Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Proc Natl Acad Sci U S A ; 120(39): e2308079120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37733743

ABSTRACT

TAK1 is a key modulator of both NF-κB signaling and RIPK1. In TNF signaling pathway, activation of TAK1 directly mediates the phosphorylation of IKK complex and RIPK1. In a search for small molecule activators of RIPK1-mediated necroptosis, we found R406/R788, two small molecule analogs that could promote sustained activation of TAK1. Treatment with R406 sensitized cells to TNF-mediated necroptosis and RIPK1-dependent apoptosis by promoting sustained RIPK1 activation. Using click chemistry and multiple biochemical binding assays, we showed that treatment with R406 promotes the activation of TAK1 by directly binding to TAK1, independent of its original target Syk kinase. Treatment with R406 promoted the ubiquitination of TAK1 and the interaction of activated TAK1 with ubiquitinated RIPK1. Finally, we showed that R406/R788 could promote the cancer-killing activities of TRAIL in vitro and in mouse models. Our studies demonstrate the possibility of developing small molecule TAK1 activators to potentiate the effect of TRAIL as anticancer therapies.


Subject(s)
Apoptosis , Neoplasms , Animals , Mice , Cell Death , Cytosol , Neoplasms/drug therapy , Neoplasms/genetics , Ubiquitination
2.
Proc Natl Acad Sci U S A ; 115(9): E2001-E2009, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440439

ABSTRACT

RIPK1 is a critical mediator of cell death and inflammation downstream of TNFR1 upon stimulation by TNFα, a potent proinflammatory cytokine involved in a multitude of human inflammatory and degenerative diseases. RIPK1 contains an N-terminal kinase domain, an intermediate domain, and a C-terminal death domain (DD). The kinase activity of RIPK1 promotes cell death and inflammation. Here, we investigated the involvement of RIPK1-DD in the regulation of RIPK1 kinase activity. We show that a charge-conserved mutation of a lysine located on the surface of DD (K599R in human RIPK1 or K584R in murine RIPK1) blocks RIPK1 activation in necroptosis and RIPK1-dependent apoptosis and the formation of complex II. Ripk1K584R/K584R knockin mutant cells are resistant to RIPK1 kinase-dependent apoptosis and necroptosis. The resistance of K584R cells, however, can be overcome by forced dimerization of RIPK1. Finally, we show that the K584R RIPK1 knockin mutation protects mice against TNFα-induced systematic inflammatory response syndrome. Our study demonstrates the role of RIPK1-DD in mediating RIPK1 dimerization and activation of its kinase activity during necroptosis and RIPK1-dependent apoptosis.


Subject(s)
Apoptosis , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Tumor Necrosis Factor-alpha/chemistry , Amino Acid Motifs , Animals , Cell Survival , Enzyme Activation , Exons , Genetic Complementation Test , HEK293 Cells , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mutation , Necrosis/genetics , Phosphorylation , Protein Binding , Protein Domains , Protein Multimerization , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
3.
BMC Cancer ; 19(1): 1044, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31690270

ABSTRACT

BACKGROUND: The receptor tyrosine kinases (RTKs) play critical roles in the development of cancers. Clear cell renal cell carcinoma (ccRCC) accounts for 75% of the RCC. The previous studies on the RTKs in ccRCCs mainly focused on their gene expressions. The activation and function of the RTKs in ccRCC have not been fully investigated. METHODS: In the present study, we analyzed the phosphorylation patterns of RTKs in human ccRCC patient samples, human ccRCC and papillary RCC cell lines, and other kidney tumor samples using human phospho-RTK arrays. We further established ccRCC patient-derived xenograft models in nude mice and assessed the effects of RTKIs (RTK Inhibitors) on the growth of these cancer cells. Immunofluorescence staining was used to detect the localization of keratin, vimentin and PDGFRß in ccRCCs. RESULTS: We found that the RTK phosphorylation patterns of the ccRCC samples were all very similar, but different from that of the cell lines, other kidney tumor samples, as well as the adjacent normal tissues. 9 RTKs, EGFR1-3, Insulin R, PDGFRß, VEGFR1, VEGFR2, HGFR and M-CSFR were found to be phosphorylated in the ccRCC samples. The adjacent normal tissues, on the other hand, had predominantly only two of the 4 EGFR family members, EGFR and ErbB4, phosphorylated. What's more, the RTK phosphorylation pattern of the xenograft, however, was different from that of the primary tissue samples. Treatment of the xenograft nude mice with corresponding RTK inhibitors effectively inhibited the Erk1/2 signaling pathway as well as the growth of the tumors. In addition, histological staining of the cancer samples revealed that most of the PDGFRß expressing cells were localized in the vimentin-positive periepithelial stroma. CONCLUSIONS: Overall, we have identified a set of RTKs that are characteristically phosphorylated in ccRCCs. The phosphorylation of RTKs in ccRCCs were determined by the growing environments. These phosphorylated/activated RTKs will guide targeting drugs development of more effective therapies in ccRCCs. The synergistical inhibition of RTKIs combination on the ccRCC suggest a novel strategy to use a combination of RTKIs to treat ccRCCs.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Kidney/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Female , Heterografts , Humans , Kidney/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Neoplasm Transplantation , Phosphorylation/immunology
4.
Cancer Sci ; 105(4): 473-80, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24450414

ABSTRACT

Constitutive activation of the signal transducer and activator of transcription 3 (STAT3) or the nuclear factor-κB (NF-κB) pathway occurs frequently in cancer cells and contributes to oncogenesis. The activation of Janus kinase 2 (JAK2) and IκB kinase (IKK) are key events in STAT3 and NF-κB signaling, respectively. We have identified 2-methoxystypandrone (2-MS) from a traditional Chinese medicinal herb Polygonum cuspidatum as a novel dual inhibitor of JAK2 and IKK. 2-MS inhibits both interleukin-6-induced and constitutively-activated STAT3, as well as tumor necrosis factor-α-induced NF-κB activation. 2-MS specifically inhibits JAK and IKKß kinase activities but has little effect on activities of other kinases tested. The inhibitory effects of 2-MS on STAT3 and NF-κB signaling can be eliminated by DTT or glutathione and can last for 4 h after a pulse treatment. Furthermore, 2-MS inhibits growth and induces death of tumor cells, particularly those with constitutively-activated STAT3 or NF-κB signaling. We propose that the natural compound 2-MS, as a potent dual inhibitor of STAT3 and NF-κB pathways, is a promising anticancer drug candidate.


Subject(s)
I-kappa B Kinase/biosynthesis , Janus Kinase 2/biosynthesis , NF-kappa B/genetics , Naphthoquinones/administration & dosage , STAT3 Transcription Factor/biosynthesis , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Interleukin-6/biosynthesis , Janus Kinase 2/genetics , Medicine, Chinese Traditional , Phosphorylation/drug effects , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
5.
Acta Pharm Sin B ; 14(1): 319-334, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38261830

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a key regulator in inflammation and cell death and is involved in mediating a variety of inflammatory or degenerative diseases. A number of allosteric RIPK1 inhibitors (RIPK1i) have been developed, and some of them have already advanced into clinical evaluation. Recently, selective RIPK1i that interact with both the allosteric pocket and the ATP-binding site of RIPK1 have started to emerge. Here, we report the rational development of a new series of type-II RIPK1i based on the rediscovery of a reported but mechanistically atypical RIPK3i. We also describe the structure-guided lead optimization of a potent, selective, and orally bioavailable RIPK1i, 62, which exhibits extraordinary efficacies in mouse models of acute or chronic inflammatory diseases. Collectively, 62 provides a useful tool for evaluating RIPK1 in animal disease models and a promising lead for further drug development.

6.
Sci Adv ; 9(47): eadj0123, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37992178

ABSTRACT

Transcriptional dysregulation is a recurring pathogenic hallmark and an emerging therapeutic vulnerability in ovarian cancer. Here, we demonstrated that ovarian cancer exhibited a unique dependency on the regulatory machinery of transcriptional termination, particularly, cleavage and polyadenylation specificity factor (CPSF) complex. Genetic abrogation of multiple CPSF subunits substantially hampered neoplastic cell viability, and we presented evidence that their indispensable roles converged on the endonuclease CPSF3. Mechanistically, CPSF perturbation resulted in lengthened 3'-untranslated regions, diminished intronic polyadenylation and widespread transcriptional readthrough, and consequently suppressed oncogenic pathways. Furthermore, we reported the development of specific CPSF3 inhibitors building upon the benzoxaborole scaffold, which exerted potent antitumor activity. Notably, CPSF3 blockade effectively exacerbated genomic instability by down-regulating DNA damage repair genes and thus acted in synergy with poly(adenosine 5'-diphosphate-ribose) polymerase inhibition. These findings establish CPSF3-dependent transcriptional termination as an exploitable driving mechanism of ovarian cancer and provide a promising class of boron-containing compounds for targeting transcription-addicted human malignancies.


Subject(s)
Neoplasm Recurrence, Local , Ovarian Neoplasms , Female , Humans , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
7.
J Med Chem ; 65(20): 13645-13659, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36221183

ABSTRACT

Deubiquitinating enzymes (DUBs) are key regulatory components of the ubiquitination system. Many DUBs have been revealed to play key roles in normal physiology and diseases. However, only very limited DUB members have well-characterized inhibitors. OTUB1 and USP8 are two DUBs reported to promote both immune evasion and tumorigenesis in tumor models, yet their targeted inhibitors are in the early stages of development. Here, we describe the lead identification and optimization of an OTUB1/USP8 dual inhibitor, 61, which exhibits highly potent and selective inhibition of both targets with subnanomolar IC50s in vitro. By inhibiting both DUBs, 61 phenocopies the double knockdown of OTUB1/USP8 and exerts pronounced antiproliferative effects in H1975 and other non-small-cell lung cancer (NSCLC) cell lines. Moreover, 61 efficaciously mitigates tumor growth in vivo. Collectively, our results provide a useful tool for pharmacological perturbation of OTUB1/USP8 and introduce a promising therapeutic strategy of dual DUB inhibition for treating NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Proteostasis , Lung Neoplasms/drug therapy , Ubiquitination , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism
8.
Oncogene ; 41(12): 1767-1779, 2022 03.
Article in English | MEDLINE | ID: mdl-35124696

ABSTRACT

Müllerian tissue-specific oncogenes, prototyped by PAX8, underlie ovarian tumorigenesis and represent unique molecular vulnerabilities. Further delineating such lineage-dependency factors and associated therapeutic implications would provide valuable insights into ovarian cancer biology and treatment. In this study, we identified SOX17 as a new lineage-survival master transcription factor, which shared co-expression pattern with PAX8 in epithelial ovarian carcinoma. Genetic disruption of SOX17 or PAX8 analogously inhibited neoplastic cell viability and downregulated a spectrum of lineage-related transcripts. Mechanistically, we showed that SOX17 physically interacted with PAX8 in cultured cell lines and clinical tumor specimens. The two nuclear proteins bound to overlapping genomic regions and regulated a common set of downstream genes, including those involved in cell cycle and tissue morphogenesis. In addition, we revealed that small-molecule inhibitors of transcriptional cyclin-dependent kinases (CDKs) effectively reduced SOX17 and PAX8 expression. ZSQ1722, a novel orally bioavailable CDK12/13 covalent antagonist, exerted potent anti-tumor activity in xenograft models. These findings shed light on an actionable lineage-survival transcriptional complex in ovarian cancer, and facilitated drug discovery by generating a serial of candidate compounds to pharmacologically target this difficult-to-treat malignancy.


Subject(s)
Ovarian Neoplasms , PAX8 Transcription Factor , SOXF Transcription Factors , Cell Cycle , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
9.
Cell Res ; 31(12): 1230-1243, 2021 12.
Article in English | MEDLINE | ID: mdl-34663909

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the ongoing global pandemic that poses substantial challenges to public health worldwide. A subset of COVID-19 patients experience systemic inflammatory response, known as cytokine storm, which may lead to death. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is an important mediator of inflammation and cell death. Here, we examined the interaction of RIPK1-mediated innate immunity with SARS-CoV-2 infection. We found evidence of RIPK1 activation in human COVID-19 lung pathological samples, and cultured human lung organoids and ACE2 transgenic mice infected by SARS-CoV-2. Inhibition of RIPK1 using multiple small-molecule inhibitors reduced the viral load of SARS-CoV-2 in human lung organoids. Furthermore, therapeutic dosing of the RIPK1 inhibitor Nec-1s reduced mortality and lung viral load, and blocked the CNS manifestation of SARS-CoV-2 in ACE2 transgenic mice. Mechanistically, we found that the RNA-dependent RNA polymerase of SARS-CoV-2, NSP12, a highly conserved central component of coronaviral replication and transcription machinery, promoted the activation of RIPK1. Furthermore, NSP12 323L variant, encoded by the SARS-CoV-2 C14408T variant first detected in Lombardy, Italy, that carries a Pro323Leu amino acid substitution in NSP12, showed increased ability to activate RIPK1. Inhibition of RIPK1 downregulated the transcriptional induction of proinflammatory cytokines and host factors including ACE2 and EGFR that promote viral entry into cells. Our results suggest that SARS-CoV-2 may have an unexpected and unusual ability to hijack the RIPK1-mediated host defense response to promote its own propagation and that inhibition of RIPK1 may provide a therapeutic option for the treatment of COVID-19.


Subject(s)
COVID-19/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/mortality , COVID-19/virology , Coronavirus RNA-Dependent RNA Polymerase/genetics , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/drug effects , ErbB Receptors/metabolism , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Lung/pathology , Lung/virology , Mice , Mice, Transgenic , Mutation , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , SARS-CoV-2/isolation & purification , SARS-CoV-2/metabolism , Survival Rate , Transcriptome/drug effects , Viral Load/drug effects , Virus Internalization , COVID-19 Drug Treatment
10.
Eur J Pharmacol ; 580(1-2): 161-8, 2008 Feb 02.
Article in English | MEDLINE | ID: mdl-18053981

ABSTRACT

Psychomotor effects elicited by systemic administration of the noncompetitive NMDA (N-methyl-D-aspartate) receptor antagonist MK-801 (dizocilpine maleate) represent perturbation of glutamatergic pathways, providing an animal model for psychotic symptoms of schizophrenia. Hyperlocomotion and stereotypy are the two main psychomotor behaviors induced by MK-801. This study compared MK-801-induced hyperlocomotion and stereotypy in young (1-month old) and aged mice (12-month old), in order to determine how the aging process may influence these behaviors. The tested MK-801 doses ranged from 0.015 to 1 mg/kg. The data indicated that MK-801 impacted the aged mice more pronouncedly than the young mice, as both hyperlocomotion and stereotypy were increased significantly more in the aged mice relative to the young mice. These results suggest an age-related increase in MK-801 sensitivity in mice.


Subject(s)
Disease Models, Animal , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Age Factors , Aging/physiology , Animals , Dizocilpine Maleate/administration & dosage , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Schizophrenia/physiopathology , Stereotyped Behavior/drug effects
11.
Cell Rep ; 10(11): 1836-49, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25801023

ABSTRACT

Multiple sclerosis (MS), a common neurodegenerative disease of the CNS, is characterized by the loss of oligodendrocytes and demyelination. Tumor necrosis factor α (TNF-α), a proinflammatory cytokine implicated in MS, can activate necroptosis, a necrotic cell death pathway regulated by RIPK1 and RIPK3 under caspase-8-deficient conditions. Here, we demonstrate defective caspase-8 activation, as well as activation of RIPK1, RIPK3, and MLKL, the hallmark mediators of necroptosis, in the cortical lesions of human MS pathological samples. Furthermore, we show that MS pathological samples are characterized by an increased insoluble proteome in common with other neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Finally, we show that necroptosis mediates oligodendrocyte degeneration induced by TNF-α and that inhibition of RIPK1 protects against oligodendrocyte cell death in two animal models of MS and in culture. Our findings demonstrate that necroptosis is involved in MS and suggest that targeting RIPK1 may represent a therapeutic strategy for MS.


Subject(s)
Apoptosis , Multiple Sclerosis/metabolism , Animals , Caspase 8/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Necrosis , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Protein Kinases/genetics , Proteome/genetics , Proteome/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/toxicity
12.
Elife ; 4: e06734, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25821988

ABSTRACT

Autophagy is an important intracellular catabolic mechanism involved in the removal of misfolded proteins. Atg14L, the mammalian ortholog of Atg14 in yeast and a critical regulator of autophagy, mediates the production PtdIns3P to initiate the formation of autophagosomes. However, it is not clear how Atg14L is regulated. In this study, we demonstrate that ubiquitination and degradation of Atg14L is controlled by ZBTB16-Cullin3-Roc1 E3 ubiquitin ligase complex. Furthermore, we show that a wide range of G-protein-coupled receptor (GPCR) ligands and agonists regulate the levels of Atg14L through ZBTB16. In addition, we show that the activation of autophagy by pharmacological inhibition of GPCR reduces the accumulation of misfolded proteins and protects against behavior dysfunction in a mouse model of Huntington's disease. Our study demonstrates a common molecular mechanism by which the activation of GPCRs leads to the suppression of autophagy and a pharmacological strategy to activate autophagy in the CNS for the treatment of neurodegenerative diseases.


Subject(s)
Heterocyclic Compounds/pharmacology , Huntington Disease/drug therapy , Huntington Disease/genetics , Kruppel-Like Transcription Factors/genetics , Receptors, CXCR4/genetics , Vesicular Transport Proteins/genetics , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Proteins , Benzylamines , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chromones/pharmacology , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclams , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Huntington Disease/mortality , Huntington Disease/pathology , Kruppel-Like Transcription Factors/metabolism , Mice , Morpholines/pharmacology , Phagosomes , Phosphatidylinositol Phosphates/biosynthesis , Promyelocytic Leukemia Zinc Finger Protein , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Psychomotor Performance/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Rotarod Performance Test , Signal Transduction , Survival Analysis , Ubiquitination , Vesicular Transport Proteins/metabolism
13.
Chin J Nat Med ; 12(8): 607-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25156286

ABSTRACT

AIM: To investigate the active constituents of Lignum Sappan (Caesalpinia sappan L.) on growth-related signaling and cell mitosis. METHOD: The influence of the ethyl acetate (EtOAc) extract of Lignum Sappan and its constituents on growth-related signaling were evaluated by a luciferase assay in cells stably-transfected with NF-κB, STAT1, or STAT3 responsive luciferase reporter plasmid. The inhibitory effect on the cell cycle was determined by flow cytometric analysis. The anti-tumor activities were assessed in vitro and in vivo. RESULTS: The EtOAc extract of Lignum Sappan had inhibitory activities on growth-related signaling and cell mitosis. Three major active compounds were sappanchalcone, brazilin, and butein. Sappanchalcone blocked cell cycle progression in the G2/M phase, brazilin inhibited TNFα/NF-κB signaling, while butein inhibited IL-6/STAT3 signaling, as well as TNFα/NF-κB signaling. The three compounds all demonstrated cytotoxic activities against human tumor cells in vitro. In a S180 tumor cell-bearing mice model, the anti-tumor efficacy of the EtOAc extract was better than the individual compounds acting alone. CONCLUSION: These results indicate that Lignum Sappan contains multiple active compounds with different antitumor activities, which act synergistically to enhance their anti-tumor effects. The EtOAc extract of Lignum Sappan may be better than individual active constituent as a novel medicine for the treatment of cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzopyrans/pharmacology , Caesalpinia , Cell Cycle Checkpoints/drug effects , Chalcones/pharmacology , Mitosis/drug effects , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Benzopyrans/therapeutic use , Chalcones/therapeutic use , Hep G2 Cells , Humans , Interleukin-6/metabolism , Male , Mice, Inbred BALB C , NF-kappa B/metabolism , Phytotherapy , Plant Extracts/therapeutic use , STAT3 Transcription Factor/metabolism , Sarcoma/drug therapy , Sarcoma/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
14.
Biochem Pharmacol ; 84(4): 468-76, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22687625

ABSTRACT

Selectively eradicating cancer cells with minimum adverse effects on normal cells is a major challenge in the development of anticancer therapy. We hypothesize that nutrient-limiting conditions frequently encountered by cancer cells in poorly vascularized solid tumors might provide an opportunity for developing selective therapy. In this study, we investigated the function and molecular mechanisms of a natural compound, arctigenin, in regulating tumor cell growth. We demonstrated that arctigenin selectively promoted glucose-starved A549 tumor cells to undergo necrosis by inhibiting mitochondrial respiration. In doing so, arctigenin elevated cellular level of reactive oxygen species (ROS) and blocked cellular energy metabolism in the glucose-starved tumor cells. We also demonstrated that cellular ROS generation was caused by intracellular ATP depletion and played an essential role in the arctigenin-induced tumor cell death under the glucose-limiting condition. Furthermore, we combined arctigenin with the glucose analogue 2-deoxyglucose (2DG) and examined their effects on tumor cell growth. Interestingly, this combination displayed preferential cell-death inducing activity against tumor cells compared to normal cells. Hence, we propose that the combination of arctigenin and 2DG may represent a promising new cancer therapy with minimal normal tissue toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Furans/pharmacology , Glucose/metabolism , Lignans/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/isolation & purification , Arctium/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Energy Metabolism/drug effects , Furans/isolation & purification , Glucose/deficiency , Humans , Lignans/isolation & purification , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis , Reactive Oxygen Species/metabolism , Tumor Microenvironment
15.
Acta Pharmacol Sin ; 27(11): 1401-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17049114

ABSTRACT

AIM: To examine the effect of GNTI [5'-guanidinyl-17-(cyclopropylmethyl)-6,7- dehydro-4,5alpha-epoxy-3,14-dihydroxy-6,7-2',3'-indolomorphinan], a selective antagonist for the kappa opioid receptor, in the MK-801 (dizocilpine maleate)-induced behavioral model of psychosis in schizophrenia as a way to explore the involvement of the kappa opioid receptor in modulating psychotic symptoms of schizophrenia. METHODS: Two doses of MK-801 (0.3 mg/kg and 0.6 mg/kg) were administered by systemic injection in mice to induce psychosis-like behavior as a rodent schizophrenia model, preceded by an injection of different doses of GNTI. Both locomotion and stereotypy were measured as the behavioral endpoints for quantitative analysis. RESULTS: GNTI inhibited MK-801-induced hyperlocomotion and stereotypy. In particular, GNTI showed differential modulation of stereotypy induced by 0.3 mg/kg vs 0.6 mg/kg MK-801. CONCLUSION: Antagonism of kappa opioid receptors attenuates MK-801-induced behavior, suggesting a potential involvement of the kappa opioid receptor in psychosis-like symptoms of schizophrenia. GNTI appears to be a useful pharmacological tool to explore the kappa opioid receptor function in vivo.


Subject(s)
Dizocilpine Maleate/antagonists & inhibitors , Guanidines/pharmacology , Locomotion/drug effects , Morphinans/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Stereotyped Behavior/drug effects , Animals , Male , Mice , Mice, Inbred BALB C , Schizophrenia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL