Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38743208

ABSTRACT

Non-small cell lung cancer (NSCLC) is a common cancer with several accepted treatments, such as chemotherapy, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, and immune checkpoint inhibitors. Nevertheless, NSCLC cells often become insensitive to these treatments, and therapeutic resistance is a major reason NSCLC still has a high mortality rate. The induction of therapeutic resistance in NSCLC often involves hedgehog, and suppression of hedgehog can increase NSCLC cell sensitivity to several conventional therapies. In our previous work, we demonstrated that the marine antimicrobial peptide tilapia piscidin 4 (TP4) exhibits potent anti-NSCLC activity in both EGFR-WT and EGFR-mutant NSCLC cells. Here, we sought to further explore whether hedgehog might influence the sensitivity of NSCLC cells to TP4. Our results showed that hedgehog was activated by TP4 in both WT and EGFR-mutant NSCLC cells and that pharmacological inhibition of hedgehog by vismodegib, a Food and Drug Administration-approved hedgehog inhibitor, potentiated TP4-induced cytotoxicity. Mechanistically, vismodegib acted by enhancing TP4-mediated increases in mitochondrial membrane potential and intracellular reactive oxygen species (ROS). MitoTempo, a specific mitochondrial ROS scavenger, abolished vismodegib/TP4 cytotoxicity. The combination of vismodegib with TP4 also reduced the levels of the antioxidant proteins catalase and superoxide dismutase, and it diminished the levels of chemoresistance-related proteins, Bcl-2 and p21. Thus, we conclude that hedgehog regulates the cytotoxic sensitivity of NSCLC cells to TP4 by protecting against mitochondrial dysfunction and suppressing oxidative stress. These findings suggest that combined treatment of vismodegib and TP4 may be a promising therapeutic strategy for NSCLC.

2.
Bone Joint Res ; 13(4): 157-168, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38569602

ABSTRACT

Aims: Osteosarcoma is the most common primary bone malignancy among children and adolescents. We investigated whether benzamil, an amiloride analogue and sodium-calcium exchange blocker, may exhibit therapeutic potential for osteosarcoma in vitro. Methods: MG63 and U2OS cells were treated with benzamil for 24 hours. Cell viability was evaluated with the MTS/PMS assay, colony formation assay, and flow cytometry (forward/side scatter). Chromosome condensation, the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay, cleavage of poly-ADP ribose polymerase (PARP) and caspase-7, and FITC annexin V/PI double staining were monitored as indicators of apoptosis. Intracellular calcium was detected by flow cytometry with Fluo-4 AM. The phosphorylation and activation of focal adhesion kinase (FAK) and signal transducer and activator of transcription 3 (STAT3) were measured by western blot. The expression levels of X-linked inhibitor of apoptosis protein (XIAP), B-cell lymphoma 2 (Bcl-2), B-cell lymphoma-extra large (Bcl-xL), SOD1, and SOD2 were also assessed by western blot. Mitochondrial status was assessed with tetramethylrhodamine, ethyl ester (TMRE), and intracellular adenosine triphosphate (ATP) was measured with BioTracker ATP-Red Live Cell Dye. Total cellular integrin levels were evaluated by western blot, and the expression of cell surface integrins was assessed using fluorescent-labelled antibodies and flow cytometry. Results: Benzamil suppressed growth of osteosarcoma cells by inducing apoptosis. Benzamil reduced the expression of cell surface integrins α5, αV, and ß1 in MG63 cells, while it only reduced the expression of αV in U2OS cells. Benzamil suppressed the phosphorylation and activation of FAK and STAT3. In addition, mitochondrial function and ATP production were compromised by benzamil. The levels of anti-apoptotic proteins XIAP, Bcl-2, and Bcl-xL were reduced by benzamil. Correspondingly, benzamil potentiated cisplatin- and methotrexate-induced apoptosis in osteosarcoma cells. Conclusion: Benzamil exerts anti-osteosarcoma activity by inducing apoptosis. In terms of mechanism, benzamil appears to inhibit integrin/FAK/STAT3 signalling, which triggers mitochondrial dysfunction and ATP depletion.

3.
Int J Hyperthermia ; 41(1): 2310017, 2024.
Article in English | MEDLINE | ID: mdl-38350654

ABSTRACT

Objective: Gastric cancer with peritoneal metastasis is considered to be final stage gastric cancer. One current treatment approach for this condition is combined cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC). However, the therapeutic mechanisms of HIPEC remain largely undescribed. Method: In order to assess the cellular effects of HIPEC in vitro, we treated AGS human gastric adenocarcinoma cells with or without 5-fluorouracil (5-Fu) at 37 °C or at 43 °C (hyperthermic temperature) for 1 h followed by incubation at 37 °C for 23 h. The impacts of hyperthermia/5-Fu on apoptosis, cell survival signals, oxidative stress, chemoresistance-related proteins and programmed death-ligand 1 (PD-L1) expression were measured. Results: Our results showed that hyperthermia potentiates 5-Fu-mediated cytotoxicity in AGS cells. Furthermore, the combination of 5-Fu and hyperthermia reduces levels of both phosphorylated STAT3 and STAT3, while increasing the levels of phosphorylated Akt and ERK. In addition, 5-Fu/hyperthermia enhances reactive oxygen species and suppresses superoxide dismutase 1. Chemoresistance-related proteins, such as multidrug resistance 1 and thymidylate synthase, are also suppressed by 5-Fu/hyperthermia. Interestingly, hyperthermia enhances 5-Fu-mediated induction of glycosylated PD-L1, but 5-Fu-mediated upregulation of PD-L1 surface expression is prevented by hyperthermia. Conclusion: Taken together, our findings provide insights that may aid in the development of novel therapeutic strategies and enhanced therapeutic efficacy of HIPEC.


Subject(s)
Hyperthermia, Induced , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , B7-H1 Antigen/therapeutic use , Drug Resistance, Neoplasm , Hyperthermia, Induced/methods , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy
4.
Cell Metab ; 35(10): 1782-1798.e8, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37586363

ABSTRACT

Glucose metabolism is known to orchestrate oncogenesis. Whether glucose serves as a signaling molecule directly regulating oncoprotein activity for tumorigenesis remains elusive. Here, we report that glucose is a cofactor binding to methyltransferase NSUN2 at amino acid 1-28 to promote NSUN2 oligomerization and activation. NSUN2 activation maintains global m5C RNA methylation, including TREX2, and stabilizes TREX2 to restrict cytosolic dsDNA accumulation and cGAS/STING activation for promoting tumorigenesis and anti-PD-L1 immunotherapy resistance. An NSUN2 mutant defective in glucose binding or disrupting glucose/NSUN2 interaction abolishes NSUN2 activity and TREX2 induction leading to cGAS/STING activation for oncogenic suppression. Strikingly, genetic deletion of the glucose/NSUN2/TREX2 axis suppresses tumorigenesis and overcomes anti-PD-L1 immunotherapy resistance in those cold tumors through cGAS/STING activation to facilitate apoptosis and CD8+ T cell infiltration. Our study identifies NSUN2 as a direct glucose sensor whose activation by glucose drives tumorigenesis and immunotherapy resistance by maintaining TREX2 expression for cGAS/STING inactivation.


Subject(s)
Nucleotidyltransferases , Signal Transduction , Humans , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction/genetics , Carcinogenesis , Immunotherapy , Methyltransferases/metabolism
5.
J Biol Chem ; 299(4): 104570, 2023 04.
Article in English | MEDLINE | ID: mdl-36870679

ABSTRACT

Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies have uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.


Subject(s)
AMP-Activated Protein Kinases , Neoplasms , Protein Processing, Post-Translational , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Liver/metabolism , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Neoplasms/enzymology
6.
Semin Cancer Biol ; 86(Pt 3): 1158-1174, 2022 11.
Article in English | MEDLINE | ID: mdl-36244530

ABSTRACT

Metabolic reprogramming is an important cancer hallmark that plays a key role in cancer malignancies and therapy resistance. Cancer cells reprogram the metabolic pathways to generate not only energy and building blocks but also produce numerous key signaling metabolites to impact signaling and epigenetic/transcriptional regulation for cancer cell proliferation and survival. A deeper understanding of the mechanisms by which metabolic reprogramming is regulated in cancer may provide potential new strategies for cancer targeting. Recent studies suggest that deregulated transcription factors have been observed in various human cancers and significantly impact metabolism and signaling in cancer. In this review, we highlight the key transcription factors that are involved in metabolic control, dissect the crosstalk between signaling and transcription factors in metabolic reprogramming, and offer therapeutic strategies targeting deregulated transcription factors for cancer treatment.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplasms/pathology , Metabolic Networks and Pathways
7.
ACS Omega ; 7(30): 26015-26020, 2022 Aug 02.
Article in English | MEDLINE | ID: mdl-35936457

ABSTRACT

PROTACs represent a promising modality that has gained significant attention for the treatment of cancer, Alzheimer's disease, and so forth. Due to limited structural information of the POI-PROTAC-E3 ligase ternary complex, the discovery of active PROTACs relies on the screening of diversity-oriented PROTAC libraries. VH032 amine is a key building block for the synthesis of VHL E3 ligase-based PROTACs. To construct VHL PROTAC libraries rapidly, the availability of VH032 amine is crucial. In this paper, we report a column chromatography-free process which enables the production of 42.5 g of VH032 amine hydrochloride in 65% overall yield with 97% purity in a week.

8.
J Clin Invest ; 132(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35229723

ABSTRACT

Cancer stem-like cells (CSLCs) acquire enhanced immune checkpoint responses to evade immune cell killing and promote tumor progression. Here we showed that signal regulatory protein γ (SIRPγ) determined CSLC properties and immune evasiveness in a small population of lung adenocarcinoma (LUAD) cancer cells. A SIRPγhi population displayed CSLC properties and transmitted the immune escape signal through sustaining CD47 expression in both SIRPγhi and SIRPγlo/- tumor cells. SIRPγ bridged MST1 and PP2A to facilitate MST1 dephosphorylation, resulting in Hippo/YAP activation and leading to cytokine release by CSLCs, which stimulated CD47 expression in LUAD cells and consequently inhibited tumor cell phagocytosis. SIRPγ promoted tumor growth and metastasis in vivo through YAP signaling. Notably, SIRPγ targeting with genetic SIRPγ knockdown or a SIRPγ-neutralizing antibody inhibited CSLC phenotypes and elicited phagocytosis that suppressed tumor growth in vivo. SIRPG was upregulated in human LUAD and its overexpression predicted poor survival outcome. Thus, SIRPγhi cells serve as CSLCs and tumor immune checkpoint-initiating cells, propagating the immune escape signal to the entire cancer cell population. Our study identifies Hippo/YAP signaling as the first mechanism by which SIRPγ is engaged and reveals that targeting SIRPγ represents an immune- and CSLC-targeting strategy for lung cancer therapy.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/metabolism , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Line, Tumor , Hippo Signaling Pathway , Humans , Lung Neoplasms/genetics , Signal Transduction
9.
Cells ; 11(3)2022 01 30.
Article in English | MEDLINE | ID: mdl-35159290

ABSTRACT

Parkinson's disease (PD) is an age-related neurodegenerative disease caused by a selective loss of dopaminergic (DA) neurons in the substantia nigra (SN). Microglial activation is implicated in the pathogenesis of PD. This study aimed to characterize the role of microglial activation in aging-related nigral DA neuron loss and motor deficits in mice. We showed that, compared to 3-month-old mice, the number of DA neurons in the SN and the expression of dopamine transporter (DAT) in the striatum decreased during the period of 9 to 12 months of age. Motor deficits and microglial activation in the SN were also evident during these months. The number of DA neurons was negatively correlated with the degrees of microglial activation. The inhibition of age-related microglial activation by ibuprofen during these 3 months decreased DA neuron loss in the SN. Eliminating the microglia prevented systemic inflammation-induced DA neuron death. Forcing mice to run during these 3 months inhibited microglial activation and DA neuron loss. Blocking the brain-derived neurotrophic factor (BDNF) signaling eliminated the exercise-induced protective effects. In conclusion, nigral DA neurons were susceptible to local microglial activation. Running exercise upregulated BDNF-TrkB signaling and inhibited microglial activation during aging. Long-term exercise can be considered as a non-pharmacological strategy to ameliorate microglial activation and related neurodegeneration.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dopaminergic Neurons/metabolism , Mice , Microglia/metabolism , Nerve Degeneration/pathology , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism
10.
Mol Cell ; 81(18): 3803-3819.e7, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547240

ABSTRACT

Mitochondrial dynamics regulated by mitochondrial fusion and fission maintain mitochondrial functions, whose alterations underline various human diseases. Here, we show that inositol is a critical metabolite directly restricting AMPK-dependent mitochondrial fission independently of its classical mode as a precursor for phosphoinositide generation. Inositol decline by IMPA1/2 deficiency elicits AMPK activation and mitochondrial fission without affecting ATP level, whereas inositol accumulation prevents AMPK-dependent mitochondrial fission. Metabolic stress or mitochondrial damage causes inositol decline in cells and mice to elicit AMPK-dependent mitochondrial fission. Inositol directly binds to AMPKγ and competes with AMP for AMPKγ binding, leading to restriction of AMPK activation and mitochondrial fission. Our study suggests that the AMP/inositol ratio is a critical determinant for AMPK activation and establishes a model in which AMPK activation requires inositol decline to release AMPKγ for AMP binding. Hence, AMPK is an inositol sensor, whose inactivation by inositol serves as a mechanism to restrict mitochondrial fission.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Inositol/metabolism , Mitochondrial Dynamics/physiology , AMP-Activated Protein Kinases/physiology , Animals , Cell Line , Humans , Inositol/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , PC-3 Cells , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Stress, Physiological/physiology
11.
Mol Cell ; 80(2): 263-278.e7, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33022274

ABSTRACT

Cancer metastasis accounts for the major cause of cancer-related deaths. How disseminated cancer cells cope with hostile microenvironments in secondary site for full-blown metastasis is largely unknown. Here, we show that AMPK (AMP-activated protein kinase), activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain TCA cycle (tricarboxylic acid cycle) and promotes cancer metastasis by adapting cancer cells to metabolic and oxidative stresses. This AMPK-PDHc axis is activated in advanced breast cancer and predicts poor metastasis-free survival. Mechanistically, AMPK localizes in the mitochondrial matrix and phosphorylates the catalytic alpha subunit of PDHc (PDHA) on two residues S295 and S314, which activates the enzymatic activity of PDHc and alleviates an inhibitory phosphorylation by PDHKs, respectively. Importantly, these phosphorylation events mediate PDHc function in cancer metastasis. Our study reveals that AMPK-mediated PDHA phosphorylation drives PDHc activation and TCA cycle to empower cancer cells adaptation to metastatic microenvironments for metastasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Citric Acid Cycle , Pyruvate Dehydrogenase Complex/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Cell Survival , Enzyme Activation , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Phosphoserine/metabolism , Signal Transduction , Stress, Physiological , Survival Analysis
12.
J Neuroinflammation ; 17(1): 210, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32664974

ABSTRACT

BACKGROUND: Excessive microglial activation is implicated in the pathogenesis of various age-related neurodegenerative diseases. In addition to neurons, brain-derived neurotrophic factor (BDNF) and its receptor TrkB are also expressed in microglia. However, the direct effect of BDNF on age-related microglial activation has rarely been investigated. METHODS: We began to address this question by examining the effect of age on microglial activation and the BDNF-TrkB pathway in mice. By using pharmacological and genetic approaches, the roles of BDNF and downstream signaling pathways in microglial activation and related neurotoxicity were examined in microglial cell line and primary microglial cells. RESULTS: We showed that microglial activation was evident in the brains of aged mice. The levels of BDNF and TrkB in microglia decreased with age and negatively correlated with their activation statuses in mice during aging. Interestingly, aging-related microglial activation could be reversed by chronic, subcutaneous perfusion of BDNF. Peripheral lipopolysaccharide (LPS) injection-induced microglial activation could be reduced by local supplement of BDNF, while shTrkB induced local microglial activation in naïve mice. In cultured microglial cell line and primary microglial cells, BDNF inhibited LPS-induced microglial activation, including morphological changes, activations of p38, JNK, and NF-кB, and productions of proinflammatory cytokines. These effects were blocked by shTrkB. BDNF induced activations of ErK and CREB which then competed with LPS-induced activation of NF-кB for binding to a common coactivator, CREB-binding protein. CONCLUSIONS: Decreasing BDNF-TrkB signaling during aging favors microglial activation, while upregulation BDNF signaling inhibits microglial activation via the TrkB-Erk-CREB pathway.


Subject(s)
Aging/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Membrane Glycoproteins/metabolism , Microglia/metabolism , Protein-Tyrosine Kinases/metabolism , Aging/drug effects , Aging/pathology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology
13.
Semin Cancer Biol ; 67(Pt 2): 16-33, 2020 12.
Article in English | MEDLINE | ID: mdl-32014608

ABSTRACT

Strictly regulated protein degradation by ubiquitin-proteasome system (UPS) is essential for various cellular processes whose dysregulation is linked to serious diseases including cancer. Skp2, a well characterized component of Skp2-SCF E3 ligase complex, is able to conjugate both K48-linked ubiquitin chains and K63-linked ubiquitin chains on its diverse substrates, inducing proteasome mediated proteolysis or modulating the function of tagged substrates respectively. Overexpression of Skp2 is observed in various human cancers associated with poor survival and adverse therapeutic outcomes, which in turn suggests that Skp2 engages in tumorigenic activity. To that end, the oncogenic properties of Skp2 are demonstrated by various genetic mouse models, highlighting the potential of Skp2 as a target for tackling cancer. In this article, we will describe the downstream substrates of Skp2 as well as upstream regulators for Skp2-SCF complex activity. We will further summarize the comprehensive oncogenic functions of Skp2 while describing diverse strategies and therapeutic platforms currently available for developing Skp2 inhibitors.


Subject(s)
Drug Resistance, Neoplasm/genetics , Molecular Targeted Therapy/methods , Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , Animals , Carcinogens , Female , Gene Expression Regulation , Humans , Male , Metabolic Networks and Pathways , Mice , Neoplasms/drug therapy , Neoplasms/genetics , S-Phase Kinase-Associated Proteins/genetics , Ubiquitination
14.
Cell ; 178(1): 176-189.e15, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155231

ABSTRACT

RLR-mediated type I IFN production plays a pivotal role in elevating host immunity for viral clearance and cancer immune surveillance. Here, we report that glycolysis, which is inactivated during RLR activation, serves as a barrier to impede type I IFN production upon RLR activation. RLR-triggered MAVS-RIG-I recognition hijacks hexokinase binding to MAVS, leading to the impairment of hexokinase mitochondria localization and activation. Lactate serves as a key metabolite responsible for glycolysis-mediated RLR signaling inhibition by directly binding to MAVS transmembrane (TM) domain and preventing MAVS aggregation. Notably, lactate restoration reverses increased IFN production caused by lactate deficiency. Using pharmacological and genetic approaches, we show that lactate reduction by lactate dehydrogenase A (LDHA) inactivation heightens type I IFN production to protect mice from viral infection. Our study establishes a critical role of glycolysis-derived lactate in limiting RLR signaling and identifies MAVS as a direct sensor of lactate, which functions to connect energy metabolism and innate immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Lactic Acid/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Animals , Female , Glycolysis , HEK293 Cells , Humans , Interferon-beta/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RAW 264.7 Cells , Receptors, Immunologic , Signal Transduction/drug effects , Transfection
15.
Cancer Med ; 8(8): 3949-3964, 2019 07.
Article in English | MEDLINE | ID: mdl-31145545

ABSTRACT

Testicular cancer is the most commonly diagnosed cancer in men at 15-44 years of age, and radical orchidectomy combined with chemotherapy is currently considered as the standard treatment. However, drugs resistance and side effects that impact the quality of life for patients with testicular cancer have not been markedly improved in recent decades. In this study, we characterized the pharmacological exacerbation of the unfolded protein response (UPR), which is an effective approach to kill testicular cancer cells, by carrying out a clustering analysis of mRNA expression profiles and the immunobloting examination of cordycepin-treated MA-10 cells. The UPR is executed in response to endoplasmic reticulum stress to complement by an apoptotic response if the defect cannot be resolved. Results showed that cordycepin significantly modulated FoxO/P15/P27, PERK-eIF2α (apoptotic), and the IRE1-XBP1 (adaptive) UPR pathways. Interestingly, a fraction of MA-10 cells survived after cordycepin treatment, the AKT, LC3 I/II, and MAPK signaling pathways were highly induced in attached cells as compared to the suspended cells, illustrating the drug resistance to cordycepin via activating AKT and MAPK pathways in MA-10 cells. In summary, PERK-eIF2α signaling pathway is required for pro-apoptotic UPR in MA-10 cell death following cordycepin treatment, suggesting a potential therapeutic application in treating testicular cancer. However, activation of AKT and MAPK pathways could possibly result in drug resistance to cordycepin in MA-10 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxyadenosines/pharmacology , Drug Resistance, Neoplasm , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Unfolded Protein Response/drug effects , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Gene Expression Profiling , Male , Mice , Models, Biological , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
16.
Cell Discov ; 4: 54, 2018.
Article in English | MEDLINE | ID: mdl-30302273

ABSTRACT

The century-old embryonal/gametogenesis hypothesis of tumors could link diverse tumors' malignant features together likely representing the real "stemness" of tumors. However, the genetic evidence to validate abnormal gametogenesis in tumors remains lacking. Here we show that p53 deficiency elicits abnormal gametogenesis from primordial germ cell-like stage to late oocyte-like stage and subsequent parthenogenetic activation. The similar upregulation of abnormal gametogenesis by p53 deficiency is observed both in p53-/- mouse model and cultured cancer cells. Notably, germ cell-like cells isolated from distinct tumors from p53-/- mice and cancer cell lines display potent tumorigenicity potential. Abnormal oogenesis induced by p53 deficiency and then spontaneous parthenogenetic activation endow tumors with imitated embryonic development, life cycle, and therapeutic resistance. Our study establishes the genetic evidence to support embryonal/gametogenesis theory of tumors and reveals a pivotal role of p53 in restricting abnormal gametogenesis that may represent a novel aspect for p53's tumor suppression.

17.
Cancer Sci ; 109(11): 3503-3518, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30191630

ABSTRACT

Fibroblast growth factor 9 (FGF9) promotes cancer progression; however, its role in cell proliferation related to tumorigenesis remains elusive. We investigated how FGF9 affected MA-10 mouse Leydig tumor cell proliferation and found that FGF9 significantly induced cell proliferation by activating ERK1/2 and retinoblastoma (Rb) phosphorylations within 15 minutes. Subsequently, the expressions of E2F1 and the cell cycle regulators: cyclin D1, cyclin E1 and cyclin-dependent kinase 4 (CDK4) in G1 phase and cyclin A1, CDK2 and CDK1 in S-G2 /M phases were increased at 12 hours after FGF9 treatment; and cyclin B1 in G2 /M phases were induced at 24 hours after FGF9 stimulation, whereas the phosphorylations of p53, p21 and p27 were not affected by FGF9. Moreover, FGF9-induced effects were inhibited by MEK inhibitor PD98059, indicating FGF9 activated the Rb/E2F pathway to accelerate MA-10 cell proliferation by activating ERK1/2. Immunoprecipitation assay and ChIP-quantitative PCR results showed that FGF9-induced Rb phosphorylation led to the dissociation of Rb-E2F1 complexes and thereby enhanced the transactivations of E2F1 target genes, Cyclin D1, Cyclin E1 and Cyclin A1. Silencing of FGF receptor 2 (FGFR2) using lentiviral shRNA inhibited FGF9-induced ERK1/2 phosphorylation and cell proliferation, indicating that FGFR2 is the obligate receptor for FGF9 to bind and activate the signaling pathway in MA-10 cells. Furthermore, in a severe combined immunodeficiency mouse xenograft model, FGF9 significantly promoted MA-10 tumor growth, a consequence of increased cell proliferation and decreased apoptosis. Conclusively, FGF9 interacts with FGFR2 to activate ERK1/2, Rb/E2F1 and cell cycle pathways to induce MA-10 cell proliferation in vitro and tumor growth in vivo.


Subject(s)
E2F1 Transcription Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 9/metabolism , Leydig Cell Tumor/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Retinoblastoma Protein/metabolism , Testicular Neoplasms/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Male , Mice , Phosphorylation , Signal Transduction
18.
Onco Targets Ther ; 11: 1475-1490, 2018.
Article in English | MEDLINE | ID: mdl-29588601

ABSTRACT

BACKGROUND: Midazolam (MDZ) has powerful hypnosis, amnesia, anti-anxiety and anticonvulsant effects. Studies have shown that prenatally developmental toxicity of diazepam can be observed in many organs/tissues. However, it remains elusive in male reproductive system. MATERIALS AND METHODS: TM3 mouse Leydig progenitor cell line was used to determine whether MDZ has any unfavorable effects. RESULTS: Midazolam significantly decreased cell viability in dose- and time-dependent manners in TM3 cells. In flow cytometry analysis, midazolam significantly increased subG1 phase cell numbers, and annexin V/PI double staining assay further confirmed that MDZ induced apoptosis in TM3 cells. Moreover, MDZ significantly induced the expression of caspase-8 and -3 proteins and the phosphorylation of JNK, ERK1/2 and p38. Besides, MDZ didn't activate Akt pathway in TM3 cells. Furthermore, the expressions of p-EIF2α, ATF4, ATF3 and CHOP were induced by midazolam, suggesting that midazolam could induce apoptosis through endoplasmic reticulum (ER) stress in TM3 cells. Additionally, the expressions of cyclin A, cyclin B and CDK1 were inhibited by midazolam through the regulation of p53 in TM3 cells, indicating that midazolam could regulate cell cycle to induce apoptosis. CONCLUSION: Midazolam could activate caspase, MAPKs and ER stress pathways and impede Akt pathway and cell cycle to induce apoptosis in TM3 mouse Leydig progenitor cells.

19.
J Food Drug Anal ; 25(1): 197-205, 2017 01.
Article in English | MEDLINE | ID: mdl-28911537

ABSTRACT

Cordyceps sinensis has various biological and pharmacological functions, and it has been claimed as a tonic supplement for sexual and reproductive dysfunctions for a long time in oriental society. In this article, the in vitro and in vivo effects of C. sinensis and cordycepin on mouse Leydig cell steroidogenesis are briefly described, the stimulatory mechanisms are summarized, and the recent findings related to the alternative substances regulating male reproductive functions are also discussed.


Subject(s)
Cordyceps , Animals , Deoxyadenosines , Male , Reproduction
20.
Nat Commun ; 8: 14799, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28300060

ABSTRACT

Dynamic changes in histone modifications under various physiological cues play important roles in gene transcription and cancer. Identification of new histone marks critical for cancer development is of particular importance. Here we show that, in a glucose-dependent manner, E3 ubiquitin ligase NEDD4 ubiquitinates histone H3 on lysine 23/36/37 residues, which specifically recruits histone acetyltransferase GCN5 for subsequent H3 acetylation. Genome-wide analysis of chromatin immunoprecipitation followed by sequencing reveals that NEDD4 regulates glucose-induced H3 K9 acetylation at transcription starting site and enhancer regions. Integrative analysis of ChIP-seq and microarray data sets also reveals a consistent role of NEDD4 in transcription activation and H3 K9 acetylation in response to glucose. Functionally, we show that NEDD4-mediated H3 ubiquitination, by transcriptionally activating IL1α, IL1ß and GCLM, is important for tumour sphere formation. Together, our study reveals the mechanism for glucose-induced transcriptome reprograming and epigenetic regulation in cancer by inducing NEDD4-dependent H3 ubiquitination.


Subject(s)
Carcinogenesis/metabolism , Histones/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Ubiquitination , Acetylation/drug effects , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glucose/pharmacology , HEK293 Cells , Humans , Lysine/metabolism , Mice, Nude , Nedd4 Ubiquitin Protein Ligases/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...