling of these hormones. Thus, in the current study, we evaluated the role of acute exercise in the leptin and insulin pathways in the hypothalamus, as well as in food intake control in obese mice. Although acute physical exercise was not able to modulate leptin signalling, this protocol suppressed the increase in the suppressor of cytokine signalling 3 (SOCS3) protein levels. In addition, acute exercise increased the content of PI3k-p110α protein in the hypothalamus. The exercised animals showed a strong tendency to reduction in cumulative food intake. For the first time, our results indicate physical exercise can increase PI3k-p110α protein content in the hypothalamus of obese mice and regulate food intake.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Obesity/therapy , Physical Conditioning, Animal/physiology , Animals , Male , Mice , Obesity/metabolism
9.
Nutrients ; 12(6)2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32575897

ABSTRACT

Present study was conducted to investigate ameliorating effects of Mori Cortex radicis on cognitive impair and neuronal defects in HFD-induced (High Fat Diet-Induced) obese mice. To induce obesity, C57BL/6 mice were fed an HFD for 8 weeks, and then mice were fed the HFD plus Mori Cortex radicis extract (MCR) (100 or 200 mg/kg/day) for 6 weeks. Prior to sacrifice, body weights were measured, and Y-maze test and oral glucose tolerance test were performed. Serum lipid metabolic biomarkers (TG, LDL, and HDL/total cholesterol ratio) and antioxidant enzymes (glutathione, superoxide dismutase, and catalase), malondialdehyde (MDA), and acetylcholinesterase (AChE) levels were measured in brain tissues. The expressions of proteins related to insulin signaling (p-IRS, PI3K, p-Akt, and GLUT4) and neuronal protection (p-Tau, Bcl-2, and Bax) were examined. MCR suppressed weight gain, improved serum lipid metabolic biomarker and glucose tolerance, inhibited AChE levels and MDA production, and restored antioxidant enzyme levels in brain tissue. In addition, MCR induced neuronal protective effects by inhibiting p-Tau expression and increasing Bcl-2/Bax ratio, which was attributed to insulin-induced increases in the expressions p-IRS, PI3K, p-Akt, and GLUT4. These indicate MCR may reduce HFD-induced insulin dysfunction and neuronal damage and suggest MCR be considered a functional material for the prevention of T2DM-associated neuronal disease.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , Cognition Disorders/metabolism , Cognition/drug effects , Insulin/metabolism , Morus , Obesity/metabolism , Acetylcholinesterase/metabolism , Animals , Antioxidants/metabolism , Antioxidants/therapeutic use , Blood Glucose/metabolism , Brain/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Diabetes Complications/metabolism , Diabetes Complications/prevention & control , Diet, High-Fat , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Phytotherapy , Plant Bark , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Roots , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
10.
Med Oncol ; 37(2): 12, 2019 Dec 21.
Article in English | MEDLINE | ID: mdl-31865465

ABSTRACT

We performed a prospective trial to assess the clinical benefit of a tailored gene set built on a next-generation sequencing (NGS) platform in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). Archived tumor tissue obtained from patients with recurrent or metastatic HNSCC was analyzed for variants by a tailored Comprehensive Cancer Gene set of 40 genes (CCG-40) performed on a NGS platform. These data were provided to clinicians to inform treatment decisions. The primary endpoint was clinical benefit (disease control) that resulted from selection and administration of a targeted therapy based on results of the CCG-40. Barriers to performance and implementation of the assay were recorded. Forty patients enrolled. Primary tumor sites included oropharynx (14), larynx/hypopharynx (14), oral cavity (9), and nasopharynx (3). The CCG-40 assay was performed in 23 patients (57.5%), but not in 17 patients due inadequate financial coverage (12) or insufficient tumor tissue (5). Potentially actionable tumor variants were identified in 3 patients (7.5%); all were PIK3CA variants. Due to inability to obtain access to candidate drugs (2) or rapid decline in performance status (1), none of these patients received targeted therapy informed by the CCG-40 results. The CCG-40 assay did not provide clinical benefit to the patients on this trial. Identification of limitations of the assay and barriers to the test's performance and application may be used to optimize this strategy in future trials.


Subject(s)
Biomarkers, Tumor/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Head and Neck Neoplasms/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Neoplasm Recurrence, Local/genetics , Adult , Aged , Biomarkers, Tumor/analysis , Class I Phosphatidylinositol 3-Kinases/metabolism , Databases, Genetic , Disease-Free Survival , Female , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Patient Selection , Prospective Studies
11.
Sci Rep ; 9(1): 16647, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719636

ABSTRACT

The present state of cancer chemotherapy is unsatisfactory. New anticancer drugs that marginally improve the survival of patients continue to be developed at an unsustainably high cost. The study aimed to elucidate the effects of insulin (INS), an inexpensive drug with a convincing safety profile, on the susceptibility of colon cancer to chemotherapeutic agents: 5-fluorouracil (FU), oxaliplatin (OXA), irinotecan (IRI), cyclophosphamide (CPA) and docetaxel (DOC). To examine the effects of insulin on cell viability and apoptosis, we performed an in vitro analysis on colon cancer cell lines Caco-2 and SW480. To verify the results, we performed in vivo analysis on mice bearing MC38 colon tumors. To assess the underlying mechanism of the therapy, we examined the mRNA expression of pathways related to the signaling downstream of insulin receptors (INSR). Moreover, we performed Western blotting to confirm expression patterns derived from the genetic analysis. For the quantification of circulating tumor cells in the peripheral blood, we used the maintrac method. The results of our study show that insulin-pretreated colon cancer cells are significantly more susceptible to commonly used chemotherapeutics. The apoptosis ratio was also enhanced when INS was administered complementary to the examined drugs. The in vivo study showed that the combination of INS and FU resulted in significant inhibition of tumor growth and reduction of the number of circulating tumor cells. This combination caused a significant downregulation of the key signaling substrates downstream of INSR. The results indicate that the downregulation of PIK3CA (phosphatidylinositol 3-kinase catalytic subunit alpha), which plays a critical role in cell signaling and GRB2 (growth factor receptor-bound protein 2), a regulator of cell proliferation and differentiation may be responsible for the sensitizing effect of INS. These findings were confirmed at protein levels by Western blotting. In conclusion, these results suggest that INS might be potentially applied to clinical use to enhance the therapeutic effectiveness of chemotherapeutic drugs. The findings may become a platform for the future development of new and inexpensive strategies for the clinical chemotherapy of tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , GRB2 Adaptor Protein/antagonists & inhibitors , Insulin/pharmacology , Animals , Blotting, Western , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Colorectal Neoplasms/metabolism , Cyclophosphamide/therapeutic use , Docetaxel/therapeutic use , Down-Regulation/drug effects , Drug Synergism , Female , Fluorouracil/therapeutic use , GRB2 Adaptor Protein/metabolism , Humans , Irinotecan/therapeutic use , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Oxaliplatin/therapeutic use
12.
Sci Rep ; 9(1): 7904, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31133639

ABSTRACT

Pyrrolone-fused benzosuberene (PBS) compounds were semi-synthesized from α,ß,γ-Himachalenes extracted from the essential oil of Cedrus deodara following amino-vinyl-bromide substituted benzosuberenes as intermediates. These PBSs compounds classified as an attractive source of therapeutics. The α-isoform of PI3K which is a pivotal modulator of PI3K/AKT/mTOR signaling pathway, responsible for neurological disorders like epilepsy, found as a potential target molecule against these 17 semi-synthesized PBS compounds using in silico ligand-based pharmacophore mapping and target screening. The compounds screened using binding affinities, ADMET properties, and toxicity that were accessed by in silico docking simulations and pharmacokinetics profiling. Ultimately two compounds viz., PBS-8 and PBS-9 were selected for further in vivo evaluation using a zebrafish (Danio rerio) model of pentylenetetrazol (PTZ)-induced clonic convulsions. Additionally, gene expression studies performed for the genes of the PI3K/AKT/mTOR pathway which further validated our results. In conclusion, these findings suggested that PBS-8 is a promising candidate that could bedeveloped as a potential antiepileptic.


Subject(s)
Anticonvulsants/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Coumarins/pharmacology , Epilepsy/drug therapy , Pyrroles/pharmacology , Animals , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/metabolism , Coumarins/chemistry , Coumarins/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Epilepsy/chemically induced , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Molecular Docking Simulation , Pentylenetetrazole/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Pyrroles/chemistry , Pyrroles/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Zebrafish , Zebrafish Proteins/metabolism
13.
Bone ; 121: 181-190, 2019 04.
Article in English | MEDLINE | ID: mdl-30682567

ABSTRACT

INTRODUCTION: The purpose of this study was to examine growth-promoting effects of myo-inositol nutritional supplementation on the mandible in experimental animals. METHODS: Mice were fed on diets that contained various concentration of myo-inositol for 3 to 12 weeks. The length of the mandible, maxilla, and femur were measured on µCT images. The mandible and tibia were examined histologically and immunohistochemically. The effects of myo-inositol on cell proliferation and chondrocytic differentiation were examined using ATDC5 cells. RESULTS: Myo-inositol supplementation had no effects on body weight, length, and maxilla and femur lengths. However, the length of mandible and the thickness of the mandibular condylar cartilage (MCC) were increased by myo-inositol supplement. Microarray analysis revealed that Pik3cd was highly expressed in MCC as compared to that in the cartilage of the tibial growth plate, which was confirmed by real-time RT-PCR and immunohistochemistry. ATDC5 cells also highly expressed Pik3CD. Myoinositol induced increases in cell proliferation and chondrocytic differentiation in ATDC5 cells. The addition of a PIK3CD inhibitor blocked the induction of cell proliferation by myo-inositol in ATDC5 cells. CONCLUSIONS: Nutritional supplementation with myo-inositol in growing mice augmented mandibular endochondral growth without any systemic effects. The specific promotion of mandibular growth by myoinositol is primarily dependent on the specific intensive expression of PIK3CD in the MCC.


Subject(s)
Inositol/pharmacology , Mandibular Condyle/drug effects , Mandibular Condyle/growth & development , Animals , Cartilage/drug effects , Cartilage/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Male , Mice
14.
J Inorg Biochem ; 193: 60-69, 2019 04.
Article in English | MEDLINE | ID: mdl-30684759

ABSTRACT

Our previous study revealed that selenium (Se) deficiency can cause myocardial injury through triggering autophagy. MicroRNAs (miRNAs) play crucial roles in autophagic cell death. However, the relationship between miRNAs and myocardial autophagy injury caused by Se deficiency remains unclear. We selected differential microRNA-215-5p (miR-215-5p) in Se-deficient myocardial tissue using high-throughput miRNA-sequencing. To further explore the role of miR-215-5p in myocardial injury, overexpression/knockdown of miR-215-5p in primary cardiomyocyte model was established by miRNAs interference technology. In this study, we report that miR-215-5p can promote myocardial autophagy by directly binding to the 3'untranslated region (3'UTR) of phosphatidylinositol-4, 5-bisphosphate 3-kinase (PI3K). Its target gene PI3K was confirmed by dual luciferase reporter assay, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot in cardiomyocytes. Our results showed that overexpression of miR-215-5p could trigger myocardial autophagy through PI3K-threonine-protein kinase (AKT)-target of rapamycin (TOR) pathway. Further studies revealed that autophagic cell death was dependent on the activation of extracellular signal-regulated kinase1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), p38 kinase (p38) and generation of reactive oxygen species (ROS) in overexpression of miR-215-5p in cardiomyocytes. On the contrary, miR-215-5p inhibitor can enhance cell survival capacity against autophagy by inhibiting ROS-mitogen-activated protein kinase (MAPK) pathways and activating the PI3K/AKT/TOR pathway in cardiomyocytes. Together, our findings support that miR-215-5p may modulate cell survival programs by regulating autophagy, and miR-215-5p acts as an autophagic regulator in the regulatory feedback loop that regulates cardiomyocyte survival by modulating the PI3K/AKT/TOR pathway and ROS-dependent MAPK pathways.


Subject(s)
Autophagy/physiology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , MAP Kinase Signaling System/physiology , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Animals , Cell Survival/physiology , Chickens , Class I Phosphatidylinositol 3-Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/cytology , Reactive Oxygen Species/metabolism , Selenium/deficiency
15.
Comput Biol Chem ; 78: 190-204, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30557817

ABSTRACT

BACKGROUND: Phosphoinositide-3-kinase Delta (PI3Kδ) plays a key role in B-cell signal transduction and inhibition of PI3Kδ is confirmed to have clinical benefit in certain types of activation of B-cell malignancies. Virtual screening techniques have been used to discover new molecules for developing novel PI3Kδ inhibitors with little side effects. METHOD: Computer aided drug design method were used to rapidly screen optimal PI3Kδ inhibitors from the Asinex database. Virtual screening based molecular docking was performed to find novel and potential lead compound targeting PI3Kδ, at first. Subsequently, drug likeness studies were carried out on the retrieved hits to evaluate and analyze their drug like properties such as absorption, distribution, metabolism, excretion, and toxicity (ADMET) for toxicity prediction. Three least toxic compounds were selected for the molecular dynamics (MD) simulations for 30 ns in order to validate its stability inside the active site of PI3Kδ receptor. RESULTS: Based on the present in silico analysis, two molecules have been identified which occupied the same binding pocket confirming the selection of active site. ASN 16296138 (Glide score: -12.175 kcal/mol, cdocker binding energy: -42.975 kcal/mol and ΔGbind value: -90.457 kcal/mol) and BAS 00227397 (Glide score: -10.988 kcal/mol, cdocker binding energy: -39.3376 kcal/mol and ΔGbind value: -81.953 kcal/mol) showed docking affinities comparatively much stronger than those of already reported known inhibitors against PI3Kδ. These two ligand's behaviors also showed consistency during the simulation of protein-ligand complexes for 30000 ps respectively, which is indicative of its stability in the receptor pocket. CONCLUSION: Compound ASN 16296138 and BAS 00227397 are potential candidates for experimental validation of biological activity against PI3Kδ in future drug discovery studies. This study smoothes the path for the development of novel leads with improved binding properties, high drug likeness, and low toxicity to humans for the treatment of cancer.


Subject(s)
Computer-Aided Design , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemistry , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Discovery , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Structure , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Skin/drug effects , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Thermodynamics
16.
Methods Mol Biol ; 1762: 31-50, 2018.
Article in English | MEDLINE | ID: mdl-29594766

ABSTRACT

Drug discovery has evolved significantly over the past two decades. Progress in key areas such as molecular and structural biology has contributed to the elucidation of the three-dimensional structure and function of a wide range of biological molecules of therapeutic interest. In this context, the integration of experimental techniques, such as X-ray crystallography, and computational methods, such as molecular docking, has promoted the emergence of several areas in drug discovery, such as structure-based drug design (SBDD). SBDD strategies have been broadly used to identify, predict and optimize the activity of small molecules toward a molecular target and have contributed to major scientific breakthroughs in pharmaceutical R&D. This chapter outlines molecular docking and structure-based virtual screening (SBVS) protocols used to predict the interaction of small molecules with the phosphatidylinositol-bisphosphate-kinase PI3Kδ, which is a molecular target for hematological diseases. A detailed description of the molecular docking and SBVS procedures and an evaluation of the results are provided.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Evaluation, Preclinical/methods , Small Molecule Libraries/chemistry , Crystallography, X-Ray , Drug Design , Drug Discovery , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Protein Conformation , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
17.
Am J Chin Med ; 43(1): 121-36, 2015.
Article in English | MEDLINE | ID: mdl-25649747

ABSTRACT

α-Phellandrene (α-PA) is a cyclic monoterpene. To investigate the induction of autophagy by α-PA and its mechanism, human liver tumor cells (J5) were incubated with α-PA and analyzed for cell viability and the molecular regulation of pre-autophagosome origination and autophagosome formation. According to the results, PI3K-I, mTOR, and Akt protein levels were decreased after α-PA treatment compared to those of the control group (p < 0.05). The phosphorylation of Bcl-2, and PI3K-III, LC3-II and Beclin-1 protein levels in J5 cells were increased after α-PA treatment (p < 0.05). In addition, α-PA up-regulated nuclear p53 and down-regulated cytoplasmic p53 expression in J5 cells. The NF-κB pathway was activated, as indicated by increase in cytosolic phosphorylated IκB, nuclear NF-κB levels, and the DNA-binding activity of NF-κB after α-PA treatment in J5 cells (p < 0.05). These results suggest that α-PA can induce J5 cell autophagy by regulating mTOR and LC-3II expression, p53 signaling, and NF-κB activation in J5 cells.


Subject(s)
Autophagy/drug effects , Autophagy/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Monoterpenes/pharmacology , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Cyclohexane Monoterpenes , Cytosol/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Phagosomes/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
18.
Adv Biol Regul ; 58: 1-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25512233

ABSTRACT

With the steady rise in the incidence of obesity and its associated comorbidities, in the last decades research aimed at understanding molecular mechanisms that control body weight has gained new interest. Fat gain is frequently associated with chronic adipose tissue inflammation and with peripheral as well as central metabolic derangements, resulting in an impaired hypothalamic regulation of energy homeostasis. Recent attention has focused on the role of phosphatidylinositol 3-kinase (PI3K) in both immune and metabolic response pathways, being involved in the pathophysiology of obesity and its associated metabolic diseases. In this review, we focus on distinct PI3K isoforms, especially class I PI3Ks, mediating inflammatory cells recruitment to the enlarged fat as well as intracellular responses to key hormonal regulators of fat storage, both in adipocytes and in the central nervous system. This integrated view of PI3K functions may ultimately help to develop new therapeutic interventions for the treatment of obesity.


Subject(s)
Adipose Tissue/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Hypothalamus/metabolism , Obesity/metabolism , Signal Transduction/immunology , Adipocytes/immunology , Adipocytes/pathology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/immunology , Energy Metabolism/genetics , Energy Metabolism/immunology , Gene Expression Regulation , Homeostasis , Humans , Hypothalamus/immunology , Hypothalamus/pathology , Immunity, Innate , Inflammation , Insulin Resistance , Leptin/genetics , Leptin/immunology , Obesity/genetics , Obesity/immunology , Obesity/pathology , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/immunology
19.
Cell Metab ; 17(4): 599-606, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23541372

ABSTRACT

The fundamental importance of the hypothalamus in the regulation of autonomic and cardiovascular functions is well established. However, the molecular processes involved are not well understood. Here, we show that the mammalian (or mechanistic) target of rapamycin (mTOR) signaling in the hypothalamus is tied to the activity of the sympathetic nervous system and to cardiovascular function. Modulation of mTOR complex 1 (mTORC1) signaling caused dramatic changes in sympathetic traffic, blood flow, and arterial pressure. Our data also demonstrate the importance of hypothalamic mTORC1 signaling in transducing the sympathetic and cardiovascular actions of leptin. Moreover, we show that the PI3K pathway links the leptin receptor to mTORC1 signaling and that changes in its activity impact sympathetic traffic and arterial pressure. These findings establish mTORC1 activity in the hypothalamus as a key determinant of sympathetic and cardiovascular regulation and suggest that dysregulated hypothalamic mTORC1 activity may influence the development of cardiovascular diseases.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Proteins/metabolism , Animals , Arterial Pressure/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Leucine/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Leptin/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL