1
|
Aksu AM, Akter A, Dhillon P, Zerbel ZJ, Bridge-Comer PE, Gbayisomore O, Reilly SM. JNK mediates serine phosphorylation of STAT3 in response to fatty acids released by lipolysis. RESEARCH SQUARE 2025:rs.3.rs-6150649. [PMID: 40092442 PMCID: PMC11908360 DOI: 10.21203/rs.3.rs-6150649/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Adipocytes play an essential role in energy balance and metabolic health. Excess nutrients are stored within the white adipose tissue (WAT) as triglycerides. Energetic demand is communicated to the adipocyte by the sympathetic nervous system. Catecholamines released by nerve terminals in the adipose tissue promote lipolysis, a process in which triglycerides are broken down into fatty acids and glycerol. Lipolytic activation of white adipocytes is associated with an increase in the rate of oxygen consumption. This lipolysis induced respiration requires phosphorylation of signal transducer and activator of transcription 3 (STAT3) at Ser727. This study identifies c-Jun N-terminal kinase 1 (JNK1) as the kinase responsible for this critical phosphorylation event, and thus a key regulator of lipolysis-driven oxidative metabolism. We demonstrate that JNK1 is activated in response to intracellular fatty acids released during lipolysis and phosphorylates lipid droplet-associated STAT3, leading to inhibition of glycerol-3-phosphate acyltransferase 3 (GPAT3) and suppression of fatty acid re-esterification. This mechanism promotes uncoupled mitochondrial respiration, increasing energy expenditure. Inhibition of JNK1 attenuated oxidative metabolism without affecting the rate of lipolysis. The MAP kinase cascade upstream of JNK1 in lipolytic adipocytes remains unclear. Neither apoptosis signal-regulating kinase 1 (ASK1) nor mitogen-activated protein kinase kinases 4/7 (MKK4/7) appear to be required. Our findings suggest that JNK1 functions as a metabolic sensor in adipocytes, activating oxidative metabolism through STAT3 phosphorylation in response to fatty acids, with implications for energy balance and obesity-related metabolic regulation.
Collapse
|
2
|
Yue P, Chen Y, Ogese MO, Sun S, Zhang X, Esan T, Buolamwini JK, Turkson J. Small Molecule Induces Time-Dependent Inhibition of Stat3 Dimerization and DNA-Binding Activity and Regresses Human Breast Tumor Xenografts. Chembiochem 2024; 25:e202400351. [PMID: 39168826 DOI: 10.1002/cbic.202400351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/26/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
Aberrantly-active signal transducer and activator of transcription (Stat)3 has a causal role in many human cancers and represents a validated anticancer drug target, though it has posed significant challenge to drug development. A new small molecule, JKB887, was identified through library screening and is predicted to interact with Lys591, Arg609 and Pro63 in the phospho-tyrosine (pTyr)-binding pocket of the Stat3 SH2 domain. JKB887 inhibited Stat3 DNA-binding activity in vitro in a time-dependent manner, with IC50 of 2.2-4.5 μM at 30-60-min incubation. It directly disrupted both the Stat3 binding to the cognate, high-affinity pTyr (pY) peptide, GpYLPQTV-NH2 in fluorescent polarization assay with IC50 of 3.5-5.5 μM at 60-90-min incubation, and to the IL-6 receptor/gp130 or Src in treated malignant cells. Treatment with JKB887 selectively blocked constitutive Stat3 phosphorylation, nuclear translocation and transcriptional activity, and Stat3-regulated gene expression, and decreased viable cell numbers, cell growth, colony formation, migration, and survival in human or mouse tumor cells. By contrast, JKB887 had minimal effects on Stat1, pErk1/2MAPK, pShc, pJAK2, or pSrc induction, or on cells that do not harbor aberrantly-active Stat3. Additionally, JKB887 inhibited growth of human breast cancer xenografts in mice. JKB887 is a Stat3-selective inhibitor with demonstrable antitumor effects against Stat3-dependent human cancers.
Collapse
Affiliation(s)
- Peibin Yue
- Department of Medicine, Division of Hematology-Oncology, Cedars Sinai Medical Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, CA, 90048, USA
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Yue Chen
- Department of Medicine, Division of Hematology-Oncology, Cedars Sinai Medical Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, CA, 90048, USA
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
- Current adress: Department of Basic Medicine, Suzhou Vocational Health College, Suzhou, 215009, China
| | - Monday O Ogese
- Department of Medicine, Division of Hematology-Oncology, Cedars Sinai Medical Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, CA, 90048, USA
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Shan Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 847 Monroe Avenue, Suite 327, Memphis, TN, 38163, USA
| | - Xiaolei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical, Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Taiwo Esan
- Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064-3095, USA
| | - John K Buolamwini
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 847 Monroe Avenue, Suite 327, Memphis, TN, 38163, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064-3095, USA
| | - James Turkson
- Department of Medicine, Division of Hematology-Oncology, Cedars Sinai Medical Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, CA, 90048, USA
- Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| |
Collapse
|
3
|
Bukowska P, Bralewska M, Pietrucha T, Sakowicz A. Nutraceuticals as Modulators of Molecular Placental Pathways: Their Potential to Prevent and Support the Treatment of Preeclampsia. Int J Mol Sci 2024; 25:12167. [PMID: 39596234 PMCID: PMC11594370 DOI: 10.3390/ijms252212167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia (PE) is a serious condition characterized by new-onset hypertension and proteinuria or organ dysfunction after the 20th week of gestation, making it a leading cause of maternal and fetal mortality worldwide. Despite extensive research, significant gaps remain in understanding the mechanisms underlying PE, contributing to the ineffectiveness of current prevention and treatment strategies. Consequently, premature cesarean sections often become the primary intervention to safeguard maternal and fetal health. Emerging evidence indicates that placental insufficiency, driven by molecular disturbances, plays a central role in the development of PE. Additionally, the maternal microbiome may be implicated in the pathomechanism of preeclampsia by secreting metabolites that influence maternal inflammation and oxidative stress, thereby affecting placental health. Given the limitations of pharmaceuticals during pregnancy due to potential risks to fetal development and concerns about teratogenic effects, nutraceuticals may provide safer alternatives. Nutraceuticals are food products or dietary supplements that offer health benefits beyond basic nutrition, including plant extracts or probiotics. Their historical use in traditional medicine has provided valuable insights into their safety and efficacy, including for pregnant women. This review will examine how the adoption of nutraceuticals can enhance dysregulated placental pathways, potentially offering benefits in the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
| | | | | | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
4
|
Naik A, Thomas R, Sikhondze M, Babiker A, Lattab B, Qasem H, Jafar U, Decock J. The LDHC-STAT3 Signaling Network Is a Key Regulator of Basal-like Breast Cancer Cell Survival. Cancers (Basel) 2024; 16:2451. [PMID: 39001513 PMCID: PMC11240808 DOI: 10.3390/cancers16132451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer treatment has evolved drastically with the addition of immunotherapy and novel targeted drugs to the current treatment options. However, achieving long-term responses with minimal adverse events remains challenging. Cancer testis antigens (CTAs) offer novel opportunities for drug development thanks to their tumor specificity, immunogenicity, pro-tumorigenic functions, and negative prognostic connotations. We previously reported that lactate dehydrogenase C (LDHC) plays a key role in regulating genomic stability and that targeting LDHC significantly improved treatment response to DNA damage response drugs in breast cancer. Here, we explored the molecular mechanisms associated with LDHC silencing in two basal-like breast cancer cell lines, MDA-MB-468 and BT-549, and a Her2-enriched breast cancer cell line, HCC-1954. Transcriptomic analyses identified the cell line-dependent differential activation of the pro-survival STAT3 pathway following LDHC depletion. While LDHC silencing significantly compromised cell survival in basal-like breast cancer cells in conjunction with a downregulation of STAT3 signaling, the opposite effect was observed in Her2-enriched breast cancer cells, which demonstrated the enhanced activation of the pro-survival STAT3 signaling pathway. The inhibition of STAT3 not only reversed the unfavorable effect of LDHC silencing in the Her2-enriched cancer cells but also demonstrated significant anti-cancer activity when used as a single agent. Our findings suggest that the LDHC-STAT3 signaling axis plays a role in regulating breast tumor cell survival in a subtype-dependent manner. Thus, LDHC-targeted therapy could be a viable therapeutic approach for a subset of breast cancer patients, particularly patients with basal-like breast cancer, whereas patients carrying Her2-enriched tumors may likely benefit more from monotherapy with STAT3 inhibitors.
Collapse
Affiliation(s)
- Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
| | - Remy Thomas
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
| | - Martin Sikhondze
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
| | - Abeer Babiker
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
| | - Boucif Lattab
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
| | - Hanan Qasem
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Umar Jafar
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar (A.B.); haqa24936@ hbku.edu.qa (H.Q.); (U.J.)
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| |
Collapse
|
5
|
Chang YC, Yu MH, Huang HP, Chen DH, Yang MY, Wang CJ. Mulberry leaf extract inhibits obesity and protects against diethylnitrosamine-induced hepatocellular carcinoma in rats. J Tradit Complement Med 2024; 14:266-275. [PMID: 38707917 PMCID: PMC11068992 DOI: 10.1016/j.jtcme.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 05/07/2024] Open
Abstract
Mulberry leaf has been recognized as a traditional Chinese medicinal plant, which was distributed throughout the Asia. The aqueous extract of mulberry leaf extract (MLE) has various biologically active components such as polyphenols and flavonoids. However, the inhibitory effect of MLE in hepatocarcinogenesis is poorly understood. In this study, we determined the role of MLE supplementation in preventing hepatocarcinogenesis in a carcinogen-initiated high-fat diet (HFD)-promoted Sprague-Dawley (SD) rat model. The rats were fed an HFD to induce obesity and spontaneous hepatomas by administering 0.01% diethylnitrosamine (DEN) in their drinking water for 12 weeks (HD group), and also to fed MLE through oral ingestion at daily doses of 0.5%, 1%, or 2%. At the end of the 12-week experimental period, the liver tumors were analyzed to identify markers of oxidative stress and antioxidant enzyme activities, and their serum was analyzed to determine their nutritional status and liver function. Histopathological analysis revealed that MLE supplementation significantly suppressed the severity and incidence of hepatic tumors. Furthermore, compared with the HFD + DEN groups, the expression of protein kinase C (PKC)-α and Rac family small GTPase 1 (Rac1) was lower in the MLE groups. These findings suggest that MLE prevents obesity-enhanced, carcinogen-induced hepatocellular carcinoma development, potentially through the protein kinase C (PKC)α/Rac1 signaling pathway. MLE might be an effective chemoprevention modality for nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH)-related hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yun-Ching Chang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Meng-Hsun Yu
- Department of Nutrition, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Hui-Pei Huang
- Department of Biochemistry, School of Medicine, Medical College, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Dong-Hui Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Mon-Yuan Yang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Chau-Jong Wang
- Department of Health Diet and Industry Management, Chung Shan Medical University, Taichung, 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, 402, Taiwan
| |
Collapse
|
6
|
Sun Y, Xu C, Jiang Z, Jiang X. DEF6(differentially exprehomolog) exacerbates pathological cardiac hypertrophy via RAC1. Cell Death Dis 2023; 14:483. [PMID: 37524688 PMCID: PMC10390462 DOI: 10.1038/s41419-023-05948-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023]
Abstract
Pathological cardiac hypertrophy involves multiple regulators and several signal transduction pathways. Currently, the mechanisms of it are not well understood. Differentially expressed in FDCP 6 homolog (DEF6) was reported to participate in immunity, bone remodeling, and cancers. The effects of DEF6 on pathological cardiac hypertrophy, however, have not yet been fully characterized. We initially determined the expression profile of DEF6 and found that DEF6 was upregulated in hypertrophic hearts and cardiomyocytes. Our in vivo results revealed that DEF6 deficiency in mice alleviated transverse aortic constriction (TAC)-induced cardiac hypertrophy, fibrosis, dilation and dysfunction of left ventricle. Conversely, cardiomyocyte-specific DEF6-overexpression aggravated the hypertrophic phenotype in mice under chronic pressure overload. Similar to the animal experiments, the in vitro data showed that adenovirus-mediated knockdown of DEF6 remarkably inhibited phenylephrine (PE)-induced cardiomyocyte hypertrophy, whereas DEF6 overexpression exerted the opposite effects. Mechanistically, exploration of the signal pathways showed that the mitogen-activated extracellular signal-regulated kinase 1/2 (MEK1/2)-extracellular signal-regulated kinase 1/2 (ERK1/2) cascade might be involved in the prohypertrophic effect of DEF6. Coimmunoprecipitation and GST (glutathione S-transferase) pulldown analyses demonstrated that DEF6 can directly interact with small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), and the Rac1 activity assay revealed that the activity of Rac1 is altered with DEF6 expression in TAC-cardiac hypertrophy and PE-triggered cardiomyocyte hypertrophy. In the end, western blot and rescue experiments using Rac1 inhibitor NSC23766 and the constitutively active mutant Rac1(G12V) verified the requirement of Rac1 and MEK1/2-ERK1/2 activation for DEF6-mediated pathological cardiac hypertrophy. Our study substantiates that DEF6 acts as a deleterious regulator of cardiac hypertrophy by activating the Rac1 and MEK1/2-ERK1/2 signaling pathways, and suggests that DEF6 may be a potential treatment target for heart failure.
Collapse
Affiliation(s)
- Yan Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, 110022, Shenyang, Liaoning Province, China
| | - Changlu Xu
- Department of Cardiology, Shengjing Hospital of China Medical University, 110022, Shenyang, Liaoning Province, China
| | - Zhongxiu Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, 110022, Shenyang, Liaoning Province, China
| | - Xi Jiang
- Department of Cardiology, Shengjing Hospital of China Medical University, 110022, Shenyang, Liaoning Province, China.
| |
Collapse
|
7
|
Kriaučiūnaitė K, Pociūtė A, Kaušylė A, Verkhratsky A, Pivoriūnas A. Basic Fibroblast Growth Factor Opens and Closes the Endothelial Blood-Brain Barrier in a Concentration-Dependent Manner. Neurochem Res 2023; 48:1211-1221. [PMID: 35859077 DOI: 10.1007/s11064-022-03678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 06/23/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Multiple paracrine factors are implicated in the regulation of barrier properties of human brain endothelial cells (BECs) in different physiologic and pathologic settings. We have recently demonstrated that autocrine secretion of basic fibroblast growth factor (bFGF) by BECs is necessary for the establishment of endothelial barrier (as demonstrated by high trans-endothelial electric resistance, TEER), whereas exogenous bFGF inhibits TEER in a concentration-dependent manner. In the present study we analysed the contribution of MAPK/ERK and STAT3 signalling pathways to the inhibitory effects of exogenous bFGF. Treatment with bFGF (8 ng/ml) for 3 days increased phosphorylation of ERK1/2 and STAT3. Treatment with FGF receptor 1 (FGFR1) inhibitor PD173074 (15 μM) suppressed both basal and bFGF-induced activation of ERK1/2 and STAT3. Suppression of STAT signalling with Janus kinase inhibitor JAKi (15 nM) alone or in the presence of bFGF did not change TEER in BEC monolayers. Exposure to JAKi affected neither proliferation, nor expression and distribution of tight junction (TJ) proteins claudin-5, occludin and zonula occludens-1 (ZO-1). In contrast, treatment with MEK 1/2 inhibitor U0126 (10 μM) partially neutralised inhibitory effect of bFGF thus increasing TEER, whereas U0126 alone did not affect resistance of endothelial barrier. Our findings demonstrate that MAPK/ERK signalling pathway does not affect autocrine bFGF signalling-dependent BECs barrier function but is largely responsible for the disruptive effects of the exogenous bFGF. We speculate that bFGF may (depending on concentration and possibly origin) dynamically regulate permeability of the endothelial blood-brain barrier.
Collapse
Affiliation(s)
- Karolina Kriaučiūnaitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania
| | - Agnė Pociūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania
| | - Aida Kaušylė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania
| | - Alexei Verkhratsky
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- IKERBASQUE, Basque Foundation for Science, Achucarro Centre for Neuroscience, 48011, Bilbao, Spain.
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania.
| |
Collapse
|
8
|
Zhu Y, Yue P, Dickinson CF, Yang JK, Datanagan K, Zhai N, Zhang Y, Miklossy G, Lopez-Tapia F, Tius MA, Turkson J. Natural product preferentially targets redox and metabolic adaptations and aberrantly active STAT3 to inhibit breast tumor growth in vivo. Cell Death Dis 2022; 13:1022. [PMID: 36473850 PMCID: PMC9726930 DOI: 10.1038/s41419-022-05477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Dysregulated gene expression programs and redox and metabolic adaptations allow cancer cells to survive under high oxidative burden. These mechanisms also represent therapeutic vulnerabilities. Using triple-negative breast cancer (TNBC) as a model, we show that compared to normal human breast epithelial cells, the TNBC cells, MDA-MB-231 and MDA-MB-468 that harbor constitutively active STAT3 also express higher glucose-6-phosphate dehydrogenase (G6PD), thioredoxin reductase (TrxR)1, NADPH, and GSH levels for survival. Present studies discover that the natural product, R001, targets these adaptation mechanisms. Treatment of TNBC cells with R001 inhibited constitutively active STAT3, STAT3-regulated gene expression, and the functions of G6PD and TrxR1. Consequently, in the TNBC, but not normal cells, R001 suppressed GSH levels, but raised NADPH levels, reflective of a loss of mitochondrial respiration and which led to reactive oxygen species (ROS) induction, all of which led to loss of viable cells and inhibition of anchorage-dependent and independent growth. R001 treatment further led to early pyroptosis and late DNA damage, cell cycle arrest, and apoptosis only in the TNBC cells. Oral administration of 5 mg/kg R001 inhibited MDA-MB-468 xenografts growth in mice, with reduced pY705-STAT3, G6PD, TrxR1, and GSH levels. R001 serves as a therapeutic entity that targets the vulnerabilities of TNBC cells to inhibit tumor growth in vivo.
Collapse
Affiliation(s)
- Yinsong Zhu
- grid.50956.3f0000 0001 2152 9905Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA ,grid.50956.3f0000 0001 2152 9905Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Peibin Yue
- grid.50956.3f0000 0001 2152 9905Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA ,grid.50956.3f0000 0001 2152 9905Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Cody F. Dickinson
- grid.410445.00000 0001 2188 0957Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825 USA
| | - Justin K. Yang
- grid.410445.00000 0001 2188 0957Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825 USA
| | - Kyrstin Datanagan
- grid.410445.00000 0001 2188 0957Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825 USA
| | - Ning Zhai
- grid.50956.3f0000 0001 2152 9905Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA ,grid.50956.3f0000 0001 2152 9905Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Yi Zhang
- grid.50956.3f0000 0001 2152 9905Biobank and Research Pathology Resource, Academic Affairs and Research Administration, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Gabriella Miklossy
- grid.516097.c0000 0001 0311 6891Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813 USA
| | - Francisco Lopez-Tapia
- grid.50956.3f0000 0001 2152 9905Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA ,grid.50956.3f0000 0001 2152 9905Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Marcus A. Tius
- grid.410445.00000 0001 2188 0957Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825 USA ,grid.516097.c0000 0001 0311 6891Cancer Biology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813 USA
| | - James Turkson
- grid.50956.3f0000 0001 2152 9905Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA ,grid.50956.3f0000 0001 2152 9905Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| |
Collapse
|
9
|
Li Y, Lai W, Zheng C, Babu JR, Xue C, Ai Q, Huggins KW. Neuroprotective Effect of Stearidonic Acid on Amyloid β-Induced Neurotoxicity in Rat Hippocampal Cells. Antioxidants (Basel) 2022; 11:2357. [PMID: 36552565 PMCID: PMC9774633 DOI: 10.3390/antiox11122357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Dietary intake of omega-3 fatty acids found in fish has been reported to reduce the risk of Alzheimer's Disease (AD). Stearidonic acid (SDA), a plant-based omega-3 fatty acid, has been targeted as a potential surrogate for fish-based fatty acids. However, its role in neuronal degeneration is unknown. This study was designed to evaluate effects of SDA on Amyloid-β(A-β)-induced neurotoxicity in rat hippocampal cells. Results showed that SDA effectively converted to eicosapentaenoic acid (EPA) in hippocampal cells. Aβ-induced apoptosis in H19-7 cells was protected by SDA pretreatment as evidenced by its regulation on the expression of relevant pro- and anti-apoptotic genes, as well as the inhibition on caspase activation. SDA also protected H19-7 cells from Aβ-induced oxidative stress by regulating the expression of relevant pro- and anti-oxidative genes, as well as the improvement in activity of catalase. As for Aβ/LPS-induced neuronal inflammation, SDA pretreatment reduced the release of IL-1β and TNFα. Further, we found that the anti-Aβ effect of SDA involves its inhibition on the expression of amyloid precursor protein and the regulation on MAPK signaling. These results demonstrated that SDAs have neuroprotective effect in Aβ-induced H19-7 hippocampal cells. This beneficial effect of SDA was attributed to its antiapoptotic, antioxidant, and anti-inflammatory properties.
Collapse
Affiliation(s)
- Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, 5 Yushan Road, Qingdao 266005, China
- Department of Nutrition, Dietetics and Hospitality Management, Auburn University, Auburn, AL 36849, USA
- College of Food Science and Engineering, Ocean University of China, Qingdao 266005, China
| | - Wencong Lai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, 5 Yushan Road, Qingdao 266005, China
| | - Chen Zheng
- Department of Nutrition, Dietetics and Hospitality Management, Auburn University, Auburn, AL 36849, USA
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics and Hospitality Management, Auburn University, Auburn, AL 36849, USA
- Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL 36849, USA
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266005, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, 5 Yushan Road, Qingdao 266005, China
| | - Kevin W. Huggins
- Department of Nutrition, Dietetics and Hospitality Management, Auburn University, Auburn, AL 36849, USA
- Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
10
|
Roads to Stat3 Paved with Cadherins. Cells 2022; 11:cells11162537. [PMID: 36010614 PMCID: PMC9406956 DOI: 10.3390/cells11162537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The engagement of cadherins, cell-to-cell adhesion proteins, triggers a dramatic increase in the levels and activity of the Rac/Cdc42 GTPases, through the inhibition of proteasomal degradation. This leads to an increase in transcription and secretion of IL6 family cytokines, activation of their common receptor, gp130, in an autocrine manner and phosphorylation of the signal transducer and activator of transcription-3 (Stat3) on tyrosine-705 by the Jak kinases. Stat3 subsequently dimerizes, migrates to the nucleus and activates the transcription of genes involved in cell division and survival. The Src oncogene also increases Rac levels, leading to secretion of IL6 family cytokines and gp130 activation, which triggers a Stat3-ptyr705 increase. Interestingly, at the same time, Src downregulates cadherins in a quantitative manner, while cadherins are required to preserve gp130 levels for IL6 family signalling. Therefore, a fine balance between Src527F/Rac/IL6 and Src527F/cadherin/gp130 levels is in existence, which is required for Stat3 activation. This further demonstrates the important role of cadherins in the activation of Stat3, through preservation of gp130 function. Conversely, the absence of cadherin engagement correlates with low Stat3 activity: In sparsely growing cells, both gp130 and Stat3-ptyr705 levels are very low, despite the fact that cSrc is active in the FAK (focal adhesion kinase)/cSrc complex, which further indicates that the engagement of cadherins is important for Stat3 activation, not just their presence. Furthermore, the caveolin-1 protein downregulates Stat3 through binding and sequestration of cadherins to the scaffolding domain of caveolin-1. We hypothesize that the cadherins/Rac/gp130 axis may be a conserved pathway to Stat3 activation in a number of systems. This fact could have significant implications in Stat3 biology, as well as in drug testing and development.
Collapse
|
11
|
Abzianidze V, Moiseeva N, Suponina D, Zakharenkova S, Rogovskaya N, Laletina L, Holder AA, Krivorotov D, Bogachenkov A, Garabadzhiu A, Ukolov A, Kosorukov V. Natural Phaeosphaeride A Derivatives Overcome Drug Resistance of Tumor Cells and Modulate Signaling Pathways. Pharmaceuticals (Basel) 2022; 15:ph15040395. [PMID: 35455394 PMCID: PMC9030166 DOI: 10.3390/ph15040395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
In the present study, natural phaeosphaeride A (PPA) derivatives are synthesized. Anti-tumor studies are carried out on the PC3, K562, HCT-116, THP-1, MCF-7, A549, NCI-H929, Jurkat, and RPMI8226 tumor cell lines, and on the human embryonic kidney (HEK293) cell line. All the compounds synthesized turned out to have better efficacy than PPA towards the tumor cell lines listed. Among them, three compounds exhibited an ability to overcome the drug resistance of tumor cells associated with the overexpression of the P-glycoprotein by modulating the work of this transporter. Luminex xMAP technology was used to assess the effect of five synthesized compounds on the activation of intracellular kinase cascades in A431 cells. MILLIPLEX MAP Multi-Pathway Magnetic Bead 9-Plex was used, which allowed for the simultaneous detection of the following nine phosphorylated protein markers of the main intracellular signaling pathways: a universal transcription factor that controls the expression of immune-response genes, apoptosis and cell cycle NFκB (pS536); cAMP-dependent transcription factor (CREB (pS133); mitogen-activated kinase p38 (pT180/pY182); stress-activated protein kinase JNK (pT183/pY185); ribosomal SK; transcription factors STAT3 (pS727) and STAT5A/B (pY694/699); protein kinase B (Akt) (pS473); and kinase regulated by extracellular signals ERK1/2 (pT185/pY187). The effect of various concentrations of PPA derivatives on the cell culture was studied using xCelligence RTCA equipment. The compounds were found to modulate JNK, ERK1/2, and p38 signaling pathways. The set of activated kinase cascades suggests that oxidative stress is the main probable mechanism of the toxic action of PPA derivatives.
Collapse
Affiliation(s)
- Victoria Abzianidze
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
- Correspondence: (V.A.); (A.H.); Tel.: +7-981-249-0902 (V.A.); +1-757-683-7102 (A.H.)
| | - Natalia Moiseeva
- Laboratory of Tumor Cell Genetics, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.M.); (L.L.)
| | - Diana Suponina
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Sofya Zakharenkova
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Nadezhda Rogovskaya
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Lidia Laletina
- Laboratory of Tumor Cell Genetics, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.M.); (L.L.)
| | - Alvin A. Holder
- Department of Chemistry and Biochemistry, Old Dominion University, 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
- Correspondence: (V.A.); (A.H.); Tel.: +7-981-249-0902 (V.A.); +1-757-683-7102 (A.H.)
| | - Denis Krivorotov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Alexander Bogachenkov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Alexander Garabadzhiu
- Saint Petersburg State Technological Institute (Technical University), 190013 Saint Petersburg, Russia;
| | - Anton Ukolov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Federal Medical Biological Agency, p/o Kuz’molovsky, 188663 Saint Petersburg, Russia; (D.S.); (S.Z.); (N.R.); (D.K.); (A.B.); (A.U.)
| | - Vyacheslav Kosorukov
- Laboratory of Transgenic Drugs, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia;
| |
Collapse
|
12
|
Miller KJ, Asim M. Unravelling the Role of Kinases That Underpin Androgen Signalling in Prostate Cancer. Cells 2022; 11:cells11060952. [PMID: 35326402 PMCID: PMC8946764 DOI: 10.3390/cells11060952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The androgen receptor (AR) signalling pathway is the key driver in most prostate cancers (PCa), and is underpinned by several kinases both upstream and downstream of the AR. Many popular therapies for PCa that target the AR directly, however, have been circumvented by AR mutation, such as androgen receptor variants. Some upstream kinases promote AR signalling, including those which phosphorylate the AR and others that are AR-regulated, and androgen regulated kinase that can also form feed-forward activation circuits to promotes AR function. All of these kinases represent potentially druggable targets for PCa. There has generally been a divide in reviews reporting on pathways upstream of the AR and those reporting on AR-regulated genes despite the overlap that constitutes the promotion of AR signalling and PCa progression. In this review, we aim to elucidate which kinases—both upstream and AR-regulated—may be therapeutic targets and require future investigation and ongoing trials in developing kinase inhibitors for PCa.
Collapse
|
13
|
Aponte E, Lafitte M, Sirvent A, Simon V, Barbery M, Fourgous E, Boublik Y, Maffei M, Armand F, Hamelin R, Pannequin J, Fort P, Pons M, Roche S. Regulation of Src tumor activity by its N-terminal intrinsically disordered region. Oncogene 2022; 41:960-970. [PMID: 34999732 PMCID: PMC8837538 DOI: 10.1038/s41388-021-02092-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 11/09/2022]
Abstract
The membrane-anchored Src tyrosine kinase is involved in numerous pathways and its deregulation is involved in human cancer. Our knowledge on Src regulation relies on crystallography, which revealed intramolecular interactions to control active Src conformations. However, Src contains a N-terminal intrinsically disordered unique domain (UD) whose function remains unclear. Using NMR, we reported that UD forms an intramolecular fuzzy complex involving a conserved region with lipid-binding capacity named Unique Lipid-Binding Region (ULBR), which could modulate Src membrane anchoring. Here we show that the ULBR is essential for Src's oncogenic capacity. ULBR inactive mutations inhibited Src transforming activity in NIH3T3 cells and in human colon cancer cells. It also reduced Src-induced tumor development in nude mice. An intact ULBR was required for MAPK signaling without affecting Src kinase activity nor sub-cellular localization. Phospho-proteomic analyses revealed that, while not impacting on the global tyrosine phospho-proteome in colon cancer cells, this region modulates phosphorylation of specific membrane-localized tyrosine kinases needed for Src oncogenic signaling, including EPHA2 and Fyn. Collectively, this study reveals an important role of this intrinsically disordered region in malignant cell transformation and suggests a novel layer of Src regulation by this unique region via membrane substrate phosphorylation.
Collapse
Affiliation(s)
- Emilie Aponte
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Marie Lafitte
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Audrey Sirvent
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Valérie Simon
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Maud Barbery
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Elise Fourgous
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Yvan Boublik
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Mariano Maffei
- Biomolecular NMR laboratory, Department of Inorganic and Organic Chemistry, University of Barcelona, Baldiri Reixac 10-12, 08028, Barcelona, Spain
- Evvivax srl-Via di Castel Romano, 100 - 00128, Rome, Italy
| | - Florence Armand
- Proteomics Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Romain Hamelin
- Proteomics Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | | | - Philippe Fort
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France
| | - Miquel Pons
- Biomolecular NMR laboratory, Department of Inorganic and Organic Chemistry, University of Barcelona, Baldiri Reixac 10-12, 08028, Barcelona, Spain.
| | - Serge Roche
- CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France.
- Equipe labellisée Ligue Contre le Cancer, CRBM, CNRS, Univ. Montpellier, F-34000, Montpellier, France.
- IGF, CNRS, Univ. Montpellier, F-34000, Montpellier, France.
| |
Collapse
|
14
|
Molinaro C, Martoriati A, Lescuyer A, Fliniaux I, Tulasne D, Cailliau K. 3-phosphoinositide-dependent protein kinase 1 (PDK1) mediates crosstalk between Src and Akt pathways in MET receptor signaling. FEBS Lett 2021; 595:2655-2664. [PMID: 34551132 DOI: 10.1002/1873-3468.14195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 11/12/2022]
Abstract
The high-affinity tyrosine kinase receptor MET plays a pivotal role in several facets of cell regulation. Although its mitogenic effect is well documented, some aspects of connection patterns between signaling pathways involved in cell cycle progression remain to be deciphered. We have used a tractable heterologous expression system, the Xenopus oocyte, to detect connections between distinct MET signaling cascades involved in G2/M progression. Our results reveal that Src acts as an adapter via its SH2 domain to recruit 3-phosphoinositide-dependent protein kinase 1 (PDK1) to the MET signaling complex leading to Akt phosphorylation. These data define an original crosstalk between Src and Akt signaling pathways that contributes to MET-induced entry into the M phase, and deserves further investigation in pathologies harboring deregulation of this receptor.
Collapse
Affiliation(s)
- Caroline Molinaro
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Alain Martoriati
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Arlette Lescuyer
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Ingrid Fliniaux
- Inserm U1003-PHYCEL-Cellular Physiology, University of Lille, Lille, France
| | - David Tulasne
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, Lille, France
| | - Katia Cailliau
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
15
|
Dong J, Cheng XD, Zhang WD, Qin JJ. Recent Update on Development of Small-Molecule STAT3 Inhibitors for Cancer Therapy: From Phosphorylation Inhibition to Protein Degradation. J Med Chem 2021; 64:8884-8915. [PMID: 34170703 DOI: 10.1021/acs.jmedchem.1c00629] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor that regulates various biological processes, including proliferation, metastasis, angiogenesis, immune response, and chemoresistance. In normal cells, STAT3 is tightly regulated to maintain a transiently active state, while persistent STAT3 activation occurs frequently in cancers, associating with a poor prognosis and tumor progression. Targeting the STAT3 protein is a potentially promising therapeutic strategy for tumors. Although none of the STAT3 inhibitors has been marketed yet, a few of them have succeeded in entering clinical trials. This Review aims to systematically summarize the progress of the last 5 years in the discovery of directive STAT3 small-molecule inhibitors and degraders, focusing primarily on their structural features, design strategies, and bioactivities. We hope this Review will shed light on future drug design and inhibitor optimization to accelerate the discovery process of STAT3 inhibitors or degraders.
Collapse
Affiliation(s)
- Jinyun Dong
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Xiang-Dong Cheng
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Jiang-Jiang Qin
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
16
|
Bolanle IO, Riches-Suman K, Williamson R, Palmer TM. Emerging roles of protein O-GlcNAcylation in cardiovascular diseases: Insights and novel therapeutic targets. Pharmacol Res 2021; 165:105467. [PMID: 33515704 DOI: 10.1016/j.phrs.2021.105467] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. While the major focus of pharmacological and non-pharmacological interventions has been on targeting disease pathophysiology and limiting predisposing factors, our understanding of the cellular and molecular mechanisms underlying the pathogenesis of CVDs remains incomplete. One mechanism that has recently emerged is protein O-GlcNAcylation. This is a dynamic, site-specific reversible post-translational modification of serine and threonine residues on target proteins and is controlled by two enzymes: O-linked β-N-acetylglucosamine transferase (OGT) and O-linked β-N-acetylglucosaminidase (OGA). Protein O-GlcNAcylation alters the cellular functions of these target proteins which play vital roles in pathways that modulate vascular homeostasis and cardiac function. Through this review, we aim to give insights on the role of protein O-GlcNAcylation in cardiovascular diseases and identify potential therapeutic targets in this pathway for development of more effective medicines to improve patient outcomes.
Collapse
Key Words
- (R)-N-(Furan-2-ylmethyl)-2-(2-methoxyphenyl)-2-(2-oxo-1,2-dihydroquinoline-6-sulfonamido)-N-(thiophen-2-ylmethyl)acetamide [OSMI-1] (PubChem CID: 118634407)
- 2-(2-Amino-3-methoxyphenyl)-4H-chromen-4-one [PD98059] (PubChem CID: 4713)
- 5H-Pyrano[3,2-d]thiazole-6,7-diol, 2-(ethylamino)-3a,6,7,7a-tetrahydro-5-(hydroxymethyl)-(3aR,5R,6S,7R,7aR) [Thiamet-G] (PubChem CID: 1355663540)
- 6-Diazo-5-oxo-l-norleucine [DON] (PubChem CID: 9087)
- Alloxan (PubChem CID: 5781)
- Azaserine (PubChem CID: 460129)
- BADGP, Benzyl-2-acetamido-2-deoxy-α-d-galactopyranoside [BADGP] (PubChem CID: 561184)
- Cardiovascular disease
- Methoxybenzene-sulfonamide [KN-93] (PubChem CID: 5312122)
- N-[(5S,6R,7R,8R)-6,7-Dihydroxy-5-(hydroxymethyl)-2-(2-phenylethyl)-5,6,7,8-tetrahydroimidazo[1,2-a]pyridin-8-yl]-2-methylpropanamide [GlcNAcstatin] (PubChem CID: 122173013)
- O-(2-Acetamido-2-deoxy-d-glucopyranosyliden)amino-N-phenylcarbamate [PUGNAc] (PubChem CID: 9576811)
- O-GlcNAc transferase
- O-GlcNAcase
- Protein O-GlcNAcylation
- Streptozotocin (PubCHem CID: 7067772)
Collapse
Affiliation(s)
- Israel Olapeju Bolanle
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Kirsten Riches-Suman
- School of Chemistry and Bioscience, University of Bradford, Bradford BD7 1DP, UK
| | - Ritchie Williamson
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Timothy M Palmer
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK.
| |
Collapse
|
17
|
Hepatoprotective Potency of Chrysophanol 8- O-Glucoside from Rheum palmatum L. against Hepatic Fibrosis via Regulation of the STAT3 Signaling Pathway. Int J Mol Sci 2020; 21:ijms21239044. [PMID: 33261209 PMCID: PMC7730872 DOI: 10.3390/ijms21239044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Rhubarb is a well-known herb worldwide and includes approximately 60 species of the Rheum genus. One of the representative plants is Rheum palmatum, which is prescribed as official rhubarb due to its pharmacological potential in the Korean and Chinese pharmacopoeia. In our bioactive screening, we found out that the EtOH extract of R. palmatum inhibited hepatic stellate cell (HSC) activation by transforming growth factor β1 (TGF-β1). Chemical investigation of the EtOH extract led to the isolation of chrysophanol 8-O-glucoside, which was determined by structural analysis using NMR spectroscopic techniques and electrospray ionization mass spectrometry (ESIMS). To elucidate the effects of chrysophanol 8-O-glucoside on HSC activation, activated LX-2 cells were treated for 48 h with chrysophanol 8-O-glucoside, and α-SMA and collagen, HSC activation markers, were measured by comparative quantitative real-time PCR (qPCR) and western blotting analysis. Chrysophanol 8-O-glucoside significantly inhibited the protein and mRNA expression of α-SMA and collagen compared with that in TGF-β1-treated LX-2 cells. Next, the expression of phosphorylated SMAD2 (p-SMAD2) and p-STAT3 was measured and the translocation of p-STAT3 to the nucleus was analyzed by western blotting analysis. The expression of p-SMAD2 and p-STAT3 showed that chrysophanol 8-O-glucoside strongly downregulated STAT3 phosphorylation by inhibiting the nuclear translocation of p-STAT3, which is an important mechanism in HSC activation. Moreover, chrysophanol 8-O-glucoside suppressed the expression of p-p38, not that of p-JNK or p-Erk, which can activate STAT3 phosphorylation and inhibit MMP2 expression, the downstream target of STAT3 signaling. These findings provided experimental evidence concerning the hepatoprotective effects of chrysophanol 8-O-glucoside against liver damage and revealed the molecular basis underlying its anti-fibrotic effects through the blocking of HSC activation.
Collapse
|
18
|
Wang Y, Liu S, Jiang G, Zhai W, Yang L, Li M, Chang Z, Zhu B. NOK associates with c-Src and promotes c-Src-induced STAT3 activation and cell proliferation. Cell Signal 2020; 75:109762. [PMID: 32871210 DOI: 10.1016/j.cellsig.2020.109762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/24/2022]
Abstract
Signal transducers and activators of transcription 3 (STAT3) is reported to regulate cell proliferation, survival, and differentiation, and thus plays a central role in development and carcinogenesis. Accumulating evidence demonstrated the involvement of cellular Src (c-Src) tyrosine kinase in the activation of STAT3. Additionally, novel oncogene with kinase-domain (NOK), a receptor protein tyrosine kinase that involves in cell transformation and tumorigenesis, was found to activate STAT3 signaling by a JAK2-dependent mechanism. However, whether the existence of the interaction between c-Src/STAT3 and NOK/STAT3 signals is still unknown. In this study, we showed that NOK formed a complex with c-Src and facilitated the interaction between c-Src and STAT3. In the complex, NOK greatly elevated the c-Src-mediated STAT3 activation by increasing the phosphorylation level of STAT3 on Tyr705. Truncated and mutation experiments further demonstrated that the kinase activity was responsible for the synergistic effect of NOK and c-Src on STAT3 activation. In addition, NOK and c-Src synergistically promoted cell proliferation and tumor growth in nude mice. Taken together, our results indicate that NOK associates with c-Src and promotes c-Src-induced STAT3 activation in a kinase-dependent manner. We proposed that the axis that NOK promoted c-Src-induced STAT3 activation is critical in cell proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Sihan Liu
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Guancheng Jiang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wanli Zhai
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Liu Yang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Mengdi Li
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Bingtao Zhu
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
19
|
Reilly SM, Hung CW, Ahmadian M, Zhao P, Keinan O, Gomez AV, DeLuca JH, Dadpey B, Lu D, Zaid J, Poirier B, Peng X, Yu RT, Downes M, Liddle C, Evans RM, Murphy AN, Saltiel AR. Catecholamines suppress fatty acid re-esterification and increase oxidation in white adipocytes via STAT3. Nat Metab 2020; 2:620-634. [PMID: 32694788 PMCID: PMC7384260 DOI: 10.1038/s42255-020-0217-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Catecholamines stimulate the mobilization of stored triglycerides in adipocytes to provide fatty acids (FAs) for other tissues. However, a large proportion is taken back up and either oxidized or re-esterified. What controls the disposition of these FAs in adipocytes remains unknown. Here, we report that catecholamines redirect FAs for oxidation through the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Adipocyte STAT3 is phosphorylated upon activation of β-adrenergic receptors, and in turn suppresses FA re-esterification to promote FA oxidation. Adipocyte-specific Stat3 KO mice exhibit normal rates of lipolysis, but exhibit defective lipolysis-driven oxidative metabolism, resulting in reduced energy expenditure and increased adiposity when they are on a high-fat diet. This previously unappreciated, non-genomic role of STAT3 explains how sympathetic activation can increase both lipolysis and FA oxidation in adipocytes, revealing a new regulatory axis in metabolism.
Collapse
Affiliation(s)
- Shannon M Reilly
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Chao-Wei Hung
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Maryam Ahmadian
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Peng Zhao
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omer Keinan
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Andrew V Gomez
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Julia H DeLuca
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Benyamin Dadpey
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Donald Lu
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Zaid
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - BreAnne Poirier
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoling Peng
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Christopher Liddle
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Anne N Murphy
- Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Cytokinetics, South San Francisco, CA, USA
| | - Alan R Saltiel
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
20
|
Bhattacharyya S, Feferman L, Han X, Xia K, Zhang F, Linhardt RJ, Tobacman JK. Increased CHST15 follows decline in arylsulfatase B (ARSB) and disinhibition of non-canonical WNT signaling: potential impact on epithelial and mesenchymal identity. Oncotarget 2020; 11:2327-2344. [PMID: 32595831 PMCID: PMC7299535 DOI: 10.18632/oncotarget.27634] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Expression of CHST15 (carbohydrate sulfotransferase 15; chondroitin 4-sulfate-6-sulfotransferase; BRAG), the sulfotransferase enzyme that adds 6-sulfate to chondroitin 4-sulfate (C4S) to make chondroitin 4,6-disulfate (chondroitin sulfate E, CSE), was increased in malignant prostate epithelium obtained by laser capture microdissection and following arylsulfatase B (ARSB; N-acetylgalactosamine-4-sulfatase) silencing in human prostate epithelial cells. Experiments in normal and malignant human prostate epithelial and stromal cells and tissues, in HepG2 cells, and in the ARSB-null mouse were performed to determine the pathway by which CHST15 expression is up-regulated when ARSB expression is reduced. Effects of Wnt-containing prostate stromal cell spent media and selective inhibitors of WNT, JNK, p38, SHP2, β-catenin, Rho, and Rac-1 signaling pathways were determined. Activation of WNT signaling followed declines in ARSB and Dickkopf WNT Signaling Pathway Inhibitor (DKK)3 and was required for increased CHST15 expression. The increase in expression of CHST15 followed activation of non-canonical WNT signaling and involved Wnt3A, Rac-1 GTPase, phospho-p38 MAPK, and nuclear DNA-bound GATA-3. Inhibition of JNK, Sp1, β-catenin nuclear translocation, or Rho kinase had no effect. Consistent with higher expression of CHST15 in prostate epithelium, disaccharide analysis showed higher levels of CSE and chondroitin 6-sulfate (C6S) disaccharides in prostate epithelial cells. In contrast, chondroitin 4-sulfate (C4S) disaccharides were greater in prostate stromal cells. CSE may contribute to increased C4S in malignant epithelium when GALNS (N-aceytylgalactosamine-6-sulfate sulfatase) is increased and ARSB is reduced. These effects increase chondroitin 4-sulfates and reduce chondroitin 6-sulfates, consistent with enhanced stromal characteristics and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VAMC, Chicago, IL, USA
| | - Leo Feferman
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VAMC, Chicago, IL, USA
| | - Xiaorui Han
- Department of Chemistry and Chemical Biology Rensselaer Polytechnic Insitute, Troy, NY, USA
| | - Ke Xia
- Department of Chemistry and Chemical Biology Rensselaer Polytechnic Insitute, Troy, NY, USA
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology Rensselaer Polytechnic Insitute, Troy, NY, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology Rensselaer Polytechnic Insitute, Troy, NY, USA
| | - Joanne K Tobacman
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VAMC, Chicago, IL, USA
| |
Collapse
|
21
|
Bharadwaj U, Kasembeli MM, Robinson P, Tweardy DJ. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol Rev 2020; 72:486-526. [PMID: 32198236 PMCID: PMC7300325 DOI: 10.1124/pr.119.018440] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Before it was molecularly cloned in 1994, acute-phase response factor or signal transducer and activator of transcription (STAT)3 was the focus of intense research into understanding the mammalian response to injury, particularly the acute-phase response. Although known to be essential for liver production of acute-phase reactant proteins, many of which augment innate immune responses, molecular cloning of acute-phase response factor or STAT3 and the research this enabled helped establish the central function of Janus kinase (JAK) family members in cytokine signaling and identified a multitude of cytokines and peptide hormones, beyond interleukin-6 and its family members, that activate JAKs and STAT3, as well as numerous new programs that their activation drives. Many, like the acute-phase response, are adaptive, whereas several are maladaptive and lead to chronic inflammation and adverse consequences, such as cachexia, fibrosis, organ dysfunction, and cancer. Molecular cloning of STAT3 also enabled the identification of other noncanonical roles for STAT3 in normal physiology, including its contribution to the function of the electron transport chain and oxidative phosphorylation, its basal and stress-related adaptive functions in mitochondria, its function as a scaffold in inflammation-enhanced platelet activation, and its contributions to endothelial permeability and calcium efflux from endoplasmic reticulum. In this review, we will summarize the molecular and cellular biology of JAK/STAT3 signaling and its functions under basal and stress conditions, which are adaptive, and then review maladaptive JAK/STAT3 signaling in animals and humans that lead to disease, as well as recent attempts to modulate them to treat these diseases. In addition, we will discuss how consideration of the noncanonical and stress-related functions of STAT3 cannot be ignored in efforts to target the canonical functions of STAT3, if the goal is to develop drugs that are not only effective but safe. SIGNIFICANCE STATEMENT: Key biological functions of Janus kinase (JAK)/signal transducer and activator of transcription (STAT)3 signaling can be delineated into two broad categories: those essential for normal cell and organ development and those activated in response to stress that are adaptive. Persistent or dysregulated JAK/STAT3 signaling, however, is maladaptive and contributes to many diseases, including diseases characterized by chronic inflammation and fibrosis, and cancer. A comprehensive understanding of JAK/STAT3 signaling in normal development, and in adaptive and maladaptive responses to stress, is essential for the continued development of safe and effective therapies that target this signaling pathway.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Moses M Kasembeli
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Prema Robinson
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
22
|
Swiatek-Machado K, Kaminska B. STAT Signaling in Glioma Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:203-222. [PMID: 32034715 DOI: 10.1007/978-3-030-30651-9_10] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
STAT (signal transducers and activators of transcription) are latent cytoplasmic transcription factors that function as downstream effectors of cytokine and growth factor receptor signaling. The canonical JAK/STAT signaling pathway involves the activation of Janus kinases (JAK) or growth factors receptor kinases, phosphorylation of STAT proteins, their dimerization and translocation into the nucleus where STATs act as transcription factors with pleiotropic downstream effects. STAT signaling is tightly controlled with restricted kinetics due to action of its negative regulators. While STAT1 is believed to play an important role in growth arrest and apoptosis, and to act as a tumor suppressor, STAT3 and 5 are involved in promoting cell cycle progression, cellular transformation, and preventing apoptosis. Aberrant activation of STATs, in particular STAT3 and STAT5, have been found in a large number of human tumors, including gliomas and may contribute to oncogenesis. In this chapter, we have (1) summarized the mechanisms of STAT activation in normal and malignant signaling; (2) discussed evidence for the critical role of constitutively activated STAT3 and STAT5 in glioma pathobiology; (3) disclosed molecular and pharmacological strategies to interfere with STAT signaling for potential therapeutic intervention in gliomas.
Collapse
Affiliation(s)
- Karolina Swiatek-Machado
- Laboratory of Transcription Regulation, Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, PL 02-093, Warsaw, Poland.
| | - Bozena Kaminska
- Laboratory of Transcription Regulation, Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, PL 02-093, Warsaw, Poland
| |
Collapse
|
23
|
Baturcam E, Vollmer S, Schlüter H, Maciewicz RA, Kurian N, Vaarala O, Ludwig S, Cunoosamy DM. MEK inhibition drives anti-viral defence in RV but not RSV challenged human airway epithelial cells through AKT/p70S6K/4E-BP1 signalling. Cell Commun Signal 2019; 17:78. [PMID: 31319869 PMCID: PMC6639958 DOI: 10.1186/s12964-019-0378-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/29/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The airway epithelium is a major target tissue in respiratory infections, and its antiviral response is mainly orchestrated by the interferon regulatory factor-3 (IRF3), which subsequently induces type I (β) and III (λ) interferon (IFN) signalling. Dual specificity mitogen-activated protein kinase kinase (MEK) pathway contributes to epithelial defence, but its role in the regulation of IFN response in human primary airway epithelial cells (AECs) is not fully understood. Here, we studied the impact of a small-molecule inhibitor (MEKi) on the IFN response following challenge with two major respiratory viruses rhinovirus (RV2) and respiratory syncytial virus (RSVA2) and a TLR3 agonist, poly(I:C). METHODS The impact of MEKi on viral load and IFN response was evaluated in primary AECs with or without a neutralising antibody against IFN-β. Quantification of viral load was determined by live virus assay and absolute quantification using qRT-PCR. Secretion of cytokines was determined by AlphaLISA/ELISA and expression of interferon-stimulated genes (ISGs) was examined by qRT-PCR and immunoblotting. A poly(I:C) model was also used to further understand the molecular mechanism by which MEK controls IFN response. AlphaLISA, siRNA-interference, immunoblotting, and confocal microscopy was used to investigate the effect of MEKi on IRF3 activation and signalling. The impact of MEKi on ERK and AKT signalling was evaluated by immunoblotting and AlphaLISA. RESULTS Here, we report that pharmacological inhibition of MEK pathway augments IRF3-driven type I and III IFN response in primary human AECs. MEKi induced activation of PI3K-AKT pathway, which was associated with phosphorylation/inactivation of the translational repressor 4E-BP1 and activation of the protein synthesis regulator p70 S6 kinase, two critical translational effectors. Elevated IFN-β response due to MEKi was also attributed to decreased STAT3 activation, which consequently dampened expression of the transcriptional repressor of IFNB1 gene, PRDI-BF1. Augmented IFN response translated into inhibition of rhinovirus 2 replication in primary AECs but not respiratory syncytial virus A2. CONCLUSIONS Our findings unveil MEK as a key molecular mechanism by which rhinovirus dampens the epithelial cell's antiviral response. Our study provides a better understanding of the role of signalling pathways in shaping the antiviral response and suggests the use of MEK inhibitors in anti-viral therapy against RV.
Collapse
Affiliation(s)
- Engin Baturcam
- Early Respiratory, Inflammation & Autoimmunity, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden.
| | - Stefan Vollmer
- Early Respiratory, Inflammation & Autoimmunity, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Holger Schlüter
- Early Respiratory, Inflammation & Autoimmunity, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Rose A Maciewicz
- Early Respiratory, Inflammation & Autoimmunity, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Nisha Kurian
- Precision Medicine, R&D Oncology, AstraZeneca, Gothenburg, Sweden
| | - Outi Vaarala
- Early Respiratory, Inflammation & Autoimmunity, R&D BioPharmaceuticals, Gaithersburg, USA
| | - Stephan Ludwig
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | | |
Collapse
|
24
|
Choi JY, Yun J, Hwang CJ, Lee HP, Kim HD, Chun H, Park PH, Choi DY, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) Phenol Ameliorates MPTP-Induced Dopaminergic Neurodegeneration by Inhibiting the STAT3 Pathway. Int J Mol Sci 2019; 20:ijms20112632. [PMID: 31146332 PMCID: PMC6600543 DOI: 10.3390/ijms20112632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is implicated in dopaminergic neurodegeneration. We have previously demonstrated that (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a selective signal transducer and activator of transcription 3 (STAT3) inhibitor, has anti-inflammatory properties in several inflammatory disease models. We investigated whether MMPP could protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic cell loss and behavioral impairment. Imprinting control region (ICR) mice (8 weeks old, n = 10 per group) were administered MMPP (5 mg/kg) in drinking water for 1 month, and injected with MPTP (15 mg/kg, four times with 2 h intervals) during the last 7 days of treatment. MMPP decreased MPTP-induced behavioral impairments in rotarod, pole, and gait tests. We also showed that MMPP ameliorated dopamine depletion in the striatum and inflammatory marker elevation in primary cultured neurons by high-performance liquid chromatography and immunohistochemical analysis. Increased activation of STAT3, p38, and monoamine oxidase B (MAO-B) were observed in the substantia nigra and striatum after MPTP injection, effects that were attenuated by MMPP treatment. Furthermore, MMPP inhibited STAT3 activity and expression of neuroinflammatory proteins, including ionized calcium binding adaptor molecule 1 (Iba1), inducible nitric oxide synthase (iNOS), and glial fibrillary acidic protein (GFAP) in 1-methyl-4-phenylpyridinium (MPP+; 0.5 mM)-treated primary cultured cells. However, mitogen-activated protein kinase (MAPK) inhibitors augmented the activity of MMPP. Collectively, our results suggest that MMPP may be an anti-inflammatory agent that attenuates dopaminergic neurodegeneration and neuroinflammation through MAO-B and MAPK pathway-dependent inhibition of STAT3 activation.
Collapse
Affiliation(s)
- Ji Yeon Choi
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hae Deun Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hyungok Chun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Dong Young Choi
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| |
Collapse
|
25
|
Zhang H, Huang L, Tao L, Zhang J, Wang F, Zhang X, Fu L. Secalonic acid D induces cell apoptosis in both sensitive and ABCG2-overexpressing multidrug resistant cancer cells through upregulating c-Jun expression. Acta Pharm Sin B 2019; 9:516-525. [PMID: 31193763 PMCID: PMC6543021 DOI: 10.1016/j.apsb.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/04/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
Secalonic acid D (SAD) could inhibit cell growth in not only sensitive cells but also multidrug resistant (MDR) cells. However, the molecular mechanisms need to be elucidated. Here, we identified that SAD possessed potent cytotoxicity in 3 pairs of MDR and their parental sensitive cells including S1-MI-80 and S1, H460/MX20 and H460, MCF-7/ADR and MCF-7 cells. Furthermore, SAD induced cell G2/M phase arrest via the downregulation of cyclin B1 and the increase of CDC2 phosphorylation. Importantly, JNK pathway upregulated the expression of c-Jun in protein level and increased c-Jun phosphorylation induced by SAD, which was linked to cell apoptosis via c-Jun/Src/STAT3 pathway. To investigate the mechanisms of upregulation of c-Jun protein by SAD, the mRNA expression level and degradation of c-Jun were examined. We found that SAD did not alter the mRNA level of c-Jun but inhibited its proteasome-dependent degradation. Taken together, these results implicate that SAD induces cancer cell death through c-Jun/Src/STAT3 signaling axis by inhibiting the proteasome-dependent degradation of c-Jun in both sensitive cells and ATP-binding cassette transporter sub-family G member 2 (ABCG2)-mediated MDR cells.
Collapse
Key Words
- ABCB1, ATP-binding cassette subfamily B member 1
- ABCG2
- ABCG2, ATP-binding cassette transporter sub-family G member 2
- AP-1, activating protein-1
- Apoptosis
- CHX, cycloheximide
- HUVEC, human umbilical vein endothelial cells
- JNKs, c-Jun N-terminal kinases
- MAPKs, mitogen-activated protein kinases
- MDR, multidrug resistance
- MTT, 3-(4,5-dimethylthiazol-yl)-2,5-diphenyltetrazolium bromide
- Multidrug resistance
- NCM460, human normal colon epithelial cells
- RT-PCR, Real-time polymerase chain reaction
- SAD, Secalonic acid D
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- SP, side population
- Secalonic acid D
- c-Jun
Collapse
Affiliation(s)
- Hong Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
| | - Liyan Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
| | - Liyang Tao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
| | - Jianye Zhang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Fang Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
| | - Xu Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
| | - Liwu Fu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou 510060, China
- Corresponding author. Tel.: +86 20 87343163; fax: +86 20 87343170.
| |
Collapse
|
26
|
Icli B, Wu W, Ozdemir D, Li H, Haemmig S, Liu X, Giatsidis G, Cheng HS, Avci SN, Kurt M, Lee N, Guimaraes RB, Manica A, Marchini JF, Rynning SE, Risnes I, Hollan I, Croce K, Orgill DP, Feinberg MW. MicroRNA-135a-3p regulates angiogenesis and tissue repair by targeting p38 signaling in endothelial cells. FASEB J 2019; 33:5599-5614. [PMID: 30668922 PMCID: PMC6436660 DOI: 10.1096/fj.201802063rr] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/02/2019] [Indexed: 12/26/2022]
Abstract
Angiogenesis is a critical process in repair of tissue injury that is regulated by a delicate balance between pro- and antiangiogenic factors. In disease states associated with impaired angiogenesis, we identified that miR-135a-3p is rapidly induced and serves as an antiangiogenic microRNA (miRNA) by targeting endothelial cell (EC) p38 signaling in vitro and in vivo. MiR-135a-3p overexpression significantly inhibited EC proliferation, migration, and network tube formation in matrigel, whereas miR-135-3p neutralization had the opposite effects. Mechanistic studies using transcriptomic profiling, bioinformatics, 3'-UTR reporter and miRNA ribonucleoprotein complex -immunoprecipitation assays, and small interfering RNA dependency studies revealed that miR-135a-3p inhibits the p38 signaling pathway in ECs by targeting huntingtin-interacting protein 1 (HIP1). Local delivery of miR-135a-3p inhibitors to wounds of diabetic db/db mice markedly increased angiogenesis, granulation tissue thickness, and wound closure rates, whereas local delivery of miR-135a-3p mimics impaired these effects. Finally, through gain- and loss-of-function studies in human skin organoids as a model of tissue injury, we demonstrated that miR-135a-3p potently modulated p38 signaling and angiogenesis in response to VEGF stimulation by targeting HIP1. These findings establish miR-135a-3p as a pivotal regulator of pathophysiological angiogenesis and tissue repair by targeting a VEGF-HIP1-p38K signaling axis, providing new targets for angiogenic therapy to promote tissue repair.-Icli, B., Wu, W., Ozdemir, D., Li, H., Haemmig, S., Liu, X., Giatsidis, G., Cheng, H. S., Avci, S. N., Kurt, M., Lee, N., Guimaraes, R. B., Manica, A., Marchini, J. F., Rynning, S. E., Risnes, I., Hollan, I., Croce, K., Orgill, D. P., Feinberg, M. W. MicroRNA-135a-3p regulates angiogenesis and tissue repair by targeting p38 signaling in endothelial cells.
Collapse
Affiliation(s)
- Basak Icli
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Winona Wu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Denizhan Ozdemir
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Hao Li
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan Haemmig
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xin Liu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giorgio Giatsidis
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Henry S. Cheng
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Seyma Nazli Avci
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Merve Kurt
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathan Lee
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raphael Boesche Guimaraes
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, Rio Grande do Sul, Brazil
| | - Andre Manica
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, Rio Grande do Sul, Brazil
| | - Julio F. Marchini
- Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - Stein Erik Rynning
- Department of Cardiac Surgery, LHL Hospital Gardermoen, Jessheim, Norway
| | - Ivar Risnes
- Department of Cardiac Surgery, LHL Hospital Gardermoen, Jessheim, Norway
| | - Ivana Hollan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Rheumatology Department, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Kevin Croce
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dennis P. Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Prasad S, Ramachandran S, Gupta N, Kaushik I, Srivastava SK. Cancer cells stemness: A doorstep to targeted therapy. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165424. [PMID: 30818002 DOI: 10.1016/j.bbadis.2019.02.019] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 02/07/2023]
Abstract
Recent advances in research on cancer have led to understand the pathogenesis of cancer and development of new anticancer drugs. Despite of these advancements, many tumors have been found to recur, undergo metastasis and develop resistance to therapy. Accumulated evidences suggest that small population of cancer cells known as cancer stem cells (CSC) are responsible for reconstitution and propagation of the disease. CSCs possess the ability to self-renew, differentiate and proliferate like normal stem cells. CSCs also appear to have resistance to anti-cancer therapies and subsequent relapse. The underlying stemness properties of the CSCs are reliant on multiple molecular targets such as signaling pathways, cell surface molecules, tumor microenvironment, apoptotic pathways, microRNA, stem cell differentiation, and drug resistance markers. Thus an effective therapeutic strategy relies on targeting CSCs to overcome the possible tumor relapse and chemoresistance. The targeted inhibition of these stem cell biomarkers is one of the promising approaches to eliminate cancer stemness. This review article summarizes possible targets of cancer cell stemness for the complete treatment of cancer.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sharavan Ramachandran
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Nehal Gupta
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Itishree Kaushik
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
28
|
PI3k and Stat3: Oncogenes that are Required for Gap Junctional, Intercellular Communication. Cancers (Basel) 2019; 11:cancers11020167. [PMID: 30717267 PMCID: PMC6406562 DOI: 10.3390/cancers11020167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/21/2019] [Accepted: 01/26/2019] [Indexed: 12/12/2022] Open
Abstract
Gap junctional, intercellular communication (GJIC) is interrupted in cells transformed by oncogenes such as activated Src. The Src effector, Ras, is required for this effect, so that Ras inhibition restores GJIC in Src-transformed cells. Interestingly, the inhibition of the Src effector phosphatidyl-inositol-3 kinase (PI3k) or Signal Transducer and Activator of Transcription-3 (Stat3) pathways does not restore GJIC. In the contrary, inhibition of PI3k or Stat3 in non-transformed rodent fibroblasts or epithelial cells or certain human lung carcinoma lines with extensive GJIC inhibits communication, while mutational activation of PI3k or Stat3 increases GJIC. Therefore, it appears that oncogenes such as activated Src have a dual role upon GJIC; acting as inhibitors of communication through the Ras pathway, and as activators through activation of PI3k or Stat3. In the presence of high Src activity the inhibitory functions prevail so that the net effect is gap junction closure. PI3k and Stat3 constitute potent survival signals, so that their inhibition in non-transformed cells triggers apoptosis which, in turn, has been independently demonstrated to suppress GJIC. The interruption of gap junctional communication would confine the apoptotic event to single cells and this might be essential for the maintenance of tissue integrity. We hypothesize that the GJIC activation by PI3k or Stat3 may be linked to their survival function.
Collapse
|
29
|
The actin cytoskeleton is important for rotavirus internalization and RNA genome replication. Virus Res 2019; 263:27-33. [PMID: 30639190 PMCID: PMC7173133 DOI: 10.1016/j.virusres.2019.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/13/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
Different stages of the rotavirus lifecycle depend on the dynamics of the actin cytoskeleton. Alpha-actinin, Diaph, and the GTPase Cdc42 are important for virus entry. The GTPAse Rac1 is required for maximal viral RNA synthesis.
Numerous host factors are required for the efficient replication of rotavirus, including the activation and inactivation of several cell signaling pathways. One of the cellular structures that are reorganized during rotavirus infection is the actin cytoskeleton. In this work, we report that the dynamics of the actin microfilaments are important at different stages of the virus life cycle, specifically, during virus internalization and viral RNA synthesis at 6 h post-infection. Our results show that the actin-binding proteins alpha-actinin 4 and Diaph, as well as the Rho-family small GTPase Cdc42 are necessary for an efficient virus entry, while GTPase Rac1 is required for maximal viral RNA synthesis.
Collapse
|
30
|
A high-throughput fluorescence polarization assay for discovering inhibitors targeting the DNA-binding domain of signal transducer and activator of transcription 3 (STAT3). Oncotarget 2018; 9:32690-32701. [PMID: 30220975 PMCID: PMC6135694 DOI: 10.18632/oncotarget.26013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/31/2018] [Indexed: 11/25/2022] Open
Abstract
Anti-cancer drug discovery efforts to directly inhibit the signal transducer and activator of transcription 3 (STAT3) have been active for over a decade following the discovery that 70% of cancers exhibit elevated STAT3 activity. The majority of research has focused on attenuating STAT3 activity through preventing homo-dimerization by targeting the SH2 or transcriptional activation domains. Such dimerization inhibitors have not yet reached the market. However, an alternative strategy focussed on preventing STAT3 DNA-binding through targeting the DNA-binding domain (DBD) offers new drug design opportunities. Currently, only EMSA and ELISA-based methods have been implemented with suitable reliability to characterize STAT3 DBD inhibitors. Herein, we present a new orthogonal, fluorescence polarization (FP) assay suitable for high-throughput screening of molecules. This assay, using a STAT3127-688 construct, was developed and optimized to screen molecules that attenuate the STAT3:DNA association with good reliability (Z’ value > 0.6) and a significant contrast (signal-to-noise ratio > 15.0) at equilibrium. The assay system was stable over a 48 hour period. Significantly, the assay is homogeneous and simple to implement for high-throughput screening compared to EMSA and ELISA. Overall, this FP assay offers a new way to identify and characterize novel molecules that inhibit STAT3:DNA association.
Collapse
|
31
|
Choi HE, Kwak HJ, Kim SK, Cheon HG. Foenumoside B isolated from Lysimachia foenum-graecum extract suppresses LPS-induced inflammatory response via NF-κB/AP-1 inactivation in murine macrophages and in endotoxin-induced shock model. Eur J Pharmacol 2018; 832:120-128. [DOI: 10.1016/j.ejphar.2018.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 10/16/2022]
|
32
|
O-Glycosylation with O-linked β-N-acetylglucosamine increases vascular contraction: Possible modulatory role on Interleukin-10 signaling pathway. Life Sci 2018; 209:78-84. [PMID: 30075176 DOI: 10.1016/j.lfs.2018.07.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 01/29/2023]
Abstract
AIMS The interleukin-10 (IL-10) is an immuno-regulatory cytokine that plays a protective effect in the vasculature. IL-10 binding to its receptor, activating the IL-10/JAK1/STAT3 cascade to exert its effects. Therefore, STAT3 phosphorylation is essential for IL-10 actions. O-Glycosylation with linked β-N-acetylglucosamine (O-GlcNAc) is a post-translational modification able to regulate many proteins by interfering with protein on a phosphorylation level. Our aim was to determine whether O-GlcNAc promotes the inhibition of IL-10-pathway (JAK1/STAT3/IL-10), inactivationg its action in the vasculature. MAIN METHODS Mice (C57BL/6) aortic segments were incubated with vehicle or Thiamet G (0.1 mM, for 24 h) to increase global O-GlcNAc levels. Aortas from knockout mice for IL-10 were also used. Vascular reactivity and western blot tests were performed to evaluate protein expression. KEY FINDINGS High levels of O-GlcNAc, induced by Thiamet G incubation, increased vascular expression of JAK1, but decreased expression and activity of STAT3. In addition, IL-10 levels were diminished in arteries treated with Thiamet G. Absence of IL-10, as well as augmented O-GlcNAcylation, increased vascular reactivity to constrictor stimuli, an effect that was abolished by ERK 1/2 inhibitor. High levels of O-GlcNAc and the absence of IL-10 also leads to increased vascular expression of ERK1/2. SIGNIFICANCE Our data suggest that O-GlcNAc modification seems to (dys)regulate IL-10 signaling pathway and consequently, compromise the protective effect of this cytokine in vasculature. It is possible that there is a promising relationship in pathophysiological conditions where changes in O-GlcNAcylation and IL-10 levels are observed, such as hypertension and diabetes.
Collapse
|
33
|
Galoczova M, Coates P, Vojtesek B. STAT3, stem cells, cancer stem cells and p63. Cell Mol Biol Lett 2018; 23:12. [PMID: 29588647 PMCID: PMC5863838 DOI: 10.1186/s11658-018-0078-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/07/2018] [Indexed: 12/15/2022] Open
Abstract
Signal Transducer and Activator of Transcription 3 (STAT3) is a transcription factor with many important functions in the biology of normal and transformed cells. Its regulation is highly complex as it is involved in signaling pathways in many different cell types and under a wide variety of conditions. Besides other functions, STAT3 is an important regulator of normal stem cells and cancer stem cells. p63 which is a member of the p53 protein family is also involved in these functions and is both physically and functionally connected with STAT3. This review summarizes STAT3 function and regulation, its role in stem cell and cancer stem cell properties and highlights recent reports about its relationship to p63.
Collapse
Affiliation(s)
- Michaela Galoczova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic
| | - Philip Coates
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic
| |
Collapse
|
34
|
Chernikov AV, Gudkov SV, Usacheva AM, Bruskov VI. Exogenous 8-oxo-7,8-dihydro-2′-deoxyguanosine: Biomedical properties, mechanisms of action, and therapeutic potential. BIOCHEMISTRY (MOSCOW) 2018. [DOI: 10.1134/s0006297917130089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Khawar MB, Mukhtar M, Abbasi MH, Sheikh N. IL-32θ: a recently identified anti-inflammatory variant of IL-32 and its preventive role in various disorders and tumor suppressor activity. Am J Transl Res 2017; 9:4726-4737. [PMID: 29218075 PMCID: PMC5714761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
Interleukin-32 theta (IL-32θ) is newly identified isoform of IL-32 which plays a vital role in inflammatory responses. Like IL-32α and IL-32β, IL-32θ isoform acts as an intracellular inflammatory modulator. It results in reduction of IL-1β production by attenuating the expression of PU.1 and inhibition of monocytes differentiation into macrophages. IL-32θ hinders TNF-α expression by inhibiting p38 MAPK and inhibitor of κB (IκB) as well. It also reserved STAT3-ZEB1 pathway leading to the inhibition of epithelial-mesenchymal transition (EMT) and stemness. Hence, it can be concluded that IL-32θ is an anti-inflammatory cytokine that can act as a tumor suppressor and can play vital role in colon cancer therapies. IL-32θ also plays a crucial role in immune system responses and cellular differentiation during disease pathogenesis. To our best knowledge this is the first ever review to condense the importance, precise mode of action in disease progression and latent remedial implications of IL-32θ in several inflammatory disorders.
Collapse
Affiliation(s)
| | - Maryam Mukhtar
- Department of Zoology, University of The Punjab, Q-A CampusLahore, 54590, Pakistan
| | | | - Nadeem Sheikh
- Department of Zoology, University of The Punjab, Q-A CampusLahore, 54590, Pakistan
- Centre for Applied Molecular Biology (CAMB), University of The Punjab87-West Canal Bank Road, Thokar Niaz Baig Lahore, Pakistan
| |
Collapse
|
36
|
Jo AR, Han HS, Seo S, Shin JS, Lee JY, Kim HJ, Lee KT. Inhibitory effect of moschamine isolated from Carthamus tinctorius on LPS-induced inflammatory mediators via AP-1 and STAT1/3 inactivation in RAW 264.7 macrophages. Bioorg Med Chem Lett 2017; 27:5245-5251. [PMID: 29102229 DOI: 10.1016/j.bmcl.2017.10.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/11/2017] [Accepted: 10/17/2017] [Indexed: 11/16/2022]
Abstract
Seeds of Carthamus tinctorius L. (Compositae) have been used in Korean traditional medicines for the treatment of cardiovascular and bone diseases. In this study, we investigated the anti-inflammatory effects of known serotonin derivatives (1-9) isolated from the ethyl acetate (EtOAc) soluble fraction from the seeds of C. tinctorius. Compound 2, identified as moschamine, most potently inhibited lipopolysaccharide (LPS)-induced production of prostaglandin E2 (PGE2) and nitric oxide (NO) in RAW 264.7 macrophages. Moschamine concentration-dependently inhibited LPS-induced PGE2 and NO production in RAW 264.7 macrophages. Consistent with these findings, moschamine suppressed the protein and mRNA levels of cyclooxygenase-2 (COX-2), microsomal prostaglandin E2 synthase (mPGES)-1, and inducible NO synthase (iNOS), interleukin (IL)-6, and IL-1β. In addition, pretreatment of moschamine significantly inhibited LPS-stimulated the transcriptional activity of activator protein-1 (AP-1) and the phosphorylation of signal transducer and activator of transcription (STAT)1/3 in RAW 264.7 macrophages. Moreover, moschamine inhibited LPS-induced the phosphorylation of p38 mitogen-activated protein kinase (p38) and extracellular signal-regulated kinase (ERK), but it had no effect on c-Jun N-terminal kinase (JNK). These results suggest that the mechanism of anti-inflammatory activity of moschamine is associated with the downregulation of COX-2, mPGES-1, iNOS, IL-6, and IL-1β expression through the suppression of AP-1 and STAT1/3 activation in LPS-induced RAW 264.7 macrophages.
Collapse
Affiliation(s)
- A-Ra Jo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hee-Soo Han
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seunghwan Seo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ji-Sun Shin
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hyoung Ja Kim
- Molecular Recognition Research Center, Materials and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
37
|
|
38
|
Vanlandingham PA, Nuno DJ, Quiambao AB, Phelps E, Wassel RA, Ma JX, Farjo KM, Farjo RA. Inhibition of Stat3 by a Small Molecule Inhibitor Slows Vision Loss in a Rat Model of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2017; 58:2095-2105. [PMID: 28395025 PMCID: PMC5386345 DOI: 10.1167/iovs.16-20641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Diabetic retinopathy is a leading cause of vision loss. Previous studies have shown signaling pathways mediated by Stat3 (signal transducer and activator of transcription 3) play a primary role in diabetic retinopathy progression. This study tested CLT-005, a small molecule inhibitor of Stat3, for its dose-dependent therapeutic effects on vision loss in a rat model of diabetic retinopathy. Methods Brown Norway rats were administered streptozotocin (STZ) to induce diabetes. CLT-005 was administered daily by oral gavage for 16 weeks at concentrations of 125, 250, or 500 mg/kg, respectively, beginning 4 days post streptozotocin administration. Systemic and ocular drug concentration was quantified with mass spectrometry. Visual function was monitored at 2-week intervals from 6 to 16 weeks using optokinetic tracking to measure visual acuity and contrast sensitivity. The presence and severity of cataracts was visually monitored and correlated to visual acuity. The transcription and translation of multiple angiogenic factors and inflammatory cytokines were measured by real-time polymerase chain reaction and Multiplex immunoassay. Results Streptozotocin-diabetic rats sustain progressive vision loss over 16 weeks, and this loss in visual function is rescued in a dose-dependent manner by CLT-005. This positive therapeutic effect correlates to the positive effects of CLT-005 on vascular leakage and the presence of inflammatory cytokines in the retina. Conclusions The present study indicates that Stat3 inhibition has strong therapeutic potential for the treatment of vision loss in diabetic retinopathy.
Collapse
Affiliation(s)
| | - Didier J Nuno
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | | | - Eric Phelps
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | - Ronald A Wassel
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Krysten M Farjo
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rafal A Farjo
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| |
Collapse
|
39
|
Momtaz S, Niaz K, Maqbool F, Abdollahi M, Rastrelli L, Nabavi SM. STAT3 targeting by polyphenols: Novel therapeutic strategy for melanoma. Biofactors 2017; 43:347-370. [PMID: 27896891 DOI: 10.1002/biof.1345] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 09/17/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023]
Abstract
Melanoma or malignant melanocytes appear with the low incidence rate, but very high mortality rate worldwide. Epidemiological studies suggest that polyphenolic compounds contribute for prevention or treatment of several cancers particularly melanoma. Such findings motivate to dig out novel therapeutic strategies against melanoma, including research toward the development of new chemotherapeutic and biologic agents that can target the tumor cells by different mechanisms. Recently, it has been found that signal transducer and activator of transcription 3 (STAT3) is activated in many cancer cases surprisingly. Different evidences supply the aspect that STAT3 activation plays a vital role in the metastasis, including proliferation of cells, survival, invasion, migration, and angiogenesis. This significant feature plays a vital role in various cellular processes, such as cell proliferation and survival. Here, we reviewed the mechanisms of the STAT3 pathway regulation and their role in promoting melanoma. Also, we have evaluated the emerging data on polyphenols (PPs) specifically their contribution in melanoma therapies with an emphasis on their regulatory/inhibitory actions in relation to STAT3 pathway and current progress in the development of phytochemical therapeutic techniques. An understanding of targeting STAT3 by PPs brings an opportunity to melanoma therapy. © 2016 BioFactors, 43(3):347-370, 2017.
Collapse
Affiliation(s)
- Saeideh Momtaz
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Kamal Niaz
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran
| | - Faheem Maqbool
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran
| | - Luca Rastrelli
- Dipartimento di Farmacia, University of Salerno, Fisciano, SA, Italy
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Paladino D, Yue P, Furuya H, Acoba J, Rosser CJ, Turkson J. A novel nuclear Src and p300 signaling axis controls migratory and invasive behavior in pancreatic cancer. Oncotarget 2016; 7:7253-67. [PMID: 26695438 PMCID: PMC4872783 DOI: 10.18632/oncotarget.6635] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
The presence of Src in the nuclear compartment has been previously reported, although its significance has remained largely unknown. We sought to delineate the functions of the nuclear pool of Src within the context of malignant progression. Active Src is localized within the nuclei of human pancreatic cancer cells and mouse fibroblasts over-expressing c-Src where it is associated with p300. Nuclear Src additionally promotes the tyrosine phosphorylation of p300 in pancreatic cancer Panc-1 cells. Src, together with p300, is associated with the high-mobility group AT-hook (HMGA)2 and SET and MYND domain-containing protein (SMYD)3 gene promoters and regulates their expression in a Src-dependent manner. These nuclear Src-dependent events correlate with anchorage-independent soft-agar growth and the migratory properties in both pancreatic Panc-1 cells and mouse fibroblasts over-expressing Src. Moreover, analyses of human pancreatic ductal adenocarcinoma (PDAC) tumor tissues detected the association of nuclear Src with the HMGA2 and SMYD3 gene promoters. Our findings for the first time show the critical importance of nuclear Src and p300 function in the migratory properties of pancreatic cancer cells. Further, data together identify a previously unknown role of nuclear Src in the regulation of gene expression in association with p300 within the context of cells harboring activated or over-expressing Src. This novel mechanism of nuclear Src-p300 axis in PDAC invasiveness and metastasis may provide an opportunity for developing more effective early clinical interventions for this lethal disease. Active Src is complexed with and phosphorylates p300 in the nucleus, and the complex is bound to HMGA2 and SMYD3 genes, thereby regulating their expression to promote pancreatic tumor cell migration and invasiveness.
Collapse
Affiliation(s)
- David Paladino
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA.,Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| | - Peibin Yue
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA.,Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| | - Hideki Furuya
- Clinical and Translational Research Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| | - Jared Acoba
- Clinical and Translational Research Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| | - Charles J Rosser
- Clinical and Translational Research Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| | - James Turkson
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA.,Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
41
|
Siavash H, Nikitakis N, Sauk J. Signal Transducers and Activators of Transcription: Insights into the Molecular Basis of Oral Cancer. ACTA ACUST UNITED AC 2016; 15:298-307. [DOI: 10.1177/154411130401500505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent efforts on developing more direct and effective targets for cancer therapy have revolved around a family of transcription factors known as STATs (signal transducers and activators of transcription). STAT proteins are latent cytoplasmic transcription factors that become activated in response to extracellular signaling proteins. STAT proteins have been convincingly reported to possess oncogenic properties in a plethora of human cancers, including oral and oropharyngeal cancer. Signal transduction pathways mediated by these oncogenic transcription factors and their regulation in oral cancer are the focus of this review.
Collapse
Affiliation(s)
- H. Siavash
- Department of Biomedical Sciences and
- Department of Diagnostic Sciences and Pathology, University of Maryland, Dental School, 666 West Baltimore Street, Room 4-C-02, Baltimore, MD 21201; and
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201
| | - N.G. Nikitakis
- Department of Biomedical Sciences and
- Department of Diagnostic Sciences and Pathology, University of Maryland, Dental School, 666 West Baltimore Street, Room 4-C-02, Baltimore, MD 21201; and
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201
| | - J.J. Sauk
- Department of Biomedical Sciences and
- Department of Diagnostic Sciences and Pathology, University of Maryland, Dental School, 666 West Baltimore Street, Room 4-C-02, Baltimore, MD 21201; and
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
42
|
Lee HH, Shin JS, Lee WS, Ryu B, Jang DS, Lee KT. Biflorin, Isolated from the Flower Buds of Syzygium aromaticum L., Suppresses LPS-Induced Inflammatory Mediators via STAT1 Inactivation in Macrophages and Protects Mice from Endotoxin Shock. JOURNAL OF NATURAL PRODUCTS 2016; 79:711-720. [PMID: 26977531 DOI: 10.1021/acs.jnatprod.5b00609] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Two chromone C-glucosides, biflorin (1) and isobiflorin (2), were isolated from the flower buds of Syzygium aromaticum L. (Myrtaceae). Here, inhibitory effects of 1 and 2 on lipopolysaccharide (LPS)-induced production of nitric oxide (NO) and prostaglandin E2 (PGE2) in RAW 264.7 macrophages were evaluated, and 1 (IC50 = 51.7 and 37.1 μM, respectively) was more potent than 2 (IC50 > 60 and 46.0 μM). The suppression of NO and PGE2 production by 1 correlated with inhibition of iNOS and COX-2 protein expression. Compound 1 reduced inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) mRNA expression via inhibition of their promoter activities. Compound 1 inhibited the LPS-induced production and mRNA expression of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6. Furthermore, 1 reduced p-STAT1 and p-p38 expression but did not affect the activity of nuclear factor κ light-chain enhancer of activated B cells (NF-κB) or activator protein 1 (AP-1). In a mouse model of LPS-induced endotoxemia, 1 reduced the mRNA levels of iNOS, COX-2, and TNF-α, and the phosphorylation-mediated activation of the signal transducer and activator of transcription 1 (STAT1), consequently improving the survival rates of mice. Compound 1 showed a significant anti-inflammatory effect on carrageenan-induced paw edema and croton-oil-induced ear edema in rats. The collective data indicate that the suppression of pro-inflammatory gene expression via p38 mitogen-activated protein kinase and STAT1 inactivation may be a mechanism for the anti-inflammatory activity of 1.
Collapse
Affiliation(s)
- Hwi-Ho Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Ji-Sun Shin
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Woo-Seok Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Byeol Ryu
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Dae Sik Jang
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, ‡Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, §Reactive Oxygen Species Medical Research Center College of Pharmacy, and ⊥Department of Physiology, School of Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| |
Collapse
|
43
|
Kaur T, Borse V, Sheth S, Sheehan K, Ghosh S, Tupal S, Jajoo S, Mukherjea D, Rybak LP, Ramkumar V. Adenosine A1 Receptor Protects Against Cisplatin Ototoxicity by Suppressing the NOX3/STAT1 Inflammatory Pathway in the Cochlea. J Neurosci 2016; 36:3962-77. [PMID: 27053204 PMCID: PMC4821909 DOI: 10.1523/jneurosci.3111-15.2016] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 02/16/2016] [Accepted: 02/26/2016] [Indexed: 01/19/2023] Open
Abstract
Cisplatin is a commonly used antineoplastic agent that produces ototoxicity that is mediated in part by increasing levels of reactive oxygen species (ROS) via the NOX3 NADPH oxidase pathway in the cochlea. Recent studies implicate ROS generation in mediating inflammatory and apoptotic processes and hearing loss by activating signal transducer and activator of transcription (STAT1). In this study, we show that the adenosine A1 receptor (A1AR) protects against cisplatin ototoxicity by suppressing an inflammatory response initiated by ROS generation via NOX3 NADPH oxidase, leading to inhibition of STAT1. Trans-tympanic administration of the A1AR agonist R-phenylisopropyladenosine (R-PIA) inhibited cisplatin-induced ototoxicity, as measured by auditory brainstem responses and scanning electron microscopy in male Wistar rats. This was associated with reduced NOX3 expression, STAT1 activation, tumor necrosis factor-α (TNF-α) levels, and apoptosis in the cochlea. In vitro studies in UB/OC-1 cells, an organ of Corti immortalized cell line, showed that R-PIA reduced cisplatin-induced phosphorylation of STAT1 Ser(727) (but not Tyr(701)) and STAT1 luciferase activity by suppressing the ERK1/2, p38, and JNK mitogen-activated protein kinase (MAPK) pathways.R-PIA also decreased the expression of STAT1 target genes, such as TNF-α, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and reduced cisplatin-mediated apoptosis. These data suggest that the A1AR provides otoprotection by suppressing NOX3 and inflammation in the cochlea and could serve as an ideal target for otoprotective drug therapy. SIGNIFICANCE STATEMENT Cisplatin is a widely used chemotherapeutic agent for the treatment of solid tumors. Its use results in significant and permanent hearing loss, for which no US Food and Drug Administration-approved treatment is currently available. In this study, we targeted the cochlear adenosine A1 receptor (A1AR) by trans-tympanic injections of the agonist R-phenylisopropyladenosine (R-PIA) and showed that it reduced cisplatin-induced inflammation and apoptosis in the rat cochlea and preserved hearing. The mechanism of protection involves suppression of the NOX3 NADPH oxidase enzyme, a major target of cisplatin-induced reactive oxygen species (ROS) generation in the cochlea. ROS initiates an inflammatory and apoptotic cascade in the cochlea by activating STAT1 transcription factor, which is attenuated byR-PIA. Therefore, trans-tympanic delivery of A1AR agonists could effectively treat cisplatin ototoxicity.
Collapse
Affiliation(s)
- Tejbeer Kaur
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | | | | | - Kelly Sheehan
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois 62794
| | | | | | | | - Debashree Mukherjea
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois 62794
| | - Leonard P Rybak
- Department of Pharmacology and Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois 62794
| | | |
Collapse
|
44
|
Miklossy G, Youn UJ, Yue P, Zhang M, Chen CH, Hilliard TS, Paladino D, Li Y, Choi J, Sarkaria JN, Kawakami JK, Wongwiwatthananukit S, Chen Y, Sun D, Chang LC, Turkson J. Hirsutinolide Series Inhibit Stat3 Activity, Alter GCN1, MAP1B, Hsp105, G6PD, Vimentin, TrxR1, and Importin α-2 Expression, and Induce Antitumor Effects against Human Glioma. J Med Chem 2015; 58:7734-48. [PMID: 26331426 DOI: 10.1021/acs.jmedchem.5b00686] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We report that hirsutinolide series, 6, 7, 10, 11, 20, and 22, and the semisynthetic analogues, 30, 31, 33, and 36, inhibit constitutively active signal transducer and activator of transcription (Stat)3 and malignant glioma phenotype. A position 13 lipophilic ester group is required for activity. Molecular modeling and nuclear magnetic resonance structural analyses reveal direct hirsutinolide:Stat3 binding. One-hour treatment of cells with 6 and 22 also upregulated importin subunit α-2 levels and repressed translational activator GCN1, microtubule-associated protein (MAP)1B, thioredoxin reductase (TrxR)1 cytoplasmic isoform 3, glucose-6-phosphate 1-dehydrogenase isoform a, Hsp105, vimentin, and tumor necrosis factor α-induced protein (TNAP)2 expression. Active hirsutinolides inhibited anchorage-dependent and three-dimensional spheroid growth, survival, and migration of human glioma lines and glioma patients' tumor-derived xenograft cells harboring constitutively active Stat3. Oral gavage delivery of 6 or 22 inhibited human glioma tumor growth in subcutaneous mouse xenografts. The inhibition of Stat3 signaling represents part of the hirsutinolide-mediated mechanisms to induce antitumor effects.
Collapse
Affiliation(s)
| | - Ui Joung Youn
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo , Hilo 96720, Hawaii, United States
| | | | - Mingming Zhang
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo , Hilo 96720, Hawaii, United States
| | - Chih-Hong Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope , Duarte 91010, California, United States
| | | | | | - Yifei Li
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope , Duarte 91010, California, United States
| | - Justin Choi
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope , Duarte 91010, California, United States
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic , Rochester 55905, Minnesota, United States
| | - Joel K Kawakami
- Division of Natural Sciences and Mathematics, Chaminade University , Honolulu 96816, Hawaii, United States
| | - Supakit Wongwiwatthananukit
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo , Hilo 96720, Hawaii, United States
| | - Yuan Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope , Duarte 91010, California, United States
| | - Dianqing Sun
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo , Hilo 96720, Hawaii, United States
| | - Leng Chee Chang
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo , Hilo 96720, Hawaii, United States
| | | |
Collapse
|
45
|
Arulanandam R, Batenchuk C, Angarita FA, Ottolino-Perry K, Cousineau S, Mottashed A, Burgess E, Falls TJ, De Silva N, Tsang J, Howe GA, Bourgeois-Daigneault MC, Conrad DP, Daneshmand M, Breitbach CJ, Kirn DH, Raptis L, Sad S, Atkins H, Huh MS, Diallo JS, Lichty BD, Ilkow CS, Le Boeuf F, Addison CL, McCart JA, Bell JC. VEGF-Mediated Induction of PRD1-BF1/Blimp1 Expression Sensitizes Tumor Vasculature to Oncolytic Virus Infection. Cancer Cell 2015. [PMID: 26212250 DOI: 10.1016/j.ccell.2015.06.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.
Collapse
Affiliation(s)
- Rozanne Arulanandam
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Cory Batenchuk
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Fernando A Angarita
- Toronto General Research Institute (TGRI), University Health Network, Toronto, ON M5G 2M9, Canada
| | - Kathryn Ottolino-Perry
- Toronto General Research Institute (TGRI), University Health Network, Toronto, ON M5G 2M9, Canada
| | - Sophie Cousineau
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Amelia Mottashed
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Emma Burgess
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Theresa J Falls
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Naomi De Silva
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jovian Tsang
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Grant A Howe
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | | | - David P Conrad
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Manijeh Daneshmand
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | | | - David H Kirn
- SillaJen Biotherapeutics, San Francisco, CA 94111-3380, USA
| | - Leda Raptis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Harold Atkins
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Michael S Huh
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Fabrice Le Boeuf
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Christina L Addison
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - J Andrea McCart
- Toronto General Research Institute (TGRI), University Health Network, Toronto, ON M5G 2M9, Canada; Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
46
|
Yue P, Lopez-Tapia F, Paladino D, Li Y, Chen CH, Namanja AT, Hilliard T, Chen Y, Tius MA, Turkson J. Hydroxamic Acid and Benzoic Acid-Based STAT3 Inhibitors Suppress Human Glioma and Breast Cancer Phenotypes In Vitro and In Vivo. Cancer Res 2015; 76:652-63. [PMID: 26088127 DOI: 10.1158/0008-5472.can-14-3558] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/18/2015] [Indexed: 12/31/2022]
Abstract
STAT3 offers an attractive target for cancer therapy, but small-molecule inhibitors with appealing pharmacologic properties have been elusive. Here, we report hydroxamic acid-based and benzoic acid-based inhibitors (SH5-07 and SH4-54, respectively) with robust bioactivity. Both inhibitors blocked STAT3 DNA-binding activity in vitro and in human glioma, breast, and prostate cancer cells and in v-Src-transformed murine fibroblasts. STAT3-dependent gene transcription was blocked along with Bcl-2, Bcl-xL, Mcl-1, cyclin D1, c-Myc, and survivin expression. Nuclear magnetic resonance analysis of STAT3-inhibitor complexes defined interactions with the SH2 and DNA-binding domains of STAT3. Ectopic expression of the SH2 domain in cells was sufficient to counter the STAT3-inhibitory effects of SH4-54. Neither compound appreciably affected STAT1 or STAT5 DNA-binding activities, STAT3-independent gene transcription, or activation of a panel of oncogenic kinases in malignant cells. Each compound decreased the proliferation and viability of glioma, breast, and prostate cancer cells and v-Src-transformed murine fibroblasts harboring constitutively active STAT3. Further, in mouse xenograft models of glioma and breast cancer, administration of SH5-07 or SH4-54 effectively inhibited tumor growth. Our results offer preclinical proof of concept for SH5-07 and SH4-54 as candidates for further development as cancer therapeutics.
Collapse
Affiliation(s)
- Peibin Yue
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Francisco Lopez-Tapia
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Department of Chemistry, University of Hawaii, Manoa, Honolulu, Hawaii
| | - David Paladino
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Yifei Li
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Chih-Hong Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Andrew T Namanja
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Tyvette Hilliard
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Yuan Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Marcus A Tius
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Department of Chemistry, University of Hawaii, Manoa, Honolulu, Hawaii
| | - James Turkson
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii.
| |
Collapse
|
47
|
Ebel ME, Awe O, Kaplan MH, Kansas GS. Diverse inflammatory cytokines induce selectin ligand expression on murine CD4 T cells via p38α MAPK. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:5781-8. [PMID: 25941329 PMCID: PMC4698157 DOI: 10.4049/jimmunol.1500485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/02/2015] [Indexed: 01/03/2023]
Abstract
Selectins are glycan-binding adhesion molecules that mediate the initial steps of leukocyte recognition of endothelium. Cytokines control numerous aspects of CD4 Th cell differentiation, but how cytokines control the induction of ligands for E- and P-selectin on Th cell subsets remains poorly understood. Among 20 cytokines that affect Th cell differentiation, we identified six that induce expression of selectin ligands on murine CD4 T cells above the low levels associated with TCR engagement: IL-12, IL-18, IL-27, IL-9, IL-25, and TGF-β1. Collectively, these six cytokines could potentially account for selectin ligand expression on all of the currently defined nonsessile Th cell lineages, including Th1, Th2, Th9, and Th17 cells, as well as regulatory T cells. Induction of selectin ligand expression by each of these six cytokines was almost completely inhibited by pharmacologic inhibition of p38 MAPK, but not other MAPKs, or by conditional genetic deletion of p38α MAPK. Analysis of the expression of key glycosyltransferase genes revealed that p38α signaling was selectively required for induction of Fut7 and Gcnt1 but not for the induction of St3gal4 or St3gal6. Constitutively active MKK6, an immediate upstream activator of p38 MAPK, induced selectin ligand expression equivalent to that of cytokines, and this induction was completely dependent on the expression of p38α. Our results identify the repertoire of cytokines responsible for selectin ligand induction on CD4 T cells and provide a mechanistic link between Th cell development and T cell migration.
Collapse
Affiliation(s)
- Mark E Ebel
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Olufolakemi Awe
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Mark H Kaplan
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Geoffrey S Kansas
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611;
| |
Collapse
|
48
|
Antitumor efficacy of the anti-interleukin-6 (IL-6) antibody siltuximab in mouse xenograft models of lung cancer. J Thorac Oncol 2015; 9:974-982. [PMID: 24922005 DOI: 10.1097/jto.0000000000000193] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Interleukin-6 (IL-6) can activate downstream signaling pathways in lung cancer cells, such as the STAT3 pathway, and is reported to be produced by tumor cells with activating EGFR mutations. We examined IL-6/STAT3 in lung cancer tumor tissues and the effects of siltuximab, a neutralizing antibody to human IL-6, in mouse models of lung cancer. METHODS IL-6 and STAT3 activation levels were compared with tumor histology and presence of KRAS mutations in snap-frozen, non-small-cell lung cancer tumors. The effects of siltuximab alone or in combination with erlotinib were examined in mouse xenograft models constructed using three cell line xenograft models and one primary explant mouse model. We examined the influence of cancer-associated fibroblasts (CAFs) on tumor growth and siltuximab effects. RESULTS IL-6 levels were higher in tumors of squamous cell versus adenocarcinoma histology and were not associated with presence of KRAS mutations. Tyrosine phosphorylation status of STAT3 did not correlate with tumor IL-6 levels. Serine phosphorylation of STAT3 was correlated with KRAS mutation status. Both tumor and stromal cells contributed to total IL-6 within tumors. Siltuximab had minimal effect as a single agent in xenografts with tumor cells alone; however, in models coadministered with CAFs, siltuximab had more potent effects on tumor inhibition. We observed no effects of combined erlotinib and siltuximab. CONCLUSIONS IL-6 is elevated in subsets of human NSCLCs, especially with squamous cell histology. Tumors supported by stromal production of IL-6 seem to be the most vulnerable to tumor growth inhibition by siltuximab.
Collapse
|
49
|
Yco LP, Mocz G, Opoku-Ansah J, Bachmann AS. Withaferin A Inhibits STAT3 and Induces Tumor Cell Death in Neuroblastoma and Multiple Myeloma. BIOCHEMISTRY INSIGHTS 2014; 7:1-13. [PMID: 25452693 PMCID: PMC4226392 DOI: 10.4137/bci.s18863] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/19/2014] [Accepted: 10/05/2014] [Indexed: 01/05/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor that has been implicated in many human cancers and has emerged as an ideal target for cancer therapy. Withaferin A (WFA) is a natural product with promising antiproliferative properties through its association with a number of molecular targets including STAT3. However, the effect of WFA in pediatric neuroblastoma (NB) and its interaction with STAT3 have not been reported. In this study, we found that WFA effectively induces dose-dependent cell death in high-risk and drug-resistant NB as well as multiple myeloma (MM) tumor cells, prevented interleukin-6 (IL-6)–mediated and persistently activated STAT3 phosphorylation at Y705, and blocked the transcriptional activity of STAT3. We further provide computational models that show that WFA binds STAT3 near the Y705 phospho-tyrosine residue of the STAT3 Src homology 2 (SH2) domain, suggesting that WFA prevents STAT3 dimer formation similar to BP-1-102, a well-established STAT3 inhibitor. Our findings propose that the antitumor activity of WFA is mediated at least in part through inhibition of STAT3 and provide a rationale for further drug development and clinical use in NB and MM.
Collapse
Affiliation(s)
- Lisette P Yco
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, USA. ; Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Gabor Mocz
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - John Opoku-Ansah
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, USA
| | - André S Bachmann
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, USA. ; Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA. ; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA. ; Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, USA
| |
Collapse
|
50
|
IL-32θ downregulates CCL5 expression through its interaction with PKCδ and STAT3. Cell Signal 2014; 26:3007-15. [PMID: 25280942 DOI: 10.1016/j.cellsig.2014.09.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/15/2014] [Accepted: 09/15/2014] [Indexed: 01/11/2023]
Abstract
Interleukin-32 (IL-32) exists in several isoforms and plays an important role in inflammatory response. Recently, we identified a new isoform, IL-32θ, and performed a microarray analysis to identify IL-32θ-regulated genes in THP-1 myelomonocytic cells. Upon stimulating IL-32θ-expressing THP-1 cells with phorbol myristate acetate (PMA), we found that the CCL5 transcript level was significantly reduced. We confirmed the downregulation of CCL5 protein expression by using an enzyme-linked immunosorbent assay (ELISA). Because STAT3 phosphorylation on Ser727 by PKCδ is reported to suppress CCL5 protein expression, we examined whether IL-32θ-mediated STAT3 Ser727 phosphorylation occurs through an interaction with PKCδ. In this study, we first demonstrate that IL-32θ interacts with PKCδ and STAT3 using co-immunoprecipitation (Co-IP) and pulldown assay. Moreover, STAT3 was rarely phosphorylated on Ser727 in the absence of IL-32θ, leading to the binding of STAT3 to the CCL5 promoter. These results indicate that IL-32θ, through its interaction with PKCδ, downregulates CCL5 expression by mediating the phosphorylation of STAT3 on Ser727 to render it transcriptionally inactive. Therefore, similar to what we have reported for IL-32α and IL-32β, our data from this study suggests that the newly identified IL-32θ isoform also acts as an intracellular modulator of inflammation.
Collapse
|