1
|
Sarvizadeh M, Hasanpour O, Naderi Ghale-Noie Z, Mollazadeh S, Rezaei M, Pourghadamyari H, Masoud Khooy M, Aschner M, Khan H, Rezaei N, Shojaie L, Mirzaei H. Allicin and Digestive System Cancers: From Chemical Structure to Its Therapeutic Opportunities. Front Oncol 2021; 11:650256. [PMID: 33987085 PMCID: PMC8111078 DOI: 10.3389/fonc.2021.650256] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022] Open
Abstract
Digestive system cancer tumors are one of the major causes of cancer-related fatalities; the vast majority of them are colorectal or gastric malignancies. Epidemiological evidence confirmed that allium-containing food, such as garlic, reduces the risk of developing malignancies. Among all compounds in garlic, allicin has been most researched, as it contains sulfur and produces many second degradation compounds, such as sulfur dioxide, diallyl sulfide (DAS), diallyl trisulfide (DATS), and diallyl disulfide (DADS) in the presence of enzymatic reactions in gastric juice. These substances have shown anti-inflammatory, antidiabetic, antihypertensive, antifungal, antiviral, antibacterial, and anticancer efficacy, including gastrointestinal (GI) cancers, leukemia, and skin cancers. Herein, we summarize the therapeutic potential of allicin in the treatment of GI cancers.
Collapse
Affiliation(s)
- Mahshad Sarvizadeh
- Nutrition and Endocrine Research Centre, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Hasanpour
- School of Paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Rezaei
- Department of Diabetes, Obesity and Metabolism, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pourghadamyari
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Nima Rezaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Layla Shojaie
- Department of Medicine, Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
2
|
Kohandel Z, Farkhondeh T, Aschner M, Samarghandian S. Anti-inflammatory effects of thymoquinone and its protective effects against several diseases. Biomed Pharmacother 2021; 138:111492. [PMID: 33743334 DOI: 10.1016/j.biopha.2021.111492] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Thymoquinone (TQ, 2-methyl-5-isopropyl-1, 4-benzoquinone), a monoterpene molecule present in Nigella sativa L., has an anti-inflammatory, anti-oxidant, and anti-apoptotic properties in several disorders such as asthma, hypertension, diabetes, inflammation, bronchitis, headache, eczema, fever, dizziness and influenza. TQ exerts its anti-inflammatory and anti-oxidant effects via several molecular pathways, including the release of cytokines, and activation of cyclooxygenase-2 (COX2), nuclear factor erythroid 2-related factor 2 (Nrf2), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), nuclear factor kappa-light-chain-enhancer of activated B (NF-Κβ). In this review, recent reports on the anti-inflammatory efficacy of TQ in heart disorders, respiratory diseases, neuroinflammation, diabetes and arthritis are summarized. We suggest that further investigation is necessary to better characterize the efficacy of TQ as a therapeutic agent.
Collapse
Affiliation(s)
- Zeynab Kohandel
- Department of Biology, Faculty of Sciences, University of Tehran, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran; Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, New York, USA
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
3
|
Activation of Interferon Signaling in Chronic Lymphocytic Leukemia Cells Contributes to Apoptosis Resistance via a JAK-Src/STAT3/Mcl-1 Signaling Pathway. Biomedicines 2021; 9:biomedicines9020188. [PMID: 33668421 PMCID: PMC7918075 DOI: 10.3390/biomedicines9020188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/18/2022] Open
Abstract
Besides their antiviral and immunomodulatory functions, type I (α/β) and II (γ) interferons (IFNs) exhibit either beneficial or detrimental effects on tumor progression. Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of abnormal CD5+ B lymphocytes that escape death. Drug resistance and disease relapse still occur in CLL. The triggering of IFN receptors is believed to be involved in the survival of CLL cells, but the underlying molecular mechanisms are not yet characterized. We show here that both type I and II IFNs promote the survival of primary CLL cells by counteracting the mitochondrial (intrinsic) apoptosis pathway. The survival process was associated with the upregulation of signal transducer and activator of transcription-3 (STAT3) and its target anti-apoptotic Mcl-1. Furthermore, the blockade of the STAT3/Mcl-1 pathway by pharmacological inhibitors against STAT3, TYK2 (for type I IFN) or JAK2 (for type II IFN) markedly reduced IFN-mediated CLL cell survival. Similarly, the selective Src family kinase inhibitor PP2 notably blocked IFN-mediated CLL cell survival by downregulating the protein levels of STAT3 and Mcl-1. Our work reveals a novel mechanism of resistance to apoptosis promoted by IFNs in CLL cells, whereby JAKs (TYK2, JAK2) and Src kinases activate in concert a STAT3/Mcl-1 signaling pathway. In view of current clinical developments of potent STAT3 and Mcl-1 inhibitors, a combination of conventional treatments with these inhibitors might thus constitute a new therapeutic strategy in CLL.
Collapse
|
4
|
Feng W, Ying WZ, Li X, Curtis LM, Sanders PW. Renoprotective effect of Stat1 deletion in murine aristolochic acid nephropathy. Am J Physiol Renal Physiol 2021; 320:F87-F96. [PMID: 33283645 PMCID: PMC7847048 DOI: 10.1152/ajprenal.00401.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/14/2023] Open
Abstract
Injured tubule epithelium stimulates a profibrotic milieu that accelerates loss of function in chronic kidney disease (CKD). This study tested the role of signal transducer and activator of transcription 1 (STAT1) in the progressive loss of kidney function in aristolochic acid (AA) nephropathy, a model of CKD. Mean serum creatinine concentration increased in wild-type (WT) littermates treated with AA, whereas Stat1-/- mice were protected. Focal increases in the apical expression of kidney injury molecule (KIM)-1 were observed in the proximal tubules of WT mice with AA treatment but were absent in Stat1-/- mice in the treatment group as well as in both control groups. A composite injury score, an indicator of proximal tubule injury, was reduced in Stat1-/- mice treated with AA. Increased expression of integrin-β6 and phosphorylated Smad2/3 in proximal tubules as well as interstitial collagen and fibronectin were observed in WT mice following AA treatment but were all decreased in AA-treated Stat1-/- mice. The data indicated that STAT1 activation facilitated the development of progressive kidney injury and interstitial fibrosis in AA nephropathy.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Zhong Ying
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xingsheng Li
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lisa M Curtis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
5
|
Illiano M, Conte M, Salzillo A, Ragone A, Spina A, Nebbioso A, Altucci L, Sapio L, Naviglio S. The KDM Inhibitor GSKJ4 Triggers CREB Downregulation via a Protein Kinase A and Proteasome-Dependent Mechanism in Human Acute Myeloid Leukemia Cells. Front Oncol 2020; 10:799. [PMID: 32582541 PMCID: PMC7289982 DOI: 10.3389/fonc.2020.00799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/23/2020] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a progressive hematopoietic-derived cancer arising from stepwise genetic mutations of the myeloid lineage. cAMP response element-binding protein (CREB) is a nuclear transcription factor, which plays a key role in the multistep process of leukemogenesis, thus emerging as an attractive potential drug target for AML treatment. Since epigenetic dysregulations, such as DNA methylation, histone modifications, as well as chromatin remodeling, are a frequent occurrence in AML, an increasing and selective number of epi-drugs are emerging as encouraging therapeutic agents. Here, we demonstrate that the histone lysine demethylases (KDMs) JMJD3/UTX inhibitor GSKJ4 results in both proliferation decrease and CREB protein downregulation in AML cells. We found that GSKJ4 clearly decreases CREB protein, but not CREB mRNA levels. By cycloheximide assay, we provide evidence that GSKJ4 reduces CREB protein stability; moreover, proteasome inhibition largely counteracts the GSKJ4-induced CREB downregulation. Very interestingly, a rapid CREB phosphorylation at the Ser133 residue precedes CREB protein decrease in response to GSKJ4 treatment. In addition, protein kinase A (PKA) inhibition, but not extracellular signal-regulated kinase (ERK)1/2 inhibition, almost completely prevents both GSKJ4-induced p-Ser133-CREB phosphorylation and CREB protein downregulation. Overall, our study enforces the evidence regarding CREB as a potential druggable target, identifies the small epigenetic molecule GSKJ4 as an “inhibitor” of CREB, and encourages the design of future GSKJ4-based studies for the development of innovative approaches for AML therapy.
Collapse
Affiliation(s)
- Michela Illiano
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alessia Salzillo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Ragone
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annamaria Spina
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigi Sapio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
6
|
Zhang S, Yang Z, Bao W, Liu L, You Y, Wang X, Shao L, Fu W, Kou X, Shen W, Yuan C, Hu B, Dang W, Nandakumar KS, Jiang H, Zheng M, Shen X. SNX10 (sorting nexin 10) inhibits colorectal cancer initiation and progression by controlling autophagic degradation of SRC. Autophagy 2019; 16:735-749. [PMID: 31208298 DOI: 10.1080/15548627.2019.1632122] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The non-receptor tyrosine kinase SRC is a key mediator of cellular protumorigenic signals. SRC is aberrantly over-expressed and activated in more than 80% of colorectal cancer (CRC) patients, therefore regulation of its stability and activity is essential. Here, we report a significant down regulation of SNX10 (sorting nexin 10) in human CRC tissues, which is closely related to tumor differentiation, TNM stage, lymph node metastasis and survival period. SNX10 deficiency in normal and neoplastic colorectal epithelial cells promotes initiation and progression of CRC in mice. SNX10 controls SRC levels by mediating autophagosome-lysosome fusion and SRC recruitment for autophagic degradation. These mechanisms ensure proper controlling of the activities of SRC-STAT3 and SRC-CTNNB1 signaling pathways by up-regulating SNX10 expression under stress conditions. These findings suggest that SNX10 acts as a tumor suppressor in CRC and it could be a potential therapeutic target for future development.Abbreviations: ACTB: actin beta; ATG5: autophagy related 5; ATG12: autophagy related 12; CQ: chloroquine; CRC: colorectal cancer; CTNNB1: catenin beta 1; EBSS: Earle's balanced salt solution; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAMP2: lysosomal associated membrane protein 2; MAP1LC3: microtubule associated protein 1 light chain 3; MKI67: marker of proliferation Ki-67; mRNA: messenger RNA; PX: phox homology; RT-qPCR: real time quantitative polymerase chain reaction; siRNA: small interfering RNA; SNX10: sorting nexin 10; SQSTM1: sequestosome 1; SRC: SRC proto-oncogene, non-receptor tyrosine kinase; STAT3: signal transducer and activator of transcription 3; WT: wild type.
Collapse
Affiliation(s)
- Sulin Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwen Yang
- Department of Pharmacy, Songjiang Hospital Affiliated Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weilian Bao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Lixin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Liming Shao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Fu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xinhui Kou
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Weixing Shen
- The Translational Medicine Research Center, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Congmin Yuan
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Bin Hu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wenzhen Dang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | | | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Li X, Ni J, Tang Y, Wang X, Tang H, Li H, Zhang S, Shen X. Allicin inhibits mouse colorectal tumorigenesis through suppressing the activation of STAT3 signaling pathway. Nat Prod Res 2018; 33:2722-2725. [DOI: 10.1080/14786419.2018.1465425] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Xiang Li
- Shanghai Kongjiang Senior High School, Shanghai, China
| | - Jiahui Ni
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yixin Tang
- Shanghai Kongjiang Senior High School, Shanghai, China
| | - Xu Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Huanhuan Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Haidong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Sulin Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Ferreli C, Lai C, August S, Buggy Y, Kumar P, Brownlow N, Parker P, Friedmann PS, Ardern-Jones M, Pickard C, Healy E. STAT4 expression and activation is increased during mitosis in vitro and in vivo in skin- and mucosa-derived cell types: implications in neoplastic and inflammatory skin diseases. J Eur Acad Dermatol Venereol 2017; 31:1663-1673. [PMID: 28516569 DOI: 10.1111/jdv.14342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/19/2017] [Indexed: 02/11/2024]
Abstract
BACKGROUND The signal transducer and activator of transcription-4 (STAT4/Stat4) is a transcription factor known to convey signals from interleukin-12, interleukin-23, and interferon-alpha/beta to the nucleus, resulting in activation of dendritic cells, T-helper cell differentiation and production of interferon-gamma. OBJECTIVE To demonstrate a novel role for STAT4 in cell mitosis. RESULTS Phosphoserine STAT4 (pSerSTAT4) is increased in cells undergoing mitosis and is distributed throughout the cytoplasm during this stage of the cell cycle, whilst phosphotyrosine STAT4 (pTyrSTAT4) is confined to the chromosomal compartment. This distinct pattern of pSerSTAT4 during mitosis is seen in vitro in human keratinocytes and in other cell types. This is also present in vivo in cells undergoing mitosis in normal skin, psoriasis and squamous cell carcinoma. Inhibition of STAT4 phosphorylation by lisofylline and depletion of STAT4 by RNA interference results in a delay in progression of mitosis and leads to a reduction in cells completing cytokinesis. CONCLUSION Our data demonstrate that STAT4 plays a role in enabling the normal and timely division of cells undergoing mitosis.
Collapse
Affiliation(s)
- C Ferreli
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology Unit, Department of Medical Sciences, Public Health University of Cagliari, Cagliari, Italy
| | - C Lai
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - S August
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - Y Buggy
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - P Kumar
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - N Brownlow
- London Research Institute, Lincoln's Inn Fields, London, UK
| | - P Parker
- London Research Institute, Lincoln's Inn Fields, London, UK
| | - P S Friedmann
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - M Ardern-Jones
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - C Pickard
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - E Healy
- Dermatopharmacology, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| |
Collapse
|
9
|
Goyal SN, Prajapati CP, Gore PR, Patil CR, Mahajan UB, Sharma C, Talla SP, Ojha SK. Therapeutic Potential and Pharmaceutical Development of Thymoquinone: A Multitargeted Molecule of Natural Origin. Front Pharmacol 2017; 8:656. [PMID: 28983249 PMCID: PMC5613109 DOI: 10.3389/fphar.2017.00656] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 09/04/2017] [Indexed: 12/16/2022] Open
Abstract
Thymoquinone, a monoterpene molecule is chemically known as 2-methyl-5-isopropyl-1, 4-benzoquinone. It is abundantly present in seeds of Nigella sativa L. that is popularly known as black cumin or black seed and belongs to the family Ranunculaceae. A large number of studies have revealed that thymoquinone is the major active constituent in N. sativa oil this constituent is responsible for the majority of the pharmacological properties. The beneficial organoprotective activities of thymoquinone in experimental animal models of different human diseases are attributed to the potent anti-oxidant and anti-inflammatory properties. Thymoquinone has also been shown to alter numerous molecular and signaling pathways in many inflammatory and degenerative diseases including cancer. Thymoquinone has been reported to possess potent lipophilicity and limited bioavailability and exhibits light and heat sensitivity. Altogether, these physiochemical properties encumber the successful formulation for the delivery of drug in oral dosages form and restrict the pharmaceutical development. In recent past, many efforts were undertaken to improve the bioavailability for clinical usage by manipulating the physiochemical parameters. The present review aimed to provide insights regarding the physicochemical characteristics, pharmacokinetics and the methods to promote pharmaceutical development and endorse the clinical usage of TQ in future by overcoming the associated physiochemical obstacles. It also enumerates briefly the pharmacological and molecular targets of thymoquinone as well as the pharmacological properties in various diseases and the underlying molecular mechanism. Though, a convincing number of experimental studies are available but human studies are not available with thymoquinone despite of the long history of use of black cumin in different diseases. Thus, the clinical studies including pharmacokinetic studies and regulatory toxicity studies are required to encourage the clinical development of thymoquinone.
Collapse
Affiliation(s)
- Sameer N. Goyal
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
- SVKM Institute of PharmacyDhule, India
| | - Chaitali P. Prajapati
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Prashant R. Gore
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Umesh B. Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Sandhya P. Talla
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Shreesh K. Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| |
Collapse
|
10
|
Hato SV, Figdor CG, Takahashi S, Pen AE, Halilovic A, Bol KF, Vasaturo A, Inoue Y, de Haas N, Verweij D, Van Herpen CML, Kaanders JH, van Krieken JHJM, Van Laarhoven HWM, Hooijer GKJ, Punt CJA, Asai A, de Vries IJM, Lesterhuis WJ. Direct inhibition of STAT signaling by platinum drugs contributes to their anti-cancer activity. Oncotarget 2017; 8:54434-54443. [PMID: 28903353 PMCID: PMC5589592 DOI: 10.18632/oncotarget.17661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 04/22/2017] [Indexed: 01/05/2023] Open
Abstract
Platinum-based chemotherapeutics are amongst the most powerful anti-cancer drugs. Although their exact mechanism of action is not well understood, it is thought to be mediated through covalent DNA binding. We investigated the effect of platinum-based chemotherapeutics on signaling through signal transducer and activator of transcription (STAT) proteins, which are involved in many oncogenic signaling pathways. We performed in vitro experiments in various cancer cell lines, investigating the effects of platinum chemotherapeutics on STAT phosphorylation and nuclear translocation, the expression of STAT-modulating proteins and downstream signaling pathways. Direct binding of platinum to STAT proteins was assessed using an AlphaScreen assay. Nuclear STAT3 expression was determined by immunohistochemistry and correlated with disease-free survival in retrospective cohorts of head and neck squamous cell carcinoma (HNSCC) patients treated with cisplatin-based chemoradiotherapy (n= 65) or with radiotherapy alone (n = 32). At clinically relevant concentrations, platinum compounds inhibited STAT phosphorylation, resulting in loss of constitutively activated STAT proteins in multiple distinct cancer cell lines. Platinum drugs specifically inhibited phospho-tyrosine binding to SH2 domains, thereby blocking STAT activation, and subsequently downregulating pro-survival- and anti-apoptotic- target genes. Importantly, we found that active STAT3 in tumors directly correlated with response to cisplatin-based chemoradiotherapy in HNSCC patients (p = 0.006). These findings provide insight into a novel, non-DNA-targeted mechanism of action of platinum drugs, and could be leveraged into the use of STAT expression as predictive biomarker for cisplatin chemotherapy and to potentiate other therapeutic strategies such as immunotherapy.
Collapse
Affiliation(s)
- Stanleyson V Hato
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Susumu Takahashi
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Anja E Pen
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Altuna Halilovic
- Department of Pathology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Kalijn F Bol
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Angela Vasaturo
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Yukie Inoue
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Nienke de Haas
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Dagmar Verweij
- Department of Pathology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carla M L Van Herpen
- Department of Medical Oncology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johannes H Kaanders
- Department of Radiation Oncology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johan H J M van Krieken
- Department of Pathology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Hanneke W M Van Laarhoven
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerrit K J Hooijer
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - W Joost Lesterhuis
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Current address: University of Western Australia, School of Medicine and Pharmacology, Perth, Australia
| |
Collapse
|
11
|
Thiagarajan D, Vedantham S, Ananthakrishnan R, Schmidt AM, Ramasamy R. Mechanisms of transcription factor acetylation and consequences in hearts. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:2221-2231. [PMID: 27543804 PMCID: PMC5159280 DOI: 10.1016/j.bbadis.2016.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/12/2016] [Accepted: 08/14/2016] [Indexed: 01/06/2023]
Abstract
Acetylation of proteins as a post-translational modification is gaining rapid acceptance as a cellular control mechanism on par with other protein modification mechanisms such as phosphorylation and ubiquitination. Through genetic manipulations and evolving proteomic technologies, identification and consequences of transcription factor acetylation is beginning to emerge. In this review, we summarize the field and discuss newly unfolding mechanisms and consequences of transcription factor acetylation in normal and stressed hearts. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | | | - Radha Ananthakrishnan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States.
| |
Collapse
|
12
|
STAT3 Represses Nitric Oxide Synthesis in Human Macrophages upon Mycobacterium tuberculosis Infection. Sci Rep 2016; 6:29297. [PMID: 27384401 PMCID: PMC4935992 DOI: 10.1038/srep29297] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/17/2016] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis is a successful intracellular pathogen. Numerous host innate immune responses signaling pathways are induced upon mycobacterium invasion, however their impact on M. tuberculosis replication is not fully understood. Here we reinvestigate the role of STAT3 specifically inside human macrophages shortly after M. tuberculosis uptake. We first show that STAT3 activation is mediated by IL-10 and occurs in M. tuberculosis infected cells as well as in bystander non-colonized cells. STAT3 activation results in the inhibition of IL-6, TNF-α, IFN-γ and MIP-1β. We further demonstrate that STAT3 represses iNOS expression and NO synthesis. Accordingly, the inhibition of STAT3 is detrimental for M. tuberculosis intracellular replication. Our study thus points out STAT3 as a key host factor for M. tuberculosis intracellular establishment in the early stages of macrophage infection.
Collapse
|
13
|
Bienaimé F, Muorah M, Yammine L, Burtin M, Nguyen C, Baron W, Garbay S, Viau A, Broueilh M, Blanc T, Peters D, Poli V, Anglicheau D, Friedlander G, Pontoglio M, Gallazzini M, Terzi F. Stat3 Controls Tubulointerstitial Communication during CKD. J Am Soc Nephrol 2016; 27:3690-3705. [PMID: 27153926 DOI: 10.1681/asn.2015091014] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/24/2016] [Indexed: 12/21/2022] Open
Abstract
In CKD, tubular cells may be involved in the induction of interstitial fibrosis, which in turn, leads to loss of renal function. However, the molecular mechanisms that link tubular cells to the interstitial compartment are not clear. Activation of the Stat3 transcription factor has been reported in tubular cells after renal damage, and Stat3 has been implicated in CKD progression. Here, we combined an experimental model of nephron reduction in mice from different genetic backgrounds and genetically modified animals with in silico and in vitro experiments to determine whether the selective activation of Stat3 in tubular cells is involved in the development of interstitial fibrosis. Nephron reduction caused Stat3 phosphorylation in tubular cells of lesion-prone mice but not in resistant mice. Furthermore, specific deletion of Stat3 in tubular cells significantly reduced the extent of interstitial fibrosis, which correlated with reduced fibroblast proliferation and matrix synthesis, after nephron reduction. Mechanistically, in vitro tubular Stat3 activation triggered the expression of a specific subset of paracrine profibrotic factors, including Lcn2, Pdgfb, and Timp1. Together, our results provide a molecular link between tubular and interstitial cells during CKD progression and identify Stat3 as a central regulator of this link and a promising therapeutic target.
Collapse
Affiliation(s)
- Frank Bienaimé
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling.,Service d'Explorations Fonctionnelles, and
| | - Mordi Muorah
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Lucie Yammine
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Martine Burtin
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Clément Nguyen
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Willian Baron
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Serge Garbay
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Amandine Viau
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Mélanie Broueilh
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Thomas Blanc
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Dorien Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Valeria Poli
- Department of Biotechnology and Health Sciences, Molecular Biotechnology Center, Torino University, Turin, Italy
| | - Dany Anglicheau
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling.,Service de Néphrologie-Transplantation, Hôpital Necker Enfants Malades, Paris, France
| | - Gérard Friedlander
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling.,Service d'Explorations Fonctionnelles, and
| | - Marco Pontoglio
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Morgan Gallazzini
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling
| | - Fabiola Terzi
- Institut National de la Santé et de la Recherche Médicale U1151, Université Paris Descartes, Institut Necker Enfants Malades, Department of Growth and Signaling,
| |
Collapse
|
14
|
Wang H, Su X, Yang M, Chen T, Hou J, Li N, Cao X. Reciprocal control of miR-197 and IL-6/STAT3 pathway reveals miR-197 as potential therapeutic target for hepatocellular carcinoma. Oncoimmunology 2015; 4:e1031440. [PMID: 26451302 DOI: 10.1080/2162402x.2015.1031440] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 02/07/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of the key players in liver cancer. Increased levels of phosphorylated STAT3 (p-STAT3) have been detected in many cancers including hepatocellular carcinoma (HCC), and are usually associated with a more aggressive phenotype and poor prognosis. In addition to aberrant activation of STAT3, upregulation of total STAT3 was also detected in HCC, for which the underlying mechanisms and significance remain to be fully elucidated. Here we report that a reciprocal regulation exists between miR-197 and the IL-6/STAT3 inflammatory signaling pathway in HCC. We found that IL-6 stimulation increased total STAT3 expression at protein level but not mRNA level in HCC cells, suggesting the existence of post-transcriptional regulation of STAT3. Our study showed that IL-6/STAT3 pathway decreases expression of miR-197 in HCC, which amplifies IL-6/STAT3 pathway and contributes to HCC progression. miR-197 can significantly inhibit HCC growth both in vitro and in vivo. In addition, IL-6/STAT3-induced downregulation of miR-197 in HCC may be via affecting Drosha binding to primary miR-197 (pri-miR-197) and thus reducing mature miR-197 generation. Our study suggests that miR-197 may serve as a potential therapeutic target for interfering with the IL-6/STAT3 inflammatory pathway in HCC.
Collapse
Affiliation(s)
- Huamin Wang
- Institute of Immunology; Zhejiang University School of Medicine ; Hangzhou, China
| | - Xiaoping Su
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Mingjin Yang
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China ; National Key Laboratory of Medical Molecular Biology & Department of Immunology; Chinese Academy of Medical Sciences; Institute of Basic Medical Sciences ; Beijing, China
| | - Taoyong Chen
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Jin Hou
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Nan Li
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Xuetao Cao
- Institute of Immunology; Zhejiang University School of Medicine ; Hangzhou, China ; National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China ; National Key Laboratory of Medical Molecular Biology & Department of Immunology; Chinese Academy of Medical Sciences; Institute of Basic Medical Sciences ; Beijing, China
| |
Collapse
|
15
|
Schrama D, Ugurel S, Sucker A, Ritter C, Zapatka M, Schadendorf D, Becker JC. STAT3 Single Nucleotide Polymorphism rs4796793 SNP Does Not Correlate with Response to Adjuvant IFNα Therapy in Stage III Melanoma Patients. Front Med (Lausanne) 2015; 1:47. [PMID: 25593920 PMCID: PMC4292185 DOI: 10.3389/fmed.2014.00047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/12/2014] [Indexed: 11/13/2022] Open
Abstract
Interferon alpha (IFNα) is approved for adjuvant treatment of stage III melanoma in Europe and the US. Its clinical efficacy, however, is restricted to a subpopulation of patients while side effects occur in most of treated patients. Thus, the identification of predictive biomarkers would be highly beneficial to improve the benefit to risk ratio. In this regard, STAT3 is important for signaling of the IFNα receptor. Moreover, the STAT3 single-nucleotide polymorphism (SNP) rs4796793 has recently been reported to be associated with IFNα sensitivity in metastatic renal cell carcinoma. To translate this notion to melanoma, we scrutinized the impact of rs4796793 functionally and clinically in this cancer. Interestingly, melanoma cells carrying the minor allele of rs4796793 were the most sensitive to IFNα in vitro. However, we did not detect a correlation between SNP genotype and STAT3 mRNA expression for either melanoma cells or for peripheral blood lymphocytes. Next, we analyzed the impact of rs4796793 on the clinical outcome of 259 stage III melanoma patients of which one-third had received adjuvant IFNα treatment. These analyses did not reveal a significant association between the STAT3 rs4796793 SNP and patients’ progression free or overall survival when IFNα treated and untreated patients were compared. In conclusion, STAT3 rs4796793 SNP is no predictive marker for the efficacy of adjuvant IFNα treatment in melanoma patients.
Collapse
Affiliation(s)
- David Schrama
- Department of Dermatology, University Hospital Würzburg , Würzburg , Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Würzburg , Würzburg , Germany ; Department of Dermatology, University Hospital Essen , Essen , Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen , Essen , Germany
| | - Cathrin Ritter
- Department of Dermatology, Medical University of Graz , Graz , Austria ; Department of Translational Skin Cancer Research, German Cancer Research Center , Heidelberg , Germany
| | - Marc Zapatka
- Department of Molecular Genetics, German Cancer Research Center , Heidelberg , Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen , Essen , Germany
| | - Jürgen Christian Becker
- Department of Dermatology, University Hospital Essen , Essen , Germany ; Department of Dermatology, Medical University of Graz , Graz , Austria ; Department of Translational Skin Cancer Research, German Cancer Research Center , Heidelberg , Germany
| |
Collapse
|
16
|
Snyder M, Huang J, Huang XY, Zhang JJ. A signal transducer and activator of transcription 3·Nuclear Factor κB (Stat3·NFκB) complex is necessary for the expression of fascin in metastatic breast cancer cells in response to interleukin (IL)-6 and tumor necrosis factor (TNF)-α. J Biol Chem 2014; 289:30082-9. [PMID: 25213863 DOI: 10.1074/jbc.m114.591719] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
IL-6 mediated activation of Stat3 is a major signaling pathway in the process of breast cancer metastasis. One important mechanism by which the IL-6/Stat3 pathway promotes metastasis is through transcriptional regulation of the actin-bundling protein fascin. In this study, we further analyzed the transcriptional regulation of the fascin gene promoter. We show that in addition to IL-6, TNF-α increases Stat3 and NFκB binding to the fascin promoter to induce its expression. We also show that NFκB is required for Stat3 recruitment to the fascin promoter in response to IL-6. Furthermore, Stat3 and NFκB form a protein complex in response to cytokine stimulation. Finally, we demonstrate that an overlapping STAT/NFκB site in a highly conserved 160-bp region of the fascin promoter is sufficient and necessary to induce transcription in response to IL-6 and TNF-α.
Collapse
Affiliation(s)
- Marylynn Snyder
- From the Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, New York 10065
| | - Jianyun Huang
- From the Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, New York 10065
| | - Xin-Yun Huang
- From the Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, New York 10065
| | - J Jillian Zhang
- From the Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, New York 10065
| |
Collapse
|
17
|
Abstract
All seven STAT proteins are expressed in the heart, and in this review we will focus on their contribution to cardiac physiology and to ischemic heart disease and its consequences. A substantial literature has focused on the roles of STAT1 and STAT3 in ischemic heart disease, where, at least in the acute phase, they appear to have a yin-yang relationship. STAT1 contributes to the loss of irreplaceable cardiac myocytes both by increasing apoptosis and by reducing cardioprotective autophagy. In contrast, STAT3 is cardioprotective, since STAT3-deficient mice have larger infarcts following ischemic injury, and a number of cardioprotective agents have been shown to act, at least partly, through STAT3 activation. STAT3 is also absolutely required for preconditioning—a process where periods of brief ischemia protect against a subsequent or previous prolonged ischemic episode. Prolonged activation of STAT3, however, is strongly implicated in the post-infarction remodeling of the heart which leads to heart failure, where, possibly together with STAT5, it augments activation of the renin-angiotensin system.
Collapse
Affiliation(s)
- Richard A Knight
- Medical Molecular Biology Unit; University College London; London, UK
| | | | | |
Collapse
|
18
|
Therapeutic Implications of Black Seed and Its Constituent Thymoquinone in the Prevention of Cancer through Inactivation and Activation of Molecular Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:724658. [PMID: 24959190 PMCID: PMC4052177 DOI: 10.1155/2014/724658] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/26/2014] [Accepted: 04/16/2014] [Indexed: 01/08/2023]
Abstract
The cancer is probably the most dreaded disease in both men and women and also major health problem worldwide. Despite its high prevalence, the exact molecular mechanisms of the development and progression are not fully understood. The current chemotherapy/radiotherapy regime used to treat cancer shows adverse side effect and may alter gene functions. Natural products are generally safe, effective, and less expensive substitutes of anticancer chemotherapeutics. Based on previous studies of their potential therapeutic uses, Nigella sativa and its constituents may be proved as good therapeutic options in the prevention of cancer. Black seeds are used as staple food in the Middle Eastern Countries for thousands of years and also in the treatment of diseases. Earlier studies have shown that N. sativa and its constituent thymoquinone (TQ) have important roles in the prevention and treatment of cancer by modulating cell signaling pathways. In this review, we summarize the role of N. sativa and its constituents TQ in the prevention of cancer through the activation or inactivation of molecular cell signaling pathways.
Collapse
|
19
|
Cyanidin-3-glucoside suppresses cytokine-induced inflammatory response in human intestinal cells: comparison with 5-aminosalicylic acid. PLoS One 2013; 8:e73001. [PMID: 24039842 PMCID: PMC3765207 DOI: 10.1371/journal.pone.0073001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
The potential use of polyphenols in the prevention and treatment of chronic inflammatory diseases has been extensively investigated although the mechanisms involved in cellular signaling need to be further elucidated. Cyanidin-3-glucoside is a typical anthocyanin of many pigmented fruits and vegetables widespread in the human diet. In the present study, the protection afforded by cyanidin-3-glucoside against cytokine-triggered inflammatory response was evaluated in the human intestinal HT-29 cell line, in comparison with 5-aminosalicylic acid, a well-established anti-inflammatory drug, used in inflammatory bowel disease. For this purpose, some key inflammatory mediators and inflammatory enzymes were examined. Our data showed that cyanidin-3-glucoside reduced cytokine-induced inflammation in intestinal cells, in terms of NO, PGE2 and IL-8 production and of iNOS and COX-2 expressions, at a much lower concentration than 5-aminosalicylic acid, suggesting a higher anti-inflammatory efficiency. Interestingly, cyanidin-3-glucoside and 5-aminosalicylic acid neither prevented IkB-α degradation nor the activation of NF-kB, but significantly reduced cytokine-induced levels of activated STAT1 accumulated in the cell nucleus. In addition, we established that phosphorylated p38 MAPK was not involved in the protective effect of cyanidin-3-glucoside or 5-aminosalicylic acid. Taking into account the high concentrations of dietary anthocyanins potentially reached in the gastrointestinal tract, cyanidin-3-glucoside may be envisaged as a promising nutraceutical giving complementary benefits in the context of inflammatory bowel disease.
Collapse
|
20
|
STAT5b as molecular target in pancreatic cancer--inhibition of tumor growth, angiogenesis, and metastases. Neoplasia 2013; 14:915-25. [PMID: 23097626 DOI: 10.1593/neo.12878] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/11/2012] [Accepted: 09/13/2012] [Indexed: 12/30/2022] Open
Abstract
The prognosis of patients suffering from pancreatic cancer is still poor and novel therapeutic options are urgently needed. Recently, the transcription factor signal transducer and activator of transcription 5b (STAT5b) was associated with tumor progression in human solid cancer. Hence, we assessed whether STAT5b might serve as an anticancer target in ductal pancreatic adenocarcinoma (DPAC). We found that nuclear expression of STAT5b can be detected in approximately 50% of DPAC. Blockade of STAT5b by stable shRNA-mediated knockdown showed no effects on tumor cell growth in vitro. However, inhibition of tumor cell motility was found even in response to stimulation with epidermal growth factor or interleukin-6. These findings were paralleled by a reduction of prometastatic and proangiogenic factors in vitro. Subsequent in vivo experiments revealed a strong growth inhibition on STAT5b blockade in subcutaneous and orthotopic models. These findings were paralleled by impaired tumor angiogenesis in vivo. In contrast to the subcutaneous model, the orthotopic model revealed a strong reduction of tumor cell proliferation that emphasizes the meaning of assessing targets in an appropriate microenvironment. Taken together, our results suggest that STAT5b might be a potential novel target for human DPAC.
Collapse
|
21
|
Current World Literature. Curr Opin Oncol 2013; 25:99-104. [DOI: 10.1097/cco.0b013e32835c1381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Shchelkunova A, Ermolinsky B, Boyle M, Mendez I, Lehker M, Martirosyan KS, Kazansky AV. Tuning of alternative splicing--switch from proto-oncogene to tumor suppressor. Int J Biol Sci 2012; 9:45-54. [PMID: 23289016 PMCID: PMC3535533 DOI: 10.7150/ijbs.5194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 12/07/2012] [Indexed: 12/21/2022] Open
Abstract
STAT5B, a specific member of the STAT family, is intimately associated with prostate tumor progression. While the full form of STAT5B is thought to promote tumor progression, a naturally occurring truncated isoform acts as a tumor suppressor. We previously demonstrated that truncated STAT5 is generated by insertion of an alternatively spliced exon and results in the introduction of an early termination codon. Present approaches targeting STAT proteins based on inhibition of functional domains of STAT's, such as DNA-binding, cooperative binding (protein-protein interaction), dimerization and phosphorylation will halt the action of the entire gene, both the proto-oncogenic and tumor suppressor functions of Stat5B. In this report we develop a new approach aimed at inhibiting the expression of full-length STAT5B (a proto-oncogene) while simultaneously enhancing the expression of STAT5∆B (a tumor suppressor). We have demonstrated the feasibility of using steric-blocking splice-switching oligonucleotides (SSOs) with a complimentary sequence to the targeted exon-intron boundary to enhance alternative intron/exon retention (up to 10%). The functional effect of the intron/exon proportional tuning was validated by cell proliferation and clonogenic assays. The new scheme applies specific steric-blocking splice-switching oligonucleotides and opens an opportunity for anti-tumor treatment as well as for the alteration of functional abilities of other STAT proteins.
Collapse
Affiliation(s)
- Aleksandra Shchelkunova
- Department of Biomedicine, The University of Texas at Brownsville, Brownsville, TX 78520, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
NPM-ALK: The Prototypic Member of a Family of Oncogenic Fusion Tyrosine Kinases. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:123253. [PMID: 22852078 PMCID: PMC3407651 DOI: 10.1155/2012/123253] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 04/28/2012] [Indexed: 02/07/2023]
Abstract
Anaplastic lymphoma kinase (ALK) was first identified in 1994 with the discovery that the gene encoding for this kinase was involved in the t(2;5)(p23;q35) chromosomal translocation observed in a subset of anaplastic large cell lymphoma (ALCL). The NPM-ALK fusion protein generated by this translocation is a constitutively active tyrosine kinase, and much research has focused on characterizing the signalling pathways and cellular activities this oncoprotein regulates in ALCL. We now know about the existence of nearly 20 distinct ALK translocation partners, and the fusion proteins resulting from these translocations play a critical role in the pathogenesis of a variety of cancers including subsets of large B-cell lymphomas, nonsmall cell lung carcinomas, and inflammatory myofibroblastic tumours. Moreover, the inhibition of ALK has been shown to be an effective treatment strategy in some of these malignancies. In this paper we will highlight malignancies where ALK translocations have been identified and discuss why ALK fusion proteins are constitutively active tyrosine kinases. Finally, using ALCL as an example, we will examine three key signalling pathways activated by NPM-ALK that contribute to proliferation and survival in ALCL.
Collapse
|
24
|
Qu Z, Fu J, Ma H, Zhou J, Jin M, Mapara MY, Grusby MJ, Xiao G. PDLIM2 restricts Th1 and Th17 differentiation and prevents autoimmune disease. Cell Biosci 2012; 2:23. [PMID: 22731402 PMCID: PMC3543335 DOI: 10.1186/2045-3701-2-23] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/06/2012] [Indexed: 01/31/2023] Open
Abstract
Background PDLIM2 is essential for the termination of the inflammatory transcription factors NF-κB and STAT but is dispensable for the development of immune cells and immune tissues/organs. Currently, it remains unknown whether and how PDLIM2 is involved in physiologic and pathogenic processes. Results Here we report that naive PDLIM2 deficient CD4+ T cells were prone to differentiate into Th1 and Th17 cells. PDLIM2 deficiency, however, had no obvious effect on lineage commitment towards Th2 or Treg cells. Notably, PDLIM2 deficient mice exhibited increased susceptibility to experimental autoimmune encephalitis (EAE), a Th1 and/or Th17 cell-mediated inflammatory disease model of multiple sclerosis (MS). Mechanistic studies further indicate that PDLIM2 was required for restricting expression of Th1 and Th17 cytokines, which was in accordance with the role of PDLIM2 in the termination of NF-κB and STAT activation. Conclusion These findings suggest that PDLIM2 is a key modulator of T-cell-mediated immune responses that may be targeted for the therapy of human autoimmune diseases.
Collapse
Affiliation(s)
- Zhaoxia Qu
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Marzano V, Santini S, Rossi C, Zucchelli M, D'Alessandro A, Marchetti C, Mingardi M, Stagni V, Barilà D, Urbani A. Proteomic profiling of ATM kinase proficient and deficient cell lines upon blockage of proteasome activity. J Proteomics 2012; 75:4632-46. [PMID: 22641158 PMCID: PMC3426930 DOI: 10.1016/j.jprot.2012.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 04/27/2012] [Accepted: 05/16/2012] [Indexed: 11/24/2022]
Abstract
Ataxia Telangiectasia Mutated (ATM) protein kinase is a key effector in the modulation of the functionality of some important stress responses, including DNA damage and oxidative stress response, and its deficiency is the hallmark of Ataxia Telangiectasia (A-T), a rare genetic disorder. ATM modulates the activity of hundreds of target proteins, essential for the correct balance between proliferation and cell death. The aim of this study is to evaluate the phenotypic adaptation at the protein level both in basal condition and in presence of proteasome blockage in order to identify the molecules whose level and stability are modulated through ATM expression. We pursued a comparative analysis of ATM deficient and proficient lymphoblastoid cells by label-free shotgun proteomic experiments comparing the panel of proteins differentially expressed. Through a non-supervised comparative bioinformatic analysis these data provided an insight on the functional role of ATM deficiency in cellular carbohydrate metabolism's regulation. This hypothesis has been demonstrated by targeted metabolic fingerprint analysis SRM (Selected Reaction Monitoring) on specific thermodynamic checkpoints of glycolysis. This article is part of a Special Issue entitled: Translational Proteomics.
Collapse
Affiliation(s)
- Valeria Marzano
- Proteomic and Metabonomic Laboratory, Fondazione Santa Lucia, 00143 Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Since its discovery two decades ago, the activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway by numerous cytokines and growth factors has resulted in it becoming one of the most well-studied intracellular signalling networks. The field has progressed from the identification of the individual components to high-resolution crystal structures of both JAK and STAT, and an understanding of the complexities of the molecular activation and deactivation cycle which results in a diverse, yet highly specific and regulated pattern of transcriptional responses. While there is still more to learn, we now appreciate how disruption and deregulation of this pathway can result in clinical disease and look forward to adoption of the next generation of JAK inhibitors in routine clinical treatment.
Collapse
Affiliation(s)
- Hiu Kiu
- Walter & Eliza Hall Institute, 1G Royal Parade, Parkville 3052, Australia
| | | |
Collapse
|
27
|
Souissi I, Ladam P, Cognet JAH, Le Coquil S, Varin-Blank N, Baran-Marszak F, Metelev V, Fagard R. A STAT3-inhibitory hairpin decoy oligodeoxynucleotide discriminates between STAT1 and STAT3 and induces death in a human colon carcinoma cell line. Mol Cancer 2012; 11:12. [PMID: 22423663 PMCID: PMC3325846 DOI: 10.1186/1476-4598-11-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 03/16/2012] [Indexed: 01/09/2023] Open
Abstract
Background The Signal Transducer and Activator of Transcription 3 (STAT3) is activated in tumor cells, and STAT3-inhibitors are able to induce the death of those cells. Decoy oligodeoxynucleotides (dODNs), which bind to the DNA Binding Domain (DBD) of STAT3, are efficient inhibitors. However, they also inhibit STAT1, whose activity is essential not only to resistance to pathogens, but also to cell growth inhibition and programmed cell death processes. The aim of this study was to design STAT3-specific dODNs which do not affect STAT1-mediated processes. Results New dODNs with a hairpin (hpdODNs) were designed. Modifications were introduced, based on the comparison of STAT3- and STAT1-DBD interactions with DNA using 3D structural analyses. The designed hpdODNs were tested for their ability to inhibit STAT3 but not STAT1 by determining: i) cell death in the active STAT3-dependent SW480 colon carcinoma cell line, ii) absence of inhibition of interferon (IFN) γ-dependent cell death, iii) expression of STAT1 targets, and iv) nuclear location of STAT3 and STAT1. One hpdODN was found to efficiently induce the death of SW480 cells without interfering with IFNγ-activated STAT1. This hpdODN was found in a complex with STAT3 but not with STAT1 using an original in-cell pull-down assay; this hpdODN also did not inhibit IFNγ-induced STAT1 phosphorylation, nor did it inhibit the expression of the STAT1-target IRF1. Furthermore, it prevented the nuclear transfer of STAT3 but not that of IFNγ-activated STAT1. Conclusions Comparative analyses at the atomic level revealed slight differences in STAT3 and STAT1 DBDs' interaction with their DNA target. These were sufficient to design a new discriminating hpdODN that inhibits STAT3 and not STAT1, thereby inducing tumor cell death without interfering with STAT1-dependent processes. Preferential interaction with STAT3 depends on oligodeoxynucleotide sequence modifications but might also result from DNA shape changes, known to modulate protein/DNA interactions. The finding of a STAT3-specific hpdODN establishes the first rational basis for designing STAT3 DBD-specific inhibitors.
Collapse
|
28
|
Snyder M, Huang XY, Zhang JJ. Signal transducers and activators of transcription 3 (STAT3) directly regulates cytokine-induced fascin expression and is required for breast cancer cell migration. J Biol Chem 2011; 286:38886-93. [PMID: 21937440 DOI: 10.1074/jbc.m111.286245] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The cytokines oncostatin M (OSM) and IL-6 promote breast cancer cell migration and metastasis. Both cytokines activate STAT3, a member of the STAT (signal transducers and activators of transcription) family of transcription factors. Through transcriptional regulation of its target genes, STAT3 controls a wide range of cellular processes, including cellular proliferation, oncogenesis, and cancer metastasis. Fascin is an actin-bundling protein involved in cell migration. Elevated levels of fascin expression are found in many metastatic cancers, and inhibition of fascin function by small chemical compounds leads to a block of tumor metastasis. In this work, we demonstrate that fascin is a direct STAT3 target gene in response to OSM and IL-6 in both mouse and human breast cancer cells. We show that NFκB also binds to the fascin promoter in response to cytokine treatment and this binding is STAT3-dependent. Both STAT3 and NFκB are required for the cytokine-induced expression of fascin in cancer cells. Furthermore, we demonstrate that STAT3, in directly controlling fascin expression, is both necessary and sufficient for breast cancer cell migration.
Collapse
Affiliation(s)
- Marylynn Snyder
- Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, New York 10065, USA
| | | | | |
Collapse
|