Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
FASEB J ; 35(11): e21985, 2021 11.
Article in English | MEDLINE | ID: mdl-34674317

ABSTRACT

Inflammation is broadly recognized as an important factor in the pathogenesis of acute kidney injury (AKI), but pharmacological approaches to alleviate inflammation in AKI have not been proved successful in clinical trials. Macrophage infiltration into renal tissue promotes inflammatory responses that contribute to the pathogenesis of AKI. Suppression of renal tissue inflammatory responses is postulated to improve renal injury of patients and animals. Rhodomeroterpene (RMT) is a novel meroterpenoid isolated from the Rhododendron genus that was shown to exert anti-inflammatory action in vivo or in vitro in this study. We investigated the treatment effects of RMT on LPS-induced sepsis and two different AKI models. The results showed that pretreatment with RMT (30 mg kg-1  d-1 , ip, for 3 days) significantly inhibited acute inflammatory responses in LPS-induced septic mice. In both renal ischemia-reperfusion injury (I/R) and sepsis-induced AKI models, RMT (30 mg kg-1  d-1 , ip, for 3 days) ameliorated renal function and injury and alleviated inflammation by reducing the infiltration of immune cells, including macrophages and neutrophils. Furthermore, our study demonstrated that RMT inhibits inflammatory responses in macrophages. The anti-inflammatory effects of RMT may be due to the inactivation of the IKK/NF-κB and PI3K/PDK1/Akt inflammatory signaling pathways in macrophages. Collectively, our findings indicate that RMT ameliorates renal injury and alleviates the renal inflammatory state in different AKI models, suggesting that RMT may be a potential agent for the treatment of AKI.


Subject(s)
Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Macrophages/drug effects , Rhododendron/chemistry , Terpenes/pharmacology , Animals , Bone Marrow Cells , HEK293 Cells , Humans , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
2.
Cell Mol Biol Lett ; 27(1): 28, 2022 Mar 19.
Article in English | MEDLINE | ID: mdl-35305553

ABSTRACT

BACKGROUND: Parecoxib plays an important role in inhibition of human cancer. However, the effect of parecoxib on esophageal squamous cell carcinoma (ESCC) is still not well known. The purpose of this study was to investigate the effect of parecoxib on ESCC and its underlying mechanism. METHODS: RNA-sequence analysis was performed to identify functional alterations and mechanisms. Cell cycle, proliferation, invasion, and migration were assessed using flow cytometry, CCK-8 assay, colony formation, transwell, and wound healing assays. Extracellular matrix (ECM) degradation was detected by substrate gel zymography and 3D cell culture assay. Western blotting was used to detect parecoxib-dependent mechanisms involving cell cycle, proliferation, invasion, and migration. Tumor formation in vivo was detected by mouse assay. RESULTS: Functional experiments indicated that parecoxib induced ESCC cell cycle arrest in G2 phase, and inhibited cell proliferation, invasion, and migration in vitro. Western blotting revealed that parecoxib downregulated the phosphorylation levels of AKT and PDK1, as well as the expression of the mutant p53, cyclin B1, and CDK1, while upregulating p21waf1. Parecoxib inhibited matrix metalloproteinase-2 (MMP2) secretion and invadopodia formation, which were related to ECM degradation. Furthermore, we found that parecoxib suppressed ESCC growth in heterotopic tumor models. CONCLUSION: Parecoxib inhibits ESCC progression, including cell cycle, proliferation, invasion, and migration, via the PDK1-AKT signaling pathway.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Animals , Cell Line, Tumor , Cell Movement , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/metabolism , Isoxazoles , Matrix Metalloproteinase 2 , Mice , Proto-Oncogene Proteins c-akt/metabolism
3.
Cell Mol Life Sci ; 77(8): 1623-1643, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31378829

ABSTRACT

The pathophysiology of type 2 diabetes mellitus (T2D) is characterized by reduced or absent insulin receptor (INSR) responsiveness to its ligand, elevated hepatic glucose output and impaired glucose uptake in peripheral tissues, particularly skeletal muscle. Treatments to reduce hyperglycemia and reestablish normal insulin signaling are much sought after. Any agent which could be orally administered to restore INSR function, in an insulin-independent manner, would have major implications for the management of this global disease. We have discovered a non-peptidyl small molecule, adenosine, 5'-Se-methyl-5'-seleno-, 2',3'-diacetate [referred to as non-peptidyl compound #43 (NPC43)], which restores INSR signaling in the complete absence of insulin. Initial screening of numerous compounds in human HepG2 liver cells revealed that NPC43 significantly inhibited glucose production. The compound was potently anti-hyperglycemic and anti-hyperinsulinemic in vivo, in insulin-resistant T2D Leprdb/db mice, following either acute or chronic treatment by oral gavage and intraperitoneal injection, respectively. The compound acted at the level of INSR and activated it in both liver and skeletal muscle of Leprdb/db mice. In cell culture, the compound activated INSR in both liver and skeletal muscle cells; furthermore, it cooperated with insulin to depress glucose-6-phosphatase catalytic subunit (G6pc) expression and stimulate glucose uptake, respectively. Our results indicated that the compound directly interacted with INSRα, triggering appropriate phosphorylation and activation of the receptor and its downstream targets. Unlike insulin, NPC43 did not activate insulin-like growth factor 1 receptor in either liver or skeletal muscle. We believe this compound represents a potential oral and/or injectable insulin replacement therapy for diabetes and diseases associated with insulin resistance.


Subject(s)
Adenosine/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Receptor, Insulin/metabolism , Adenosine/analogs & derivatives , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hep G2 Cells , Humans , Hyperglycemia/complications , Hyperglycemia/metabolism , Hypoglycemic Agents/chemistry , Insulin/metabolism , Insulin Resistance , Male , Methylation , Mice , Mice, Inbred C57BL , Organoselenium Compounds/chemistry , Organoselenium Compounds/therapeutic use
4.
J Cell Physiol ; 234(11): 20728-20741, 2019 11.
Article in English | MEDLINE | ID: mdl-31004362

ABSTRACT

Non-small-cell lung cancer (NSCLC) is an aggressive subtype of pulmonary carcinomas with high mortality. However, chemotherapy drug resistance and high recurrence rates hinder the curative effect of platinum-based first-line chemotherapy, which makes it urgent to develop new antitumor drugs for NSCLC. 9za, a new candidate drug synthesized by our research group, has been verified with potent antilung cancer activity in preliminary experiments. However, the underlying molecular mechanism of 9za remains largely vague. This work revealed that 9za could play important cytotoxic and proapoptotic roles in NSCLC cells. Moreover, 9za could induce autophagy and promote autophagy flux. Interestingly, the cytotoxic and proapoptotic roles were significantly dependent on 9za-induced cytoprotective autophagy. That is, the coadministration of 9za with an autophagy inhibitor such as chloroquine or 3-methyladenine exhibited increased cytotoxic and proapoptotic effects compared with 9za treatment alone. In addition, 9za exposure suppressed the phosphorylation of phosphoinositide-dependent protein kinase 1 (PDK1), protein kinase B (Akt), mammalian targets of rapamycin (mTOR), p70 S6 kinase, and 4E binding protein 1 by a dose-dependent way, manifesting that the Akt/mTOR axis was implicated in 9za-induced autophagy. In addition, the overexpression of PDK1 resulted in increased phosphorylation of PDK1 and Akt and blocking of 9za-mediated autophagy. These data showed that the PDK1/Akt/mTOR pathway was involved in 9za-induced autophagy. Hence, this work provides a theoretical basis for exploiting 9za as a new antilung cancer candidate drug and hints that the combination of 9za with an autophagy inhibitor is a feasible alternative approach for the therapy of NSCLC.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Indoles/pharmacology , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Aniline Compounds/chemistry , Aniline Compounds/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Benzodioxoles/chemistry , Benzodioxoles/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/chemistry , Indoles/therapeutic use , Molecular Structure , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics
5.
Bioorg Med Chem ; 27(6): 944-954, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30777660

ABSTRACT

The dysfunction and mutual compensatory activation of RAF-MEK-ERK and PI3K-PDK1-AKT pathways have been demonstrated as the hallmarks in several primary and recurrent cancers. The strategy of concurrent blocking of these two pathways shows clinical merits on effective cancer therapy, such as combinatory treatments and dual-pathway inhibitors. Herein, we report a novel prototype of dual-pathway inhibitors by means of merging the core structural scaffolds of a MEK1 inhibitor and a PDK1 inhibitor. A library of 43 compounds that categorized into three series (Series I-III) was synthesized and tested for antitumor activity in lung cancer cells. The results from structure-activity relationship (SAR) analysis showed the following order of antitumor activity that 3-hydroxy-5-(phenylamino) indolone (Series III) > 3-alkenyl-5-(phenylamino) indolone (Series I) > 3-alkyl-5-(phenylamino) indolone (Series II). A lead compound 9za in Series III showed most potent antitumor activity with IC50 value of 1.8 ±â€¯0.8 µM in A549 cells. Moreover, antitumor mechanism study demonstrated that 9za exerted significant apoptotic effect, and cellular signal pathway analysis revealed the potent blockage of phosphorylation levels of ERK and AKT in RAF-MEK-ERK and PI3K-PDK1-AKT pathways, respectively. The results reported here provide robust experimental basis for the discovery and optimization of dual pathway agents for anti-lung cancer therapy.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , A549 Cells , Amination , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzodioxoles/chemical synthesis , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Cell Line, Tumor , Drug Design , Humans , Indoles/chemical synthesis , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , raf Kinases/metabolism
6.
Bioorg Chem ; 87: 136-141, 2019 06.
Article in English | MEDLINE | ID: mdl-30884307

ABSTRACT

Chlorovaltrates U-W (1-3), three previously undescribed iridoids, together with four known analogues were isolated from the roots of Valeriana jatamansi. Their structures were elucidated by means of spectroscopic analyses (HRESIMS, NMR). The cytotoxicity of all isolates was evaluated. Compounds 5-7 exhibited selective cytotoxicity against HCT116 cells, with IC50 values of 9.3, 1.7 and 2.2 µM, respectively. The preliminary mechanistic study revealed that, the cytotoxicity effect of 6 was attributed to Akt/mTOR activation blockade via inhibition of PDK1 phosphorylation. Meanwhile, compound 6 could induce autophagosome formation in HCT116 cells via suppressing its downstream Akt/mTOR. These findings show that compound 6 could be of great importance to the development of anti-colon cancer agents.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Iridoids/pharmacology , Protein Kinase Inhibitors/pharmacology , Valerian/chemistry , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Iridoids/chemistry , Iridoids/isolation & purification , Models, Molecular , Molecular Structure , Plant Roots/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
7.
Proc Natl Acad Sci U S A ; 113(21): E2955-64, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27170189

ABSTRACT

Neurons migrate a long radial distance by a process known as locomotion in the developing mammalian neocortex. During locomotion, immature neurons undergo saltatory movement along radial glia fibers. The molecular mechanisms that regulate the speed of locomotion are largely unknown. We now show that the serine/threonine kinase Akt and its activator phosphoinositide-dependent protein kinase 1 (PDK1) regulate the speed of locomotion of mouse neocortical neurons through the cortical plate. Inactivation of the PDK1-Akt pathway impaired the coordinated movement of the nucleus and centrosome, a microtubule-dependent process, during neuronal migration. Moreover, the PDK1-Akt pathway was found to control microtubules, likely by regulating the binding of accessory proteins including the dynactin subunit p150(glued) Consistent with this notion, we found that PDK1 regulates the expression of cytoplasmic dynein intermediate chain and light intermediate chain at a posttranscriptional level in the developing neocortex. Our results thus reveal an essential role for the PDK1-Akt pathway in the regulation of a key step of neuronal migration.


Subject(s)
Cell Movement/physiology , Microtubules/metabolism , Neocortex/growth & development , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Dynactin Complex/genetics , Dynactin Complex/metabolism , Mice , Mice, Transgenic , Microtubules/genetics , Neocortex/cytology , Neurons/cytology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
8.
Molecules ; 24(7)2019 Apr 07.
Article in English | MEDLINE | ID: mdl-30959969

ABSTRACT

Ponatinib is a multi-target protein tyrosine kinase inhibitor, and its effects on hepatocellular carcinoma cells have not been previously explored. In the present study, we investigated its effects on hepatocellular carcinoma cell growth and the underlying mechanisms. Toward SK-Hep-1 and SNU-423 cells, ponatinib induces apoptosis by upregulation of cleaved caspase-3 and -7 and promotes cell cycle arrest in the G1 phase by inhibiting CDK4/6/CyclinD1 complex and phosphorylation of retinoblastoma protein. It inhibits the growth-stimulating mitogen-activated protein (MAP) kinase pathway, the phosphorylation of Src on both negative and positive regulation sites, and Jak2 and Stat3 phosphorylation. Surprisingly, it also activates the PDK1, the protein kinase B (Akt), and the mechanistic target of rapamycin (mTOR) signaling pathway. Blocking mTOR signaling strongly sensitizes cells to inhibition by ponatinib and makes ponatinib a much more potent inhibitor of hepatocellular carcinoma cell proliferation. These findings demonstrate that ponatinib exerts both positive and negative effects on hepatocellular cell proliferation, and eliminating its growth-stimulating effects by drug combination or potentially by chemical medication can significantly improve its efficacy as an anti-cancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Signal Transduction/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , TOR Serine-Threonine Kinases/metabolism
9.
Allergy ; 70(12): 1622-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26335809

ABSTRACT

BACKGROUND: Asthma is a heterogenetic disorder characterized by chronic inflammation with variable airflow obstruction and airway hyper-responsiveness. As the most potent and popular bronchodilators, ß2 adrenergic receptor (ß2 AR) agonists bind to the ß2 ARs that are coupled via a stimulatory G protein to adenylyl cyclase, thereby improving cAMP accumulation and resulting in airway smooth muscle relaxation. We previously demonstrated arctigenin had a synergistic function with the ß2 AR agonist, but the target for this remained elusive. METHOD: Chemical proteomics capturing was used to enrich and uncover the target of arctigenin in human bronchial smooth muscle cells, and reverse docking and molecular dynamic stimulation were performed to evaluate the binding of arctigenin and its target. In vitro enzyme activities and protein levels were demonstrated with special kits and Western blotting. Finally, guinea pig tracheal muscle segregation and ex vivo function were analysed. RESULTS: Arctigenin bound to PDK1 with an ideal binding free energy -25.45 kcal/mol and inhibited PDK1 kinase activity without changing its protein level. Additionally, arctigenin reduced PKB/Akt-induced phosphorylation of PDE4D, which was first identified in this study. Attenuation of PDE4D resulted in cAMP accumulation in human bronchial smooth muscle. The inhibition of PDK1 showed a synergistic function with ß2 AR agonists and relaxed the constriction of segregated guinea pig tracheal muscle. CONCLUSIONS: The PDK1/Akt/PDE4D axis serves as a novel asthma target, which may benefit airflow obstruction.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/metabolism , Bronchi/drug effects , Furans/pharmacology , Lignans/pharmacology , Signal Transduction/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Blotting, Western , Bronchi/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Synergism , Guinea Pigs , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism
10.
Toxics ; 12(1)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38251029

ABSTRACT

Ritonavir (RTV) is an antiviral and a component of COVID-19 treatments. Moreover, RTV demonstrates anti-cancer effects by suppressing AKT. However, RTV has cytotoxicity and suppresses sperm functions by altering AKT activity. Although abnormal AKT activity is known for causing detrimental effects on sperm functions, how RTV alters AKT signaling in spermatozoa remains unknown. Therefore, this study aimed to investigate reproductive toxicity of RTV in spermatozoa through phosphoinositide 3-kinase/phosphoinositide-dependent protein kinase-1/protein kinase B (PI3K/PDK1/AKT) signaling. Duroc spermatozoa were treated with various concentrations of RTV, and capacitation was induced. Sperm functions (sperm motility, motion kinematics, capacitation status, and cell viability) and expression levels of tyrosine-phosphorylated proteins and PI3K/PDK1/AKT pathway-related proteins were evaluated. In the results, RTV significantly suppressed sperm motility, motion kinematics, capacitation, acrosome reactions, and cell viability. Additionally, RTV significantly increased levels of phospho-tyrosine proteins and PI3K/PDK1/AKT pathway-related proteins except for AKT and PI3K. The expression level of AKT was not significantly altered and that of PI3K was significantly decreased. These results suggest RTV may suppress sperm functions by induced alterations of PI3K/PDK1/AKT pathway through abnormally increased tyrosine phosphorylation. Therefore, we suggest people who use or prescribe RTV need to consider its male reproductive toxicity.

11.
Toxicol In Vitro ; 99: 105848, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38772495

ABSTRACT

Nirmatrelvir (NMV) is a recently developed selective inhibitor of the main protease of Sars-Cov-2 that reduces the severity of infection. Despite its widespread use and various side effects, NMV's effect on male fertility is still unclear. This study was thus established to investigate how NMV affects male fertility. For experiments, Duroc spermatozoa were incubated with various concentrations of NMV (0, 0.1, 1, 10, 50, and 100 µM). Then, sperm motility, motion kinematics, capacitation status, intracellular ATP level, and cell viability were evaluated. In addition, the expression levels of phospho-PKA substrates, tyrosine-phosphorylated proteins, and PI3K/PDK1/AKT signaling pathway-related proteins were measured by western blotting. Our results showed that sperm motility, motion kinematics, proportion of capacitated spermatozoa, and intracellular ATP level were significantly decreased by NMV in a dose-dependent manner. Moreover, PKA activation was significantly suppressed by NMV, and expression levels of PI3K, phospho-PDK1, AKT, and phospho-AKT (Thr308 and Ser473) were significantly increased in a dose-dependent manner. Combining these findings, it is suggested that NMV has detrimental effects on sperm function by inducing abnormal changes in the PI3K/PDK1/AKT signaling pathway, resulting in PKA deactivation. Therefore, there is a need to pay particular attention to its male reproductive toxicity when NMV is administered.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Sperm Motility , Spermatozoa , Male , Proto-Oncogene Proteins c-akt/metabolism , Spermatozoa/drug effects , Signal Transduction/drug effects , Sperm Motility/drug effects , Animals , Phosphatidylinositol 3-Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Swine , Adenosine Triphosphate/metabolism , Sperm Capacitation/drug effects , Cell Survival/drug effects
12.
Reprod Toxicol ; 124: 108545, 2024 03.
Article in English | MEDLINE | ID: mdl-38246476

ABSTRACT

4-Nonylphenol (4-NP) is an endocrine-disrupting chemical that impairs animal and human reproduction. However, the mechanisms underlying male reproductive dysfunction by 4-NP have not been fully understood. Herein, we demonstrated the effects of 4-NP on boar sperm functions and molecular mechanisms. Spermatozoa were treated with various concentrations of 4-NP (0, 10, 25, 50, 75, and 100 µM) during capacitation. Then, we evaluated sperm motility, capacitation status, intracellular ATP level, and cell viability. Finally, we measured the expression of phosphorylated protein kinase A (PKA), tyrosine phosphorylation, and proteins related to the phosphatidylinositol 3 kinase (PI3K)/phosphoinositide-dependent kinase-1 (PDK1)/protein kinase B (AKT) signaling pathways following exposure to 4-NP. Sperm motility and motion kinematics were reduced by 4-NP, whereas intracellular ATP levels were increased significantly in a dose-dependent manner. Furthermore, the expression levels of p-PI3K, PTEN, p-PDK1, AKT, and p-AKT exhibited a significant dose-dependent increase. Moreover, abnormal activation of PKA and tyrosine phosphorylation were observed. Specifically, the ∼24 kDa p-PKA substrate demonstrated a significant reduction following exposure to 4-Np. In addition, the ∼18 kDa p-PKA substrate and tyrosine-phosphorylated proteins displayed a significant dose-dependent increase after exposure to 4-NP. Our results suggest that 4-NP may induce detrimental effects on sperm functions through abnormal changes in PKA activity and tyrosine phosphorylation during capacitation, possibly through unusual alteration of the PI3K/PDK1/AKT signaling pathway. Therefore, 4-NP must be cautiously used considering its reproductive toxicity.


Subject(s)
Phenols , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Male , Humans , Swine , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinase/pharmacology , Semen/metabolism , Sperm Motility , Signal Transduction , Spermatozoa , Phosphorylation , Cyclic AMP-Dependent Protein Kinases/metabolism , Tyrosine/metabolism , Adenosine Triphosphate/metabolism , Sperm Capacitation
13.
Reprod Toxicol ; 128: 108659, 2024 09.
Article in English | MEDLINE | ID: mdl-38972361

ABSTRACT

Oridonin, a natural terpenoid isolated from the leaves of Isodon rubescens (Hemsley) H.Hara, is widely used in oriental medicine for its anticancer properties across various cancer types. Despite its prevalent use, the toxic effects of oridonin on male reproduction, particularly its impact on sperm functions and the mechanisms involved, are not well understood. This study aimed to explore the effects and underlying mechanisms of oridonin on sperm functions. We initially treated Duroc boar spermatozoa with varying concentrations of oridonin (0, 5, 50, 75, 100, and 150 µM) and incubated them to induce capacitation. We then assessed cell viability and several sperm functions, including sperm motility and motion kinematics, capacitation status, and ATP levels. We also analyzed the expression levels of proteins associated with the phosphatidylinositol 3-kinase (PI3K)/phosphoinositide-dependent kinase-1 (PDK1)/protein kinase B (AKT) signaling pathway and phosphotyrosine proteins. Our results indicate that oridonin adversely affects most sperm functions in a dose-dependent manner. We observed significant decreases in AKT, p-AKT (Thr308), phosphatase and tensin homolog (PTEN), p-PDK1, and p-PI3K levels following oridonin treatment, alongside an abnormal increase in phosphotyrosine proteins. These findings suggest that oridonin may disrupt normal levels of tyrosine-phosphorylated proteins by inhibiting the PI3K/PDK1/AKT signaling pathway, which is crucial for cell proliferation, metabolism, and apoptosis, thus potentially harming sperm functions. Consequently, we recommend considering the reproductive toxicity of oridonin when using it as a therapeutic agent.


Subject(s)
Diterpenes, Kaurane , Signal Transduction , Sperm Motility , Spermatozoa , Animals , Male , Adenosine Triphosphate/metabolism , Cell Survival/drug effects , Diterpenes, Kaurane/adverse effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction/drug effects , Sperm Capacitation/drug effects , Sperm Motility/drug effects , Spermatozoa/drug effects , Swine
14.
Mol Med Rep ; 29(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38214335

ABSTRACT

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that the data panel for the "Huh7+BSA" experiment shown in Fig. 1D on p. 2852, showing the relative size of lipid droplets as determined in morphological studies using oil red O staining, had also appeared previously in the following article published by the same research group [Li D, Cheng M, Niu Y, Chi X, Liu X, Fan J, Fan H, Chang Y and Yang W: Identification of a novel human long non-coding RNA that regulates hepatic lipid metabolism by inhibiting SREBP-1c. Int J Biol Sci 13: 349-357, 2017]. Upon examining their original data, the authors have realized that this data panel was inadvertently selected incorrectly in Fig. 1, and the revised version of Fig. 1, containing the correct data panel for Fig. 1D, is shown on the next page. Note that this error did not significantly affect the results or the conclusions reported in this paper. All the authors agree to the publication of this Corrigendum, and are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to correct this error. Moreover, the authors apologize to the readership for any inconvenience caused. [Molecular Medicine Reports 18: 2850-2856, 2018; DOI: 10.3892/mmr.2018.9278].

15.
Genes (Basel) ; 14(4)2023 03 30.
Article in English | MEDLINE | ID: mdl-37107587

ABSTRACT

BACKGROUND: The Bcl2-associated athanogene4 (BAG4/SODD) protein could be identified as a tumor marker for several malignancies and plays a major role in the occurrence, development, and drug resistance of tumors. However, the role of Silencer of death domains (SODD) in lung carcinogenesis is still elusive. OBJECTIVE: To illuminate the effect of SODD on the proliferation, migration, invasion, and apoptosis of lung cancer cells and tumor growth in vivo and explore the corresponding mechanism. METHODS: The expression of SODD in tumor and normal tissues was determined and compared via western blot. SODD gene knockout lung cancer cells (H1299 cells) were established through a CRISPR/Cas9 gene deleting system, and a transient SODD overexpression of H1299 cells was also constructed. Then, cell proliferation and invasion were assessed through colony formation and cell counting kit-8 assays, transwell migration assays, and wound healing assays. Cell drug sensitivity is also analyzed by Cell Counting Kit-8 assay. The flow cytometer was used to perform cell circle and apoptosis analysis. The interaction of SODD and RAF-1 was confirmed by co-immunoprecipitation, and the phosphorylated level of Phosphatidylinositol 3-kinase (PI3K), Serine/threonine-protein kinase (AKT), Rapidly accelerated fibrosarcoma (RAF)-1,and extracellular signal regulated kinase (ERK) in cells was examined by western blot to evaluate the activation of PI3K/PDK1/AKT and RAF/MEK/ERK pathways. In vivo, Xenograft tumor assay of SODD knockout H1299 cells was used to evaluate further the role of SODD on the proliferation of H1299 cells. RESULTS: SODD binds to RAF-1 and is over-expressed in lung tissues, and promotes the proliferation, migration, invasion, and drug sensitivity of H1299 cells. The reduced cells in the S phase and increased cells arrested in the G2/M phase were found in SODD knockout H1299 cells, and more cells got apoptosis. The expression of 3-phosphoinositide-dependent protein kinase 1(PDK1) protein in SODD knockout H1299 cells decreases distinctively, and the phosphorylated level of AKT, RAF-1, and ERK-1 kinase in SODD knockout H1299 cells is also less than that in normal H1299 cells. In contrast, SODD overexpression significantly increases the phosphorylation of AKT. In vivo, SODD promotes the tumorigenicity of H1299 cells in nude mice. CONCLUSIONS: SODD is overexpressed in lung tissues and plays a considerable role in the development and progression of lung cancer by regulating the PI3K/PDK1/AKT and RAF/MEK/ERK pathways.


Subject(s)
Fibrosarcoma , Lung Neoplasms , Mice , Animals , Humans , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Mice, Nude , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Cell Transformation, Neoplastic , Mitogen-Activated Protein Kinase Kinases/metabolism
16.
J Cancer Res Clin Oncol ; 149(18): 16729-16739, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37725242

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) mutation is a prominent driver of lung cancer. Tyrosine kinase inhibitors (TKIs) have shown efficacy in treating EGFR-mutant lung cancer, but the emergence of drug resistance poses a significant challenge. Recent research has highlighted solute carrier family 12 member 8 (SLC12A8) as one of the highly upregulated genes in various cancer types. However, its oncogenic function remains largely unexplored. METHODS: 343 consecutive lung cancer patients were prospectively recruited and were followed for over 10 years. SLC12A8 expression in lung cancer tissues was measured by qPCR and was associated with patient survival. The association of SLC12A8 with TKI resistance was studied in in vitro EGFR-mutant lung cancer cell line as well as in in vivo xenograft tumor model. High-throughput kinome screening was employed to investigate SLC12A8-mediated oncogenic signaling pathway in lung cancer. RESULTS: SLC12A8 is a predictive biomarker of poor prognosis in lung cancer, particularly in patients with EGFR mutations. SLC12A8 overexpression diminishes the effectiveness of TKIs in EGFR-mutant lung cancer, resulting in treatment failure and disease progression. More importantly, SLC12A8-induced TKI resistance is mediated by the PDK1/AKT signaling axis, while silencing SLC12A8 expression inhibits oncogenic PDK1/AKT signaling, restoring TKI sensitivity in lung cancer cells. CONCLUSION: SLC12A8 mediates TKI resistance in EGFR-mutant lung cancer via PDK1/AKT axis. These findings not only advance our understanding of the molecular mechanisms driving TKI resistance, but also offer novel alternative strategies for the treatment of lung cancer.


Subject(s)
Lung Neoplasms , Protein Kinase Inhibitors , Sodium-Potassium-Chloride Symporters , Humans , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Sodium-Potassium-Chloride Symporters/genetics
17.
Pathol Res Pract ; 246: 154491, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37141697

ABSTRACT

Non-small cell lung cancer (NSCLC) ranks the most lethal malignancies around the world, nearly 85 % of lung cancers are NSCLC. Its high prevalence and morbidity pose a considerable burden to human health, identifying promising therapeutic targets for NSCLC is urgently needed. The essential function of long non-coding RNAs (lncRNAs) in multiple cellular progressions and pathophysiological processes are widely understood, thus we investigated the role of lncRNA T-cell leukemia/lymphoma 6 (TCL6) in NSCLC progression. LncRNA TCL6 level is increased in NSCLC samples and downregulation of lncRNA TCL6 inhibited NSCLC tumorigenesis. Moreover, Scratch Family Transcriptional Repressor 1 (SCRT1) can modulate lncRNA TCL6 expression in NSCLC cells, with lncRNA TCL6 promoting NSCLC development through Pyruvate Dehydrogenase Kinase 1 (PDK1)/AKT signaling by interacting with PDK1, thereby providing a novel framework for NSCLC research.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics
18.
Pharmaceuticals (Basel) ; 16(2)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-37259393

ABSTRACT

In a preliminary study, we synthesized a series of new PDK1/MEK dual inhibitors. Antitumor activity screening showed that Compound YZT exerts a strong inhibitory action in A549 cells. However, the specific mechanism of YZT against non-small cell lung cancer (NSCLC) is largely unknown. This work confirmed the anti-proliferation and pro-apoptosis effects of YZT in NSCLC cells. Furthermore, YZT promotes autophagy and provokes complete autophagic flux in NSCLC cells. Notably, compared with YZT alone, the combination of YZT with the autophagy inhibitor chloroquine (CQ) or 3-methyladenine (3-MA) markedly strengthened the anti-proliferative and pro-apoptotic actions, suggesting that YZT-induced autophagy is cytoprotective. We further found that YZT-induced autophagy may exert a cytoprotective function by preserving the integrity of mitochondria and decreasing mitochondrial apoptosis. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis suggested that PDK1 is an upstream protein of the Akt/mTOR axis and western blotting verified that YZT induces autophagy by the PDK1/Akt/mTOR signaling axis. Finally, YZT plus CQ significantly enhanced the anticancer activities compared to YZT alone in an animal study and immunohistochemistry showed that the level of LC3 was increased by YZT, which is in line with the in vitro results. In short, our study provides reliable experimental basis for developing Compound YZT as a new chemotherapeutic drug candidate and suggests that combined administration of YZT with CQ is a potential therapy against NSCLC.

19.
Reprod Toxicol ; 113: 103-109, 2022 10.
Article in English | MEDLINE | ID: mdl-35973673

ABSTRACT

Glucose-regulated protein 78 (GRP78), which is commonly found in the endoplasmic reticulum (ER), is involved in stabilizing ER proteins and inducing the unfolded protein response. Furthermore, GRP78 is expressed on the surface of most common cancer cells, such as cells of breast, lung, liver, and prostate cancers, and plays a role in apoptosis and cell proliferation via the PI3K/PDK1/AKT signaling pathway. Therefore, various trials have been performed for evaluating cancer treatment by inhibiting GRP78. Moreover, GRP78 is expressed on the surface of spermatozoa; however, its role in spermatozoa physiology remains unclear. Therefore, this study was designed to investigate the effects of GRP78 on sperm function during capacitation and elucidate the underlying mechanisms. Boar spermatozoa were exposed to various concentrations of HA15, a GRP78 antagonist, and sperm kinematic parameters, capacitation status, cell viability, levels of PI3K/PDK1/AKT-pathway related proteins, and tyrosine phosphorylation were evaluated. GRP78 inhibition significantly decreased sperm motility, kinematic parameters, capacitated and acrosome-reacted spermatozoa counts, and cell viability. Moreover, GRP78 expression was significantly decreased in HA15-treated spermatozoa compared to that in the control group, and levels of PI3K/PDK1/AKT-pathway related proteins changed significantly. Furthermore, tyrosine phosphorylation was significantly altered in the HA15-treated group. The results of this study suggest that GRP78 inhibition in cancer therapy may negatively affect sperm function. These results lay a strong foundation for future studies aiming to identify the molecular mechanisms related to GRP78 in spermatozoa.


Subject(s)
Sperm Capacitation , Sperm Motility , Animals , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Semen/metabolism , Spermatozoa , Swine , Tyrosine/metabolism
20.
Redox Biol ; 38: 101774, 2021 01.
Article in English | MEDLINE | ID: mdl-33152664

ABSTRACT

Skeletal muscle atrophy with high prevalence can induce weakness and fatigability and place huge burden on both health and quality of life. During skeletal muscle degeneration, excessive fibroblasts and extracellular matrix (ECM) accumulated to replace and impair the resident muscle fiber and led to loss of muscle mass. Cyclooxygenase-2 (COX-2), the rate-limiting enzyme in synthesis of prostaglandin, has been identified as a positive regulator in pathophysiological process like inflammation and oxidative stress. In our study, we found injured muscles of human subjects and mouse model overexpressed COX-2 compared to the non-damaged region and COX-2 was also upregulated in fibroblasts following TGF-ß stimulation. Then we detected the effect of selective COX-2 inhibitor celecoxib on fibrogenesis. Celecoxib mediated anti-fibrotic effect by inhibiting fibroblast differentiation, proliferation and migration as well as inactivating TGF-ß-dependent signaling pathway, non-canonical TGF-ß pathways and suppressing generation of reactive oxygen species (ROS) and oxidative stress. In vivo pharmacological inhibition of COX-2 by celecoxib decreased tissue fibrosis and increased skeletal muscle fiber preservation reflected by less ECM formation and myofibroblast accumulation with decreased p-ERK1/2, p-Smad2/3, TGF-ßR1, VEGF, NOX2 and NOX4 expression. Expression profiling further found that celecoxib could suppress PDK1 expression. The interaction between COX-2 and PDK1/AKT signaling remained unclear, here we found that COX-2 could bind to PDK1/AKT to form compound. Knockdown of COX-2 in fibroblasts by pharmacological inactivation or by siRNA restrained PDK1 expression and AKT phosphorylation induced by TGF-ß treatment. Besides, si-COX-2 prevented TGF-ß-induced K63-ubiquitination of AKT by blocking the interaction between AKT and E3 ubiquitin ligase TRAF4. In summary, we found blocking COX-2 inhibited fibrogenesis after muscle atrophy induced by injury and suppressed AKT signaling pathway by inhibiting upstream PDK1 expression and preventing the recruitment of TRAF4 to AKT, indicating that COX-2/PDK1/AKT signaling pathway promised to be target for treating muscle atrophy in the future.


Subject(s)
Cyclooxygenase 2 Inhibitors , Muscle, Skeletal/pathology , Muscular Atrophy , Proto-Oncogene Proteins c-akt , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , TNF Receptor-Associated Factor 4 , Animals , Celecoxib , Cyclooxygenase 2/genetics , Gene Silencing , Humans , Mice , Muscle, Skeletal/metabolism , Muscular Atrophy/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Quality of Life , Signal Transduction , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL