101
|
RNA-seq Reveals Dysregulation of Novel Melanocyte Genes upon Oxidative Stress: Implications in Vitiligo Pathogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2841814. [PMID: 31871544 PMCID: PMC6913168 DOI: 10.1155/2019/2841814] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/01/2019] [Indexed: 01/01/2023]
Abstract
Oxidative stress is known to induce melanocyte death, but the underlying mechanisms are incompletely understood. To identify oxidative stress-induced global gene expression changes in melanocytes, we treated PIG1 melanocytes with H2O2 in a dose- and time-dependent manner and performed RNA-seq. This approach allowed us to capture the events occurring early as well as late phase after treatment with H2O2. Our bioinformatics analysis identified differentially expressed genes involved in various biological processes of melanocytes which are known to contribute to the vitiligo development, such as apoptosis, autophagy, cell cycle regulation, cell adhesion, immune and inflammatory responses, melanocyte pluripotency, and developmental signaling such as WNT and NOTCH pathways. We uncovered several novel genes that are not previously described to be involved in melanocytic response to stress nor in vitiligo pathogenesis. Quantitative PCR and western blot analysis of selected proteins, performed on PIG1 and primary human epidermal melanocytes, confirmed the RNA-seq data. Interestingly, we discovered an aberrant regulation of several transcription factors that are involved in diabetes, neurological, and psychiatric diseases, all of which are comorbid conditions in patients with vitiligo. Our results may lead to a better understanding of the molecular mechanisms underlying vitiligo pathogenesis and help developing new drug targets for effective treatment.
Collapse
|
102
|
Teymouri S, Rakhshandeh H, Baghdar HN, Yousefi M, Salari R. Analgesic Herbal Medicines in the Treatment of Knee Osteoarthritis: A Systematic Review. Curr Rheumatol Rev 2019; 15:290-303. [DOI: 10.2174/1573397115666190328150203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 11/22/2018] [Accepted: 03/20/2019] [Indexed: 02/07/2023]
Abstract
Background:Osteoarthritis (OA) is a type of progressive rheumatoid disease, which leads to the degeneration of the articular cartilage, synovium, subchondral bone, tendons, and the surrounding ligaments.There are various treatments for knee OA, including pharmaceutical, nonpharmaceutical, and surgical treatments. Considering the chronic nature of the disease as well as the necessity for the long-term use of chemical medications, various side effects could occur that include gastrointestinal bleeding, hypertension, congestive heart failure, hyperkalemia, and kidney failure. Therefore, suitable treatments with fewer side effects should be recommended. Recent investigations suggest increased tendency in people to use Complementary and Alternative Medicine (CAM) for knee OA treatment.Objective:This systematic review aimed to assess the effectiveness and safety of herbal preparations for the treatment of OA.Methods:The searched databases were Cochrane, Scopus, and PubMed. All the selected papers pertained to randomized controlled trials until August 8, 2017 in English in which one or several specific herbs had been used in knee OA treatment.Results:We included 24 randomized trials (involving 2399 women and men). There were several different herbal medicines used within the included trials.Conclusion:The results show that the methods used in these trials may reduce symptoms and the extent of NSAID consumption and enhance the quality of life. Additional trials are suggested to investigate the safety and efficacy of herbs for the treatment of patients with OA.
Collapse
Affiliation(s)
- Soudeh Teymouri
- Ph.D Candidate of Persian Medicine, Student Research Committee, School of Persian and Complementary Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hasan Rakhshandeh
- Pharm.D, Assistant Professor, Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamideh Naghedi Baghdar
- Department of Persian Medicine, School of Persian and Complementary Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Yousefi
- Department of Persian Medicine, School of Persian and Complementary Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roshanak Salari
- Assistant Professor of Drug Control, Department of Pharmaceutical Sciences in Persian Medicine, School of Persian and Complementary Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
103
|
Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett 2019; 217:91-115. [PMID: 31747563 DOI: 10.1016/j.imlet.2019.11.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/01/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
Abstract
Chemokines are small molecules called "chemotactic cytokines" and regulate many processes like leukocyte trafficking, homing of immune cells, maturation, cytoskeletal rearrangement, physiology, migration during development, and host immune responses. These proteins bind to their corresponding 7-membrane G-protein-coupled receptors. Chemokines and their receptors are anti-inflammatory factors in autoimmune conditions, so consider as potential targets for neutralization in such diseases. They also express by cancer cells and function as angiogenic factors, and/or survival/growth factors that enhance tumor angiogenesis and development. Among chemokines, the CXCL12/CXCR4 axis has significantly been studied in numerous cancers and autoimmune diseases. CXCL12 is a homeostatic chemokine, which is acts as an anti-inflammatory chemokine during autoimmune inflammatory responses. In cancer cells, CXCL12 acts as an angiogenic, proliferative agent and regulates tumor cell apoptosis as well. CXCR4 has a role in leukocyte chemotaxis in inflammatory situations in numerous autoimmune diseases, as well as the high levels of CXCR4, observed in different types of human cancers. These findings suggest CXCL12/CXCR4 as a potential therapeutic target for therapy of autoimmune diseases and open a new approach to targeted-therapy of cancers by neutralizing CXCL12 and CXCR4. In this paper, we reviewed the current understanding of the role of the CXCL12/CXCR4 axis in disease pathology and cancer biology, and discuss its therapeutic implications in cancer and diseases.
Collapse
|
104
|
Fu Z, Fan Q, Zhou Y, Zhao Y, He Z. Elimination of Intracellular Calcium Overload by BAPTA-AM-Loaded Liposomes: A Promising Therapeutic Agent for Acute Liver Failure. ACS APPLIED MATERIALS & INTERFACES 2019; 11:39574-39585. [PMID: 31589019 DOI: 10.1021/acsami.9b13690] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In the past few decades, intracellular calcium overload has been shown to induce cell death through multiple signaling pathways. In this study, we used BAPTA-AM, a well-known membrane-permeable Ca2+ chelator, to prevent cell injury by allaying the intracellular calcium overload. We explored the clinical potentials of BAPTA-AM-loaded liposome (BAL) in the treatment of the acute liver failure (ALF) mouse model, which is characterized by severe hepatic necrosis and apoptosis. We discovered that BAL can significantly inhibit D-GalN-induced LO2 cell damage as it increased cell viability by 60% and downregulated the LPS-stimulated inflammatory response in RAW 264.7 macrophages by reversing the morphological change and modulating TNF-α and NF-κB expressions. Through systemic administration, BAL can rapidly accumulate in damaged liver tissue and exhibit excellent treatment effects on the D-GalN/LPS-induced ALF mouse model, including elevation of the survival rate (from 10 to 80%), recovery of normal liver indexes and liver health indicators, improvement of liver blood microcirculation (increased the blood flow volume by 80% and flow rate by 60%), and blood coagulation. The underlying hepatoprotective effect of BAL is presumably based on the antinecrosis and antiapoptosis abilities attributed to its inhibition on oxidative stress, restriction on TNF-α receptor, and mitochondria-mediated apoptotic pathway by effectively clearing the overloaded intercellular calcium. BAL holds great potential as a new therapeutic strategy for ALF treatment, and its prominent cell rescue ability provides ample opportunities for the treatment of many other diseases that are characterized by rapid and massive cell damage.
Collapse
Affiliation(s)
- Zailin Fu
- Department of Pharmacy , The First People's Hospital of Yuhang District , Hangzhou 310000 , P. R. China
- Department of Pharmacy , Zhejiang University of Technology , Hangzhou 310000 , P. R. China
| | - Qiaomei Fan
- Department of Pharmacy , The First Affiliated Hospital of Zhejiang Chinese Medical University , Hangzhou 310000 , P. R. China
- Department of Pharmacy , Zhejiang University of Technology , Hangzhou 310000 , P. R. China
| | - Yang Zhou
- Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , Maryland 21218 , United States
| | - Yi Zhao
- Wisconsin Institute for Discovery and Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , Wisconsin 53715 , United States
| | - Zhiyu He
- Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , Maryland 21218 , United States
| |
Collapse
|
105
|
Zhao LM, Zhang JH. Histone Deacetylase Inhibitors in Tumor Immunotherapy. Curr Med Chem 2019; 26:2990-3008. [PMID: 28762309 DOI: 10.2174/0929867324666170801102124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 04/26/2017] [Accepted: 06/27/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND With an increasing understanding of the antitumor immune response, considerable progress has been made in the field of tumor immunotherapy in the last decade. Inhibition of histone deacetylases represents a new strategy in tumor therapy and histone deacetylase inhibitors have been recently developed and validated as potential antitumor drugs. In addition to the direct antitumor effects, histone deacetylase inhibitors have been found to have the ability to improve tumor recognition by immune cells that may contribute to their antitumor activity. These immunomodolutory effects are desirable, and their in-depth comprehension will facilitate the design of novel regimens with improved clinical efficacy. OBJECTIVE Our goal here is to review recent developments in the application of histone deacetylase inhibitors as immune modulators in cancer treatment. METHODS Systemic compilation of the relevant literature in this field. RESULTS & CONCLUSION In this review, we summarize recent advances in the understanding of how histone deacetylase inhibitors alter immune process and discuss their effects on various cytokines. We also discuss the challenges to optimize the use of these inhibitors as immune modulators in cancer treatment. Information gained from this review will be valuable to this field and may be helpful for designing tumor immunotherapy trials involving histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Li-Ming Zhao
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Jie-Huan Zhang
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
106
|
Agarwal RG, Sharma P, Nyati KK. microRNAs in Mycobacterial Infection: Modulation of Host Immune Response and Apoptotic Pathways. Immune Netw 2019; 19:e30. [PMID: 31720041 PMCID: PMC6829074 DOI: 10.4110/in.2019.19.e30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/19/2019] [Accepted: 08/29/2019] [Indexed: 01/02/2023] Open
Abstract
Our current knowledge of mycobacterial infections in humans has progressively increased over the past few decades. The infection of Mycobacterium tuberculosis causes tuberculosis (TB) disease, which has reasoned for excessive morbidity and mortality worldwide, and has become a foremost issue of health problem globally. Mycobacterium leprae, another member of the family Mycobacteriaceae, is responsible for causing a chronic disease known as leprosy that mainly affects mucosa of the upper respiratory tract, skin, peripheral nerves, and eyes. Ample amount of existing data suggests that pathogenic mycobacteria have skilled in utilizing different mechanisms to escape or offset the host immune responses. They hijack the machinery of immune cells through the modulation of microRNAs (miRs), which regulate gene expression and immune responses of the host. Evidence shows that miRs have now gained considerable attention in the research, owing to their involvement in a broad range of inflammatory processes that are further implicated in the pathogenesis of several diseases. However, the knowledge of functions of miRs during mycobacterial infections remains limited. This review summarises recent findings of differential expression of miRs, which are used to good advantage by mycobacteria in offsetting host immune responses generated against them.
Collapse
Affiliation(s)
- Riddhi Girdhar Agarwal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Kishan Kumar Nyati
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| |
Collapse
|
107
|
Wang L, Sun J, Gao P, Su K, Wu H, Li J, Lou W. Wnt1-inducible signaling protein 1 regulates laryngeal squamous cell carcinoma glycolysis and chemoresistance via the YAP1/TEAD1/GLUT1 pathway. J Cell Physiol 2019; 234:15941-15950. [PMID: 30805937 DOI: 10.1002/jcp.28253] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/27/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Wnt1-inducible signaling protein 1 (WISP1) is a matricellular protein and downstream target of Wnt/β-catenin signaling. This study sought to determine the role of WISP1 in glucose metabolism and chemoresistance in laryngeal squamous cell carcinoma. WISP1 expression was silenced or upregulated in Hep-2 cells by the transfection of WISP1 siRNA or AdWISP1 vector. Ectopic WISP1 expression regulated glucose uptake and lactate production in Hep-2 cells. Subsequently, the expression of glucose transporter 1 (GLUT1) was significantly modulated by WISP1. Furthermore, WISP1 increased cell survival rates, diminished cell death rates, and suppressed ataxia-telangiectasia-mutated (ATM)-mediated DNA damage response pathway in cancer cells treated with cisplatin through GLUT1. WISP1 also promoted cancer cell tumorigenicity and growth in mice implanted with Hep-2 cells. Additionally, WISP1 activated the YAP1/TEAD1 pathway that consequently contributed to the regulation of GLUT1 expression. In summary, WISP1 regulated glucose metabolism and cisplatin resistance in laryngeal cancer by regulating GLUT1 expression. WISP1 may be used as a potential therapeutic target for laryngeal cancer.
Collapse
Affiliation(s)
- Liang Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Sun
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pei Gao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Su
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huanhuan Wu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junli Li
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weihua Lou
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
108
|
Miao L, Tao H, Peng Y, Wang S, Zhong Z, El-Seedi H, Dragan S, Zengin G, Cheang WS, Wang Y, Xiao J. The anti-inflammatory potential of Portulaca oleracea L. (purslane) extract by partial suppression on NF-κB and MAPK activation. Food Chem 2019; 290:239-245. [PMID: 31000042 DOI: 10.1016/j.foodchem.2019.04.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/02/2019] [Accepted: 04/01/2019] [Indexed: 02/05/2023]
Abstract
Portulaca oleracea L. (Purslane) has great potential as food and traditional drugs in several countries. The purpose of this study was to evaluate the anti-inflammatory effects of purslane extract on lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. Purslane extracts significantly reduced LPS-induced synthesis of NO in a dose-dependent manner, as well as the expression levels of iNOS and COX-2. The productions of TNF-α and IL-6 were also significantly reduced at the higher dose of 400 μg/ml. Meanwhile, the expression levels of P65, p-P65, p-MEK and p-IκB-α were inhibited dose-dependently. The nuclear translocation of P65 was partially prevented by the extract, which explained the inhibition of NF-κB pathway. In addition, three reported flavonoids, named luteolin, kaempferol and quercitrin, were identified in the extract, which might be responsible for its anti-inflammatory effects. Above all, our research has partially proved that purslane could be considered as a natural anti-inflammatory agent in further applications.
Collapse
Affiliation(s)
- Lingchao Miao
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Hongxun Tao
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Yu Peng
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Shengpeng Wang
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| | - Hesham El-Seedi
- Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, SE-75 123 Uppsala, Sweden.
| | - Simona Dragan
- University of Medicine and Pharmacy Victor Babes, 2 Eftimie Murgu Sq, 300041 Timisoara, Romania.
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey.
| | - Wai San Cheang
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| | - Yitao Wang
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| | - Jianbo Xiao
- State Key Laboratory of Quality Control in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
109
|
Abstract
Cancer cells overexpress several transcription factors and motor proteins, such as NFkB and kinesin, to accommodate their high energy demand as well as migratory needs via enhanced glycolysis. We hypothesize that high glucose drives cancer progression and cell aggressiveness by decreasing actin expression, increasing NFkB, and kinesin expressions, and by activating Epithelial Mesenchymal Transition (EMT). Using lowly metastatic MCF-7 and highly metastatic MDA-MB231 (MB231) breast cancer cells - highly incident cancer types - we establish how glucose metabolism regulates actin and the biochemical changes that lead to alterations of cell mechanical properties. We find that higher glucose (15 and 30 mM) increases glycolytic enzymes, glucose uptake, migration speed, kinesin, Ki-67, and NFkB expressions (biomarkers), and hybrid EMT phenotype activation (adhesion molecules/cadherins). Downregulation of actin, increased expressions of motor protein and NFkB, and decreased nuclear stiffness - induced by higher glucose - result in a significant increase in the migration speed. Moreover, glucose deprivation using the glucose analog 2-deoxyglucose decreases significantly the migration speed in both cancer cells. Thus, higher glucose promotes a more aggressive phenotype that promises to be a new target for cancer therapy and can help prevent cancer progression in diabetic patients by inhibiting glucose activated mechanisms.
Collapse
Affiliation(s)
- Julianna M Santos
- Mechanical Engineering Department, Texas Tech University, Lubbock, Texas, USA
| | - Fazle Hussain
- Mechanical Engineering Department, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
110
|
Ang RL, Chan M, Ting AT. Ripoptocide - A Spark for Inflammation. Front Cell Dev Biol 2019; 7:163. [PMID: 31457011 PMCID: PMC6700212 DOI: 10.3389/fcell.2019.00163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
The clinical success of biologics that inhibit TNF (Tumor Necrosis Factor) in inflammatory bowel diseases (IBD), psoriasis and rheumatoid arthritis (RA) has clearly established a pathogenic role for this cytokine in these inflammatory disorders. TNF binding to its receptors activates NFκB and MAPK signaling, inducing the expression of downstream pro-inflammatory genes. This is thought to be the primary mechanism by which TNF elicits inflammation. TNF is also a well-known trigger of caspase-dependent apoptosis or caspase-independent necroptosis. Whether cell death has any role in TNF-mediated inflammation has been less clear. Emerging data from animal models now suggest that cellular demise caused by TNF may indeed provoke inflammation. The default response of most cells to TNF stimulation is survival, rather than death, due to the presence of two sequential cell death checkpoints. The early checkpoint is transcription-independent involving the non-degradative ubiquitination of RIPK1 to prevent RIPK1 from becoming a death-signaling molecule. The later checkpoint requires the induction of pro-survival genes by NFκB-mediated transcription. When the early checkpoint is disrupted, RIPK1 initiates cell death and we suggest the term ripoptocide to describe this manner of death (encompassing both apoptosis and necroptosis). The sensitivity of a cell to ripoptocide is determined by the balance between regulatory molecules that enforce and those that disassemble the early checkpoint. As there is evidence suggesting that ripoptocide is inflammatory, individuals may develop inflammation due to ripoptocide brought about by genetic, epigenetic or post-translational alteration of these checkpoint regulators. For these individuals, drugs that reinforce the early checkpoint and inhibit ripoptocide could be useful in ameliorating inflammation.
Collapse
Affiliation(s)
- Rosalind L Ang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mark Chan
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,MSBS Program, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Adrian T Ting
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
111
|
The 3 Curcuminoid Analogs Comprising the Curcumin Extract Comparably Inhibit Nuclear Factor kappa-light-chain-enhancer Activation. PROGRESS IN PREVENTIVE MEDICINE 2019. [DOI: 10.1097/pp9.0000000000000023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
112
|
Zhao S, Liao T, Zhou T, Huang X, Xiang H, Chen J, Xu Z. Formyl-peptide receptor 2 suppresses proliferation, migration and invasion in human extravillous trophoblastic cells. Prostaglandins Other Lipid Mediat 2019; 143:106342. [DOI: 10.1016/j.prostaglandins.2019.106342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
|
113
|
Xie M, Zheng H, Madan-Lala R, Dai W, Gimbrone NT, Chen Z, Kinose F, Blackstone SA, Smalley KSM, Cress WD, Haura EB, Rix U, Beg AA. MEK Inhibition Modulates Cytokine Response to Mediate Therapeutic Efficacy in Lung Cancer. Cancer Res 2019; 79:5812-5825. [PMID: 31362929 DOI: 10.1158/0008-5472.can-19-0698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/19/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022]
Abstract
Activating mutations in BRAF, a key mediator of RAS signaling, are present in approximately 50% of melanoma patients. Pharmacologic inhibition of BRAF or the downstream MAP kinase MEK is highly effective in treating BRAF-mutant melanoma. In contrast, RAS pathway inhibitors have been less effective in treating epithelial malignancies, such as lung cancer. Here, we show that treatment of melanoma patients with BRAF and MEK inhibitors (MEKi) activated tumor NF-κB activity. MEKi potentiated the response to TNFα, a potent activator of NF-κB. In both melanoma and lung cancer cells, MEKi increased cell-surface expression of TNFα receptor 1 (TNFR1), which enhanced NF-κB activation and augmented expression of genes regulated by TNFα and IFNγ. Screening of 289 targeted agents for the ability to increase TNFα and IFNγ target gene expression demonstrated that this was a general activity of inhibitors of MEK and ERK kinases. Treatment with MEKi led to acquisition of a novel vulnerability to TNFα and IFNγ-induced apoptosis in lung cancer cells that were refractory to MEKi killing and augmented cell-cycle arrest. Abolishing the expression of TNFR1 on lung cancer cells impaired the antitumor efficacy of MEKi, whereas the administration of TNFα and IFNγ in MEKi-treated mice enhanced the antitumor response. Furthermore, immunotherapeutics known to induce expression of these cytokines synergized with MEKi in eradicating tumors. These findings define a novel cytokine response modulatory function of MEKi that can be therapeutically exploited. SIGNIFICANCE: Lung cancer cells are rendered sensitive to MEK inhibitors by TNFα and IFNγ, providing a strong mechanistic rationale for combining immunotherapeutics, such as checkpoint blockers, with MEK inhibitor therapy for lung cancer.See related commentary by Havel, p. 5699.
Collapse
Affiliation(s)
- Mengyu Xie
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Hong Zheng
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | | | - Wenjie Dai
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | - Nicholas T Gimbrone
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida.,Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Zhihua Chen
- Department of Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Fumi Kinose
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | | | - W Douglas Cress
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Uwe Rix
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida
| | - Amer A Beg
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. .,Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
114
|
Czauderna C, Castven D, Mahn FL, Marquardt JU. Context-Dependent Role of NF-κB Signaling in Primary Liver Cancer-from Tumor Development to Therapeutic Implications. Cancers (Basel) 2019; 11:cancers11081053. [PMID: 31349670 PMCID: PMC6721782 DOI: 10.3390/cancers11081053] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory cell death is a major risk factor for the development of diverse cancers including liver cancer. Herein, disruption of the hepatic microenvironment as well as the immune cell composition are major determinants of malignant transformation and progression in hepatocellular carcinomas (HCC). Considerable research efforts have focused on the identification of predisposing factors that promote induction of an oncogenic field effect within the inflammatory liver microenvironment. Among the most prominent factors involved in this so-called inflammation-fibrosis-cancer axis is the NF-κB pathway. The dominant role of this pathway for malignant transformation and progression in HCC is well documented. Pathway activation is significantly linked to poor prognostic traits as well as stemness characteristics, which places modulation of NF-κB signaling in the focus of therapeutic interventions. However, it is well recognized that the mechanistic importance of the pathway for HCC is highly context and cell type dependent. While constitutive pathway activation in an inflammatory etiological background can significantly promote HCC development and progression, absence of NF-κB signaling in differentiated liver cells also significantly enhances liver cancer development. Thus, therapeutic targeting of NF-κB as well as associated family members may not only exert beneficial effects but also negatively impact viability of healthy hepatocytes and/or cholangiocytes, respectively. The review presented here aims to decipher the complexity and paradoxical functions of NF-κB signaling in primary liver and non-parenchymal cells, as well as the induced molecular alterations that drive HCC development and progression with a particular focus on (immune-) therapeutic interventions.
Collapse
Affiliation(s)
- Carolin Czauderna
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Darko Castven
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Friederike L Mahn
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany.
| |
Collapse
|
115
|
Wu H, Zhuang Q, Xu J, Xu L, Zhao Y, Wang C, Yang Z, Shen F, Liu Z, Peng R. Cell-Penetrating Peptide Enhanced Antigen Presentation for Cancer Immunotherapy. Bioconjug Chem 2019; 30:2115-2126. [DOI: 10.1021/acs.bioconjchem.9b00245] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hanfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Qi Zhuang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Ligeng Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Yuhuan Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Chenya Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Zongjin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
116
|
Wang Y, Liu S, Li B, Jiang Y, Zhou X, Chen J, Li M, Ren B, Peng X, Zhou X, Cheng L. Staphylococcus aureus induces COX-2-dependent proliferation and malignant transformation in oral keratinocytes. J Oral Microbiol 2019; 11:1643205. [PMID: 31448061 PMCID: PMC6691923 DOI: 10.1080/20002297.2019.1643205] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/25/2019] [Accepted: 07/03/2019] [Indexed: 02/05/2023] Open
Abstract
The COX-2/PGE2 axis can play roles in mediating the progression of tumor. COX-2 induction was observed in oral cancer. In our previous study, we found Staphylococcus aureus, a pathogen prevalent in oral cancer, can activate the COX-2/PGE2 pathway in human oral keratinocyte (HOK) cells. Here, we investigated the proliferation of HOK cells affected by COX-2 induction and the role of COX-2 induction in the malignant transformation of HOK cells. We found S. aureus was able to facilitate HOK cell proliferation through upregulating COX-2 expression. With the induction of COX-2, expression of oral cancer-associated genes cyclin D1 was upregulated and p16 was downregulated. Transcriptome analysis showed that the “NF−kappa B signaling pathway” and “TNF signaling pathway” had the highest enrichment of differentially expressed genes (DEGs) with COX-2 over-expression. Seven upregulated genes (jun, tlr4, cxcl1, lif, cxcl3, tnfrsf1β, and il1β) in these two pathways were critical for the increased proliferation of HOK cells and might be associated with COX-2. Malignant transformation of cells was evaluated by soft agar colony formation assay and S. aureus infection promoted HOK cell colony formation. These results suggest the potential of S. aureus to induce the infection-associated malignant transformation of oral epitheliums through COX-2 activation.
Collapse
Affiliation(s)
- Yuxia Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Shiyu Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaling Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
117
|
Choi MC, Jo J, Park J, Kang HK, Park Y. NF-κB Signaling Pathways in Osteoarthritic Cartilage Destruction. Cells 2019; 8:cells8070734. [PMID: 31319599 PMCID: PMC6678954 DOI: 10.3390/cells8070734] [Citation(s) in RCA: 354] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a type of joint disease associated with wear and tear, inflammation, and aging. Mechanical stress along with synovial inflammation promotes the degradation of the extracellular matrix in the cartilage, leading to the breakdown of joint cartilage. The nuclear factor-kappaB (NF-κB) transcription factor has long been recognized as a disease-contributing factor and, thus, has become a therapeutic target for OA. Because NF-κB is a versatile and multi-functional transcription factor involved in various biological processes, a comprehensive understanding of the functions or regulation of NF-κB in the OA pathology will aid in the development of targeted therapeutic strategies to protect the cartilage from OA damage and reduce the risk of potential side-effects. In this review, we discuss the roles of NF-κB in OA chondrocytes and related signaling pathways, including recent findings, to better understand pathological cartilage remodeling and provide potential therapeutic targets that can interfere with NF-κB signaling for OA treatment.
Collapse
Affiliation(s)
- Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| | - Jiwon Jo
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea
| | - Jonggwan Park
- Department of Bioinformatics, Kongju National University, Kongju 38065, Korea
| | - Hee Kyoung Kang
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| |
Collapse
|
118
|
Kapur N, Mir H, Sonpavde GP, Jain S, Bae S, Lillard JW, Singh S. Prostate cancer cells hyper-activate CXCR6 signaling by cleaving CXCL16 to overcome effect of docetaxel. Cancer Lett 2019; 454:1-13. [PMID: 30974114 PMCID: PMC7748218 DOI: 10.1016/j.canlet.2019.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 10/27/2022]
Abstract
Molecular reprogramming in response to chemotherapeutics leads to poor therapeutic outcomes for prostate cancer (PCa). In this study, we demonstrated that CXCR6-CXCL16 axis promotes DTX resistance and acts as a counter-defense mechanism. After CXCR6 activation, cell death in response to DTX was inhibited, and blocking of CXCR6 potentiated DTX cytotoxicity. Moreover, in response to DTX, PCa cells expressed higher CXCR6, CXCL16, and ADAM-10. Furthermore, ADAM-10-mediated release of CXCL16 hyper-activated CXCR6 signaling in response to DTX. Activation of CXCR6 resulted in increased GSK-3β, NF-κB, ERK1/2 phosphorylation, and survivin expression, which reduce DTX response. Finally, treatment of PCa cells with anti-CXCR6 monoclonal antibody synergistically or additively induced cell death with ∼1.5-4.5 fold reduction in the effective concentration of DTX. In sum, our data imply that co-targeting of CXCR6 would lead to therapeutic enhancement of DTX, leading to better clinical outcomes for PCa patients.
Collapse
Affiliation(s)
- Neeraj Kapur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA; Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Hina Mir
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA; Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Guru P Sonpavde
- Department of Medical Oncology, Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Sanjay Jain
- Department of Medicine, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Sejong Bae
- Department of Medicine, Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - James W Lillard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA; Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Shailesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA; Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
119
|
Cao X, Ding L, Xie ZZ, Yang Y, Whiteman M, Moore PK, Bian JS. A Review of Hydrogen Sulfide Synthesis, Metabolism, and Measurement: Is Modulation of Hydrogen Sulfide a Novel Therapeutic for Cancer? Antioxid Redox Signal 2019; 31:1-38. [PMID: 29790379 PMCID: PMC6551999 DOI: 10.1089/ars.2017.7058] [Citation(s) in RCA: 290] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023]
Abstract
Significance: Hydrogen sulfide (H2S) has been recognized as the third gaseous transmitter alongside nitric oxide and carbon monoxide. In the past decade, numerous studies have demonstrated an active role of H2S in the context of cancer biology. Recent Advances: The three H2S-producing enzymes, namely cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3MST), have been found to be highly expressed in numerous types of cancer. Moreover, inhibition of CBS has shown anti-tumor activity, particularly in colon cancer, ovarian cancer, and breast cancer, whereas the consequence of CSE or 3MST inhibition remains largely unexplored in cancer cells. Intriguingly, H2S donation at high amounts or a long time duration has also been observed to induce cancer cell apoptosis in vitro and in vivo while sparing noncancerous fibroblast cells. Therefore, a bell-shaped model has been proposed to explain the role of H2S in cancer development. Specifically, endogenous H2S or a relatively low level of exogenous H2S may exhibit a pro-cancer effect, whereas exposure to H2S at a higher amount or for a long period may lead to cancer cell death. This indicates that inhibition of H2S biosynthesis and H2S supplementation serve as two distinct ways for cancer treatment. This paradoxical role of H2S has stimulated the enthusiasm for the development of novel CBS inhibitors, H2S donors, and H2S-releasing hybrids. Critical Issues: A clear relationship between H2S level and cancer progression remains lacking. The possibility that the altered levels of these byproducts have influenced the cell viability of cancer cells has not been excluded in previous studies when modulating H2S producing enzymes. Future Directions: The consequence of CSE or 3MST inhibition in cancer cells need to be examined in the future. Better portrayal of the crosstalk among these gaseous transmitters may not only lead to an in-depth understanding of cancer progression but also shed light on novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Xu Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lei Ding
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi-zhong Xie
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Yong Yang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, China
| | | | - Philip K. Moore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
120
|
Carpinus turczaninowii Extract May Alleviate High Glucose-Induced Arterial Damage and Inflammation. Antioxidants (Basel) 2019; 8:antiox8060172. [PMID: 31212679 PMCID: PMC6616550 DOI: 10.3390/antiox8060172] [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: 05/06/2019] [Revised: 06/02/2019] [Accepted: 06/07/2019] [Indexed: 01/06/2023] Open
Abstract
Hyperglycemia-induced oxidative stress triggers severe vascular damage and induces an inflammatory vascular state, and is, therefore, one of the main causes of atherosclerosis. Recently, interest in the natural compound Carpinus turczaninowii has increased because of its reported antioxidant and anti-inflammatory properties. We investigated whether a C. turczaninowii extract was capable of attenuating high glucose-induced inflammation and arterial damage using human aortic vascular smooth muscle cells (hASMCs). mRNA expression levels of proinflammatory response [interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α)], endoplasmic reticulum (ER) stress [CCAAT-enhancer-binding proteins (C/EBP) homologous protein (CHOP)], and adenosine monophosphate (AMP)-protein activated kinase α2 (AMPK α2)], and DNA damage [phosphorylated H2.AX (p-H2.AX)] were measured in hASMCs treated with the C. turczaninowii extracts (1 and 10 μg/mL) after being stimulated by high glucose (25 mM) or not. The C. turczaninowii extract attenuated the increased mRNA expression of IL-6, TNF-α, and CHOP in hASMCs under high glucose conditions. The expression levels of p-H2.AX and AMPK α2 induced by high glucose were also significantly decreased in response to treatment with the C. turczaninowii extract. In addition, 15 types of phenolic compounds including quercetin, myricitrin, and ellagic acid, which exhibit antioxidant and anti-inflammatory properties, were identified in the C. turczaninowii extract through ultra-performance liquid chromatography-quadrupole-time of flight (UPLC-Q-TOF) mass spectrometry. In conclusion, C. turczaninowii may alleviate high glucose-induced inflammation and arterial damage in hASMCs, and may have potential in the treatment of hyperglycemia-induced atherosclerosis.
Collapse
|
121
|
Ning Y, Wang X, Lammi MJ, Guo X. Changes in the NF-κB signaling pathway in juvenile and adult patients with Kashin-Beck disease. Exp Cell Res 2019; 379:140-149. [DOI: 10.1016/j.yexcr.2019.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 03/15/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022]
|
122
|
Aghajani M, Mansoori B, Mohammadi A, Asadzadeh Z, Baradaran B. New emerging roles of CD133 in cancer stem cell: Signaling pathway and miRNA regulation. J Cell Physiol 2019; 234:21642-21661. [PMID: 31102292 DOI: 10.1002/jcp.28824] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSC) are rare immortal cells within a tumor that are able to initiate tumor progression, development, and resistance. Advances studies show that, like normal stem cells, CSCs can be both self-renewed and given rise to many cell types, therefore form tumors. A number of cell surface markers, such as CD44, CD24, and CD133 are frequently used to identify CSCs. CD133, a transmembrane glycoprotein, either alone or in collaboration with other markers, has been mainly considered to identify CSCs from different solid tumors. However, the exactness of CD133 as a cancer stem cell biomarker has not been approved yet. The clinical importance of CD133 is as a CSC marker in many cancers. Also, it contributes to shorter survival, tumor progression, and tumor recurrence. The expression of CD133 is controlled by many extracellular or intracellular factors, such as tumor microenvironment, epigenetic factors, signaling pathways, and miRNAs. In this study, it was attempted to determine: 1) CD133 function; 2) the role of CD133 in cancer; 3) CD133 regulation; 4) the therapeutic role of CD133 in cancers.
Collapse
Affiliation(s)
- Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
123
|
Novosad J, Fiala Z, Borská L, Krejsek J. Immunosuppressive Effect of Polycyclic Aromatic Hydrocarbons by Induction of Apoptosis of pre-B Lymphocytes of Bone Marrow. ACTA MEDICA (HRADEC KRÁLOVÉ) 2019. [DOI: 10.14712/18059694.2019.68] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAH) are ubiquitous environmental pollutants, distinguished by genotoxic, hepatotoxic, nephrotoxic and immunotoxic effects. Especially secondary toxicity after bioactivation by microsomal monooxygenases (dependent on cytochromes P450) is characteristic of them. The immunotoxic effect is the result of very global impact on immunological reactivity of an organism and immunosuppression by induction of apoptosis of pre-B lymphocytes represents one of its particular forms. It has been proved that the effect of PAH is caused mostly by the following mechanisms: enzymatic induction by the way of activation of AhR (Aromatic hydrocarbon Receptor); alteration of cellular DNA; development of oxidative stress; increase in the concentration of intercellular calcium and decline of activity of NF-κB (Nuclear Factor – kappa B). Most sensitive to these changes are particularly B-lymphocytic precursors and pre-B lymphocytes. Intensity of entire manifestations is also considerably dependent on the presence and intensity of mechanisms of active or passive resistance of cells.
Collapse
|
124
|
Shi P, Su Y, Li R, Liang Z, Dong S, Huang J. PEDV nsp16 negatively regulates innate immunity to promote viral proliferation. Virus Res 2019; 265:57-66. [PMID: 30849413 PMCID: PMC7114654 DOI: 10.1016/j.virusres.2019.03.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 11/06/2022]
Abstract
Type-I IFNs (IFN-I) provide a key mediator of innate antiviral response during virus proliferation. Porcine epidemic diarrhea virus (PEDV), which causes diarrhea in swine of all ages, is a worldwide-distributed alphacoronavirus with economic importance. Here, we screened PEDV RNA modification enzymes involved in regulating antiviral response. Whereas the PEDV nsp13 barely regulates type I IFN, inflammatory cytokines (IL-6, TNF-a) and MHCII, nsp16 and nsp14 (to a lesser extent) down-regulate these antiviral effectors. Importantly, we found nsp16 KDKE tetrad appears to play a key role in interferon inhibition by mutating the D129 catalytic residue. Mechanistically, nsp16 down-regulates the activities of RIG-I and MDA5 mediated IFN-β and ISRE. In turn, the mRNA levels of IFIT family members (IFIT1, IFIT2, IFIT3) was inhibited in cells overexpressing nsp16. In addition, nsp10 enhanced the inhibitory effect of nsp16 on IFN-β. Altogether these results indicate PEDV nsp16 negatively regulates innate immunity to promote viral proliferation. Findings from this study provides novel perspective to advance the understanding in the pathogenesis of PEDV.
Collapse
Affiliation(s)
- Peidian Shi
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Yanxin Su
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Ruiqiao Li
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Zhixuan Liang
- Tianjin Center of Animal Disease Preventive and Control, Tianjin, China
| | | | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
125
|
Hassan SM, Taha AM, Eldahshan OA, Sayed AA, Salem AM. Modulatory effect of Prosopis juliflora leaves on hepatic fibrogenic and fibrolytic alterations induced in rats by thioacetamide. Biomed Pharmacother 2019; 115:108788. [PMID: 31035010 DOI: 10.1016/j.biopha.2019.108788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/23/2019] [Accepted: 03/13/2019] [Indexed: 01/15/2023] Open
Abstract
This study investigated the antifibrotic effect of Prosopis juliflora leaves crude methanolic extract (PJEL) against thioacetamide (TAA)-induced liver fibrosis. The phytochemical analysis of PJEL was performed via HPLC/MS in association with evaluating its free radical scavenging and cytotoxic activities. The antifibrotic activity of PJEL was assessed by dividing Wistar rats into 8 groups: normal control, PJEL1-administered rats (2 mg/ Kg b.w.), PJEL2-administered rats (4 mg/ Kg b.w.), PJEL3-administered rats (8 mg/Kg b.w.), TAA-induced hepatic fibrosis, TTA + PJEL1, TAA + PJEL2, and TAA + PJEL3. Results indicated that PJEL crude methanolic extract is rich in polyphenolic compounds and alkaloids. PJEL exerted free radical scavenging activity with IC50 of 123.5 μg/mL and cytotoxic activity against a well-differentiated hepatocellular cell line (IC50 = 11.1 μg/mL). PJEL at a dose of 4 mg/Kg b.w. ameliorated serum ALT activity and improved serum albumin level and hepatic hydroxyproline content in association with a reduction in the fibrosis stage. PJEL elevated hepatic tumor necrosis factor-α and interleukin-6 contents with less necrosis grade. PJEL post-therapy ameliorated the relative expression of Bcl-2, Col1A1, Mmp-9, and Mmp-2 genes in liver. CONCLUSION: PJEL possesses a good therapeutic activity against TAA-induced liver fibrosis via enhancing extracellular matrix removal and stimulating hepatic regeneration to decrease hepatic necrosis.
Collapse
Affiliation(s)
- Salah M Hassan
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - AlShaimaa M Taha
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Omayma A Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Ahmed A Sayed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt; Children's Cancer Hospital, 57357, Egypt
| | - Ahmed M Salem
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
126
|
Li H, Lu N, Yu X, Liu X, Hu P, Zhu Y, Shen L, Xu J, Li Z, Guo Q, Hui H. Oroxylin A, a natural compound, mitigates the negative effects of TNFα-treated acute myelogenous leukemia cells. Carcinogenesis 2019; 39:1292-1303. [PMID: 29346508 DOI: 10.1093/carcin/bgy004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 01/10/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a complicated cytokine which is involved in proliferation and differentiation of acute myelogenous leukemia (AML) cells through a poorly understood mechanism. Mechanistic studies indicate that TNFα induced binding of PI3K subunit p85α to N-terminal truncated nuclear receptor RXRα (tRXRα) proteins, and activated AKT. The activated PI3K/AKT pathway negatively regulated differentiation of AML cells through the upregulation of c-Myc. In addition, TNFα also induced activation of nuclear factor κB (NF-κB), a nuclear transcription factor which was shown to promote cell proliferation. The present study demonstrates that oroxylin A, a natural compound isolated from Scutellariae radix, sensitizes leukemia cells to TNFα and markedly enhances TNFα-induced growth inhibition and differentiation of AML cell including human leukemia cell lines and primary AML cells. Activation of PI3K/AKT pathway could be inhibited by oroxylin A through inhibiting expression of tRXRα in NB4 and HL-60-resistant cells. Furthermore, we found that oroxylin A inhibited the activation of NF-κB and the DNA binding activity by TNFα proved by EMSA in these two AML cell lines. Moreover, in vivo studies showed that treatment with oroxylin A in combination with TNFα decreased AML cell population and prolonged survival in NOD/SCID mice with xenografts of primary AML cells. Overall, our results indicate that oroxylin A is able to inhibit the negative effects of TNFα for AML therapy, suggesting that combination of oroxylin A and TNFα have the potential to delay growth or eliminate the abnormal leukemic cells, thus representing a promising strategy for AML treatment.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaoxuan Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiao Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Po Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yu Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China.,Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu Province, People's Republic of China
| | - Le Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jingyan Xu
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
127
|
Eser Ocak P, Ocak U, Tang J, Zhang JH. The role of caveolin-1 in tumors of the brain - functional and clinical implications. Cell Oncol (Dordr) 2019; 42:423-447. [PMID: 30993541 DOI: 10.1007/s13402-019-00447-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Caveolin-1 (cav-1) is the major structural protein of caveolae, the flask-shaped invaginations of the plasma membrane mainly involved in cell signaling. Today, cav-1 is believed to play a role in a variety of disease processes including cancer, owing to the variations of its expression in association with tumor progression, invasive behavior, metastasis and therapy resistance. Since first detected in the brain, a number of studies has particularly focused on the role of cav-1 in the various steps of brain tumorigenesis. In this review, we discuss the different roles of cav-1 and its contributions to the molecular mechanisms underlying the pathobiology and natural behavior of brain tumors including glial, non-glial and metastatic subtypes. These contributions could be attributed to its co-localization with important players in tumorigenesis within the lipid-enriched domains of the plasma membrane. In that regard, the ability of cav-1 to interact with various cell signaling molecules as well as the impact of caveolae depletion on important pathways acting in brain tumor pathogenesis are noteworthy. We also discuss conversant causes hampering the treatment of malignant glial tumors such as limited transport of chemotherapeutics across the blood tumor barrier and resistance to chemoradiotherapy, by focusing on the molecular fundamentals involving cav-1 participation. CONCLUSIONS Cav-1 has the potential to pivot the molecular basis underlying the pathobiology of brain tumors, particularly the malignant glial subtype. In addition, the regulatory effect of cav-1-dependent and caveola-mediated transcellular transport on the permeability of the blood tumor barrier could be of benefit to overcome the restricted transport across brain barriers when applying chemotherapeutics. The association of cav-1 with tumors of the brain other than malignant gliomas deserves to be underlined, as well given the evidence suggesting its potential in predicting tumor grade and recurrence rates together with determining patient prognosis in oligodendrogliomas, ependymomas, meningiomas, vestibular schwannomas and brain metastases.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
128
|
Cai Y, Li Y. Upregulation of miR-29b-3p protects cardiomyocytes from hypoxia-induced apoptosis by targeting TRAF5. Cell Mol Biol Lett 2019; 24:27. [PMID: 31011336 PMCID: PMC6460541 DOI: 10.1186/s11658-019-0151-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNAs (miRNAs) are pivotal regulators in regulating hypoxia-induced cardiomyocyte injury. This study was designed to evaluate the effects of miR-29b-3p on hypoxic cardiomyocytes. Methods Human AC16 cells were cultured under normoxic or hypoxic conditions. Hypoxic injury was confirmed based on alterations in cell viability using CCK-8 assay and apoptosis using flow cytometry and Hoechst staining. Bioinformatics analyses and the dual-luciferase reporter assay were performed to predict and validate the target gene of miR-29b-3p. Results We found that hypoxia suppressed cell viability and promoted apoptosis. TNF receptor-associated factor 5 (TRAF5) was a potential target gene of miR-29b-3p. Our in vitro experiments revealed that miR-29b-3p overexpression or TRAF6 knockdown significantly protected cardiomyocytes against hypoxia-induced injury. Moreover, knockdown of TRAF5 knockdown potentiated the protective effects of miR-29b-3p against hypoxia-induced cell injury. Conclusion These findings suggest that upregulation of miR-29b-3p could protect cardiomyocytes against hypoxia-induced injury through downregulation of TRAF5. Targeting TRAF5 with miR-29b-3p might be a potential therapeutic method for AMI.
Collapse
Affiliation(s)
- Yuhua Cai
- Department of Cardiovasology, Jingzhou First Municipal Hospital, Jingzhou, Hubei Province China
| | - Yunpeng Li
- 2Department of Cardiovasology, Dongfeng Hospital, Hubei University of Medicine, No. 16 Daling Road, Shiyan, 442008 Hubei Province China
| |
Collapse
|
129
|
Zhu D, Huang R, Fu P, Chen L, Luo L, Chu P, He L, Li Y, Liao L, Zhu Z, Wang Y. Investigating the Role of BATF3 in Grass Carp ( Ctenopharyngodon idella) Immune Modulation: A Fundamental Functional Analysis. Int J Mol Sci 2019; 20:ijms20071687. [PMID: 30987332 PMCID: PMC6479329 DOI: 10.3390/ijms20071687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Basic leucine zipper transcription factor ATF-like (BATF)-3, belonging to activator protein 1 (AP-1) superfamily transcription factors, is essential for homeostatic development of CD8α+ classical dendritic cells activating CD8 T-cell responses to intracellular pathogens. In this study, the characteristics and cDNA cloning of the CiBATF3 molecule were described in grass carp (Ctenopharyngodon idella). CiBATF3 had abundant expression in immune-related organizations, including liver, spleen and gill, and grass carp reovirus (GCRV) infection had significantly changed its expression level. After Ctenopharyngodon idella kidney (CIK) cells were challenged with pathogen-associated molecular patterns (PAMPs), polyinosinic:polycytidylic acid (poly(I:C)) stimulation induced higher mRNA levels of CiBATF3 than that of lipopolysaccharide (LPS). Subcellular localization showed that CiBATF3-GFP was entirely distributed throughout cells and nuclear translocation of CiBATF3 was found after poly(I:C) treatment. Additionally, the interaction between CiBATF3 and interleukin 10 (IL-10) was proven by bimolecular fluorescence complementation (BiFC) system. The small interfering RNA (siRNA)-mediated CiBATF3 silencing showed that the mRNA of CiBATF3 and its downstream genes were down-regulated in vitro and in vivo. CiBATF3 played a negative regulatory role in the transcriptional activities of AP-1 and NF-κB reporter gene. In summary, the results may provide valuable information on fundamental functional mechanisms of CiBATF3.
Collapse
Affiliation(s)
- Denghui Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Rong Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Peipei Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Liangming Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lifei Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Pengfei Chu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Libo He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Yongming Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Lanjie Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Zuoyan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Yaping Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
- Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
130
|
Regulation of microRNAs by molecular hydrogen contributes to the prevention of radiation-induced damage in the rat myocardium. Mol Cell Biochem 2019; 457:61-72. [PMID: 30830529 DOI: 10.1007/s11010-019-03512-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/23/2019] [Indexed: 10/27/2022]
Abstract
microRNAs (miRNAs) constitute a large class of post-transcriptional regulators of gene expression. It has been estimated that miRNAs regulate up to 30% of the protein-coding genes in humans. They are implicated in many physiological and pathological processes, including those involved in radiation-induced heart damage. Biomedical studies indicate that molecular hydrogen has potential as a radioprotective agent due to its antioxidant, anti-inflammatory, and signal-modulating effects. However, the impact of molecular hydrogen on the expression of miRNAs in the heart after irradiation has not been investigated. This study aimed to explore the involvement of miRNA-1, -15b, and -21 in the protective action of molecular hydrogen on rat myocardium damaged by irradiation. The results showed that the levels of malondialdehyde (MDA) and tumor necrosis factor alpha (TNF-α) increased in the rat myocardium after irradiation. Treatment with molecular hydrogen-rich water (HRW) reduced these values to the level of non-irradiated controls. miRNA-1 is known to be involved in cardiac hypertrophy, and was significantly decreased in the rat myocardium after irradiation. Application of HRW attenuated this decrease in all evaluated time periods. miRNA-15b is considered to be anti-fibrotic, anti-hypertrophic, and anti-oxidative. Irradiation downregulated miRNA-15b, whereas administration of HRW restored these values. miRNA-21 is connected with cardiac fibrosis. We observed significant increase in miRNA-21 expression in the irradiated rat hearts. Molecular hydrogen lowered myocardial miRNA-21 levels after irradiation. This study revealed for the first time that the protective effects of molecular hydrogen on irradiation-induced heart damage may be mediated by regulating miRNA-1, -15b, and -21.
Collapse
|
131
|
Kashem MA, Li H, Toledo NP, Omange RW, Liang B, Liu LR, Li L, Yang X, Yuan XY, Kindrachuk J, Plummer FA, Luo M. Toll-like Interleukin 1 Receptor Regulator Is an Important Modulator of Inflammation Responsive Genes. Front Immunol 2019; 10:272. [PMID: 30873160 PMCID: PMC6403165 DOI: 10.3389/fimmu.2019.00272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/31/2019] [Indexed: 01/30/2023] Open
Abstract
TILRR (Toll-like interleukin-1 receptor regulator), a transcript variant of FREM1, is a novel regulatory component, which stimulates innate immune responses through binding to IL-1R1 (Interleukin-1 receptor, type 1) and TLR (Toll-like receptor) complex. However, it is not known whether TILRR expression influences other genes in the NFκB signal transduction and pro-inflammatory responses. Our previous study identified FREM1 as a novel candidate gene in HIV-1 resistance/susceptibility in the Pumwani Sex worker cohort. In this study, we investigated the effect of TILRR overexpression on expression of genes in the NFκB signaling pathway in vitro. The effect of TILRR on mRNA expression of 84 genes related to NFκB signal transduction pathway was investigated by qRT-PCR. Overexpression of TILRR on pro-inflammatory cytokine/chemokine(s) secretion in cell culture supernatants was analyzed using Bioplex multiplex bead assay. We found that TILRR overexpression significantly influenced expression of many genes in HeLa and VK2/E6E7 cells. Several cytokine/chemokine(s), including IL-6, IL-8 (CXCL8), IP-10, MCP-1, MIP-1β, and RANTES (CCL5) were significantly increased in the cell culture supernatants following TILRR overexpression. Although how TILRR influences the expression of these genes needs to be further studied, we are the first to show the influence of TILRR on many genes in the NFκB inflammatory pathways. The NFκB inflammatory response pathways are extremely important in microbial infection and pathogenesis, including HIV-1 transmission. Further study of the role of TILRR may identify the novel intervention targets and strategies against HIV infection.
Collapse
Affiliation(s)
- Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Nikki Pauline Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Robert Were Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Binhua Liang
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Lewis Ruxi Liu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Lin Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Xuefen Yang
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Xin-Yong Yuan
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Jason Kindrachuk
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| |
Collapse
|
132
|
Ding W, Fan YY, Zhang C, Fu L, Chen X, Xu DX. Obeticholic acid differentially regulates hepatic injury and inflammation at different stages of D-galactosamine/lipopolysaccharide-evoked acute liver failure. Eur J Pharmacol 2019; 850:150-157. [PMID: 30772394 DOI: 10.1016/j.ejphar.2019.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/30/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
The farnesoid X receptor (FXR) is a ligand-activated transcription factor that regulates genes involved in bile acid metabolism. Accumulating data demonstrate that FXR has an anti-inflammatory activity. The present study aimed to investigate the effect of obeticholic acid (OCA), a novel synthetic FXR agonist, on D-galactosamine (GalN)/lipopolysaccharide (LPS)-evoked acute liver injury. All mice except controls were intraperitoneally injected with GalN (300 mg/kg) plus LPS (2.5 μg/kg). Some mice were pretreated with OCA (10 mg/kg) 48, 24 and 1 h before GalN/LPS. As expected, pretreatment with OCA alleviated hepatocyte apoptosis at early and middle stages of GalN/LPS-induced acute liver failure. By contrast, pretreatment with OCA augmented hepatic injury and inflammatory cell infiltration at middle stage of GalN/LPS-induced acute liver failure. Additional experiment found that OCA inhibited hepatic NF-κB activation at early and middle stages of GalN/LPS-induced acute liver failure. Interestingly, OCA inhibited hepatic proinflammatory cytokine tnf-α and il-6 but upregulated hepatic anti-inflammatory cytokine il-10 at early stage of GalN/LPS-induced acute liver failure. By contrast, OCA suppressed hepatic anti-inflammatory cytokine tgf-β and il-10 at middle stage of GalN/LPS-induced acute liver injury. These results suggest that FXR agonist OCA differentially regulates hepatic injury and inflammation at different stages of GalN/LPS-evoked acute liver failure.
Collapse
Affiliation(s)
- Wen Ding
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Yuan-Yuan Fan
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Lin Fu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Xi Chen
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China.
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
133
|
Yan B, Sun Y, Zeng J, Chen Y, Li C, Song P, Zhang L, Yang X, Wu Y, Ma P. Combined use of vitamin E and nimodipine ameliorates dibutyl phthalate-induced memory deficit and apoptosis in mice by inhibiting the ERK 1/2 pathway. Toxicol Appl Pharmacol 2019; 368:1-17. [PMID: 30776390 DOI: 10.1016/j.taap.2019.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
Learning disabilities (LDs) in children are a serious global problem. Dibutyl phthalate (DBP), a plasticizer widely used in daily life, has been linked to triggering childhood LDs, however the mechanism underlying this remains unclear. Studies have shown that the ERK 1/2 pathway is closely related to apoptosis of hippocampal neurons. On the basis of these links between LDs, DBP and the ERK 1/2 pathway, we explore whether DBP induces hippocampal neuron apoptosis and increases behavioral disorders in mice via the ERK 1/2 pathway. We looked at oxidative stress, examined the calcium signal, detected the ERK 1/2 pathway and evaluated apoptosis as well as using histological observations, and found that DBP significantly increased oxidative damage and apoptosis in hippocampal neurons via the ERK 1/2 pathway in mice. We also found that pretreatment with the dihydropyridine's (DHP's) Ca2+ antagonist, nimodipine (NMDP), combined with the antioxidant Vitamin E (VE), attenuated ERK 1/2 phosphorylation and DBP-mediated disorders, suggesting that a combined use of VE and NMDP can ameliorate DBP-induced memory deficit and apoptosis via inhibiting the ERK 1/2 pathway. These results indicate that DBP predisposes oxidative damage and apoptosis in hippocampal neurons by activation of the ERK 1/2 pathway, and may be proposed as a possible mechanism underlying LDs in children. Moreover, VE and NMDP may play a certain protective role in the targeted treatment of childhood LDs.
Collapse
Affiliation(s)
- Biao Yan
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Yanling Sun
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Jie Zeng
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Yingying Chen
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Chongyao Li
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Peng Song
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Lin Zhang
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Xu Yang
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Yang Wu
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China.
| | - Ping Ma
- Laboratory of Environment-Immunological and Neurological Diseases, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China.
| |
Collapse
|
134
|
Novel function of PiT1/SLC20A1 in LPS-related inflammation and wound healing. Sci Rep 2019; 9:1808. [PMID: 30755642 PMCID: PMC6372663 DOI: 10.1038/s41598-018-37551-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.
Collapse
|
135
|
Abstract
Cancer stem cells (CSCs) are rare types of cells responsible for tumor development, relapse, and metastasis. However, current research in CSC biology is largely limited by the difficulty of obtaining sufficient CSCs. Single-cell analysis techniques are promising tools for CSC-related studies. Here, we used the Single-probe mass spectrometry (MS) technique to investigate the metabolic features of live colorectal CSCs at the single-cell level. Experimental data were analyzed using statistical analysis methods, including the t-test and partial least squares discriminant analysis. Our results indicate that the overall metabolic profiles of CSCs are distinct from non-stem cancer cells (NSCCs). Specifically, we demonstrated that tricarboxylic acid (TCA) cycle metabolites are more abundant in CSCs compared to NSCCs, indicating their major energy production pathways are different. Moreover, CSCs have relatively higher levels of unsaturated lipids. Inhibiting the activities of stearoyl-CoA desaturase-1 (SCD1), nuclear factor κB (NF-κB), and aldehyde dehydrogenases (ALDH1A1) in CSCs significantly reduced the abundances of unsaturated lipids and hindered the formation of spheroids, resulting in reduced stemness of CSCs. Our techniques and experimental protocols can be potentially used for metabolomic studies of other CSCs and rare types of cells and provide a new approach to discovering functional biomarkers as therapeutic targets.
Collapse
Affiliation(s)
- Mei Sun
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
136
|
Corsetti G, Chen-Scarabelli C, Romano C, Pasini E, Dioguardi FS, Onorati F, Knight R, Patel H, Saravolatz L, Faggian G, Scarabelli TM. Autophagy and Oncosis/Necroptosis Are Enhanced in Cardiomyocytes from Heart Failure Patients. Med Sci Monit Basic Res 2019; 25:33-44. [PMID: 30713336 PMCID: PMC6373236 DOI: 10.12659/msmbr.913436] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Although originally described as a survival mechanism, it is unknown whether and to what extent autophagy is implicated in the terminal stages of heart failure. Here, we studied magnitude and evolution of autophagy in patients with intractable heart failure. Material/Methods Myocardial samples were obtained from 22 patients with ischemic cardiomyopathy and idiopathic dilated cardiomyopathy who were undergoing cardiac transplantation. Hearts from 11 patients who died from non-cardiac causes were used as control samples. Autophagy was evaluated by immunostaining with a monoclonal microtubule associated protein light chain 3 (LC3)-II antibody, while the relationship of autophagy with apoptosis and oncosis was assessed by double staining with TUNEL (terminal deoxynucleotidyl transferase – mediated deoxyuridine triphosphate nick end labeling) assay and complement 9 (C9) immunological staining, respectively. In addition, several necroptotic markers, including RIP1 and RIP3 (receptor interacting protein kinase 1 and 3), anti-C3 (cleaved-caspase-3), and anti-NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) were assessed by immunohistochemistry. Results Anti-LC3-II staining was detected in 8.7±1.6% of the heart failure patient heart samples and in 1.2±0.3% of control patient heart samples. Vacuole formation started at one nuclear pole, before becoming bipolar and involving the cytosol. Subsequently, the autophagic process extended also to the nuclei, which underwent a progressive vacuolization and disintegration, assuming a peculiar “strawberry like appearance”. Myocytes with extensive vacuole formation exhibited nuclear degeneration, which was associated with TUNEL, C3, C9, RIP1, and RIP3 positive staining. Conversely, myocytes with less extensive vacuole formation showed RIP1 and NF-κB positive staining, though not positivity for other cell death markers. Conclusions Autophagy was extensively detected in end-stage heart failure and its progression, resulted in secondary cell death, with occurrence of oncosis and necroptosis exceeding that of apoptosis. Conversely, activation of the RIP1/NF-κB pathway was associated with cell survival.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carol Chen-Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| | - Claudia Romano
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Evasio Pasini
- Scientific Clinical Institutes Maugeri, Cardiac Rehabilitation Lumezzane Institute, Brescia, Italy
| | | | - Francesco Onorati
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Richard Knight
- Medical Research Council (MRC) Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Hemang Patel
- General Medical Education, Department of Internal Medicine, Ascension St. John Hospital, Detroit, MI, USA
| | - Louis Saravolatz
- Department of Medicine, Ascension St John Hospital and Wayne State University School of Medicine, Detroit, MI, USA
| | - Giuseppe Faggian
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Tiziano M Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| |
Collapse
|
137
|
Cai W, Zhong S, Zheng F, Zhang Y, Gao F, Xu H, Cai X, Lan J, Huang D, Shi G. Angiotensin II confers resistance to apoptosis in cardiac myofibroblasts through the AT1/ERK1/2/RSK1 pathway. IUBMB Life 2019; 71:261-276. [PMID: 30452117 DOI: 10.1002/iub.1967] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 02/05/2023]
Abstract
Myofibroblast apoptosis is essential for normal resolution of wound repair, including cardiac infarction repair. Impaired cardiac myofibroblast (CMF) apoptosis is associated with excessive extracellular matrix (ECM) deposition, which could be responsible for pathological cardiac fibrosis. Conventionally, angiotensin II (Ang II), a soluble peptide, is implicated in fibrogenesis because it induces cardiac fibroblast (CFb) proliferation, differentiation, and collagen synthesis. However, the role of Ang II in regulation of CMF survival and apoptosis has not been fully clarified. In this report, we cultured neonatal rat CFbs, which transform into CMFs after passage 3 (6-8 days), and investigated the effects of Ang II on CMFs challenged by TNF-α combined with cycloheximide and the underlying mechanisms. Here, we show that Ang II rapidly activates MAPKs but not AKT in CMFs and confers apoptosis resistance, as evidenced by the inhibition of caspase-3 cleavage, early apoptotic cells and late apoptotic cells. This inhibitory effect of Ang II was reversed by blockade of AT1 or inactivation of ERK1/2 or RSK1 but not AT2, indicating that activation of the prosurvival AT1/ERK1/2/RSK1 signaling pathway mediates apoptosis resistance. TGF-β, a latent fibrotic factor, was found to have no relation to Ang II-induced apoptosis resistance in our study. Furthermore, Ang II-mediated apoptosis resistance, which was conferred by activation of the AT1/ERK1/2/RSK1 signaling pathway, was also confirmed in human adult ventricular cardiac myofibroblasts. Collectively, our findings suggest a novel profibrotic mechanism of Ang II in which it promotes myofibroblast resistance to apoptosis in addition to classical mechanisms, providing a potential novel therapeutic approach by targeting prosurvival signaling pathways. © 2018 IUBMB Life, 71(1):261-276, 2019.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin II Type 2 Receptor Blockers/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Butadienes/pharmacology
- Cell Survival/drug effects
- Cycloheximide/pharmacology
- Flavonoids/pharmacology
- Gene Expression Regulation
- Humans
- Imidazoles/pharmacology
- Losartan/pharmacology
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myofibroblasts/cytology
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Nitriles/pharmacology
- Primary Cell Culture
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Wenfeng Cai
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Shuping Zhong
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Fuchun Zheng
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongquan Xu
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiangna Cai
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Junhong Lan
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
- Department of Cardiovascular Diseases, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
138
|
Abstract
The master pro-inflammatory cytokine, tumour necrosis factor (TNF), has been shown to modulate multiple signalling pathways, with wide-ranging downstream effects. TNF plays a vital role in the typical immune response through the regulation of a number of pathways encompassing an immediate inflammatory reaction with significant innate immune involvement as well as cellular activation with subsequent proliferation and programmed cell death or necrosis. As might be expected with such a broad spectrum of cellular effects and complex signalling pathways, TNF has also been implicated in a number of disease states, such as rheumatoid arthritis, ankylosing spondylitis, and Crohn’s disease. Since the time of its discovery over 40 years ago, TNF ligand and its receptors, TNF receptor (TNFR) 1 and 2, have been categorised into two complementary superfamilies, namely TNF (TNFSF) and TNFR (TNFRSF), and 19 ligands and 29 receptors have been identified to date. There have been significant advances in our understanding of TNF signalling pathways in the last decade, and this short review aims to elucidate some of the most recent advances involving TNF signalling in health and disease.
Collapse
Affiliation(s)
- Jonathan Holbrook
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, UK.,Leeds Institute of Medical Research at St. James's, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Leeds, UK
| | - Samuel Lara-Reyna
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, UK.,Leeds Institute of Medical Research at St. James's, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Leeds, UK
| | - Heledd Jarosz-Griffiths
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, UK.,Leeds Institute of Medical Research at St. James's, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Leeds, UK
| | - Michael McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Leeds, UK
| |
Collapse
|
139
|
Luo YX, Wang XY, Huang YJ, Fang SH, Wu J, Zhang YB, Xiong TQ, Yang C, Shen JG, Sang CL, Wang Q, Fang JS. Systems pharmacology-based investigation of Sanwei Ganjiang Prescription: related mechanisms in liver injury. Chin J Nat Med 2018; 16:756-765. [PMID: 30322609 DOI: 10.1016/s1875-5364(18)30115-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Liver injury remains a significant global health problem and has a variety of causes, including oxidative stress (OS), inflammation, and apoptosis of liver cells. There is currently no curative therapy for this disorder. Sanwei Ganjiang Prescription (SWGJP), derived from traditional Chinese medicine (TCM), has shown its effectiveness in long-term liver damage therapy, although the underlying molecular mechanisms are still not fully understood. To explore the underlining mechanisms of action for SWGJP in liver injury from a holistic view, in the present study, a systems pharmacology approach was developed, which involved drug target identification and multilevel data integration analysis. Using a comprehensive systems approach, we identified 43 candidate compounds in SWGJP and 408 corresponding potential targets. We further deciphered the mechanisms of SWGJP in treating liver injury, including compound-target network analysis, target-function network analysis, and integrated pathways analysis. We deduced that SWGJP may protect hepatocytes through several functional modules involved in liver injury integrated-pathway, such as Nrf2-dependent anti-oxidative stress module. Notably, systems pharmacology provides an alternative way to investigate the complex action mode of TCM.
Collapse
Affiliation(s)
- Yun-Xia Luo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin-Yue Wang
- Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yu-Jie Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shu-Huan Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jun Wu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yong-Bin Zhang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Tian-Qin Xiong
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cong Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Gang Shen
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Chuan-Lan Sang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jian-Song Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
140
|
Chen L, Liu L, Xie Z, Wang F, Zhu L, Zhang C, Fan P, Sinkemani A, Hong X, Wu X. Protein kinase RNA‐like ER kinase/eukaryotic translation initiation factor 2α pathway attenuates
tumor necrosis factor alpha‐induced apoptosis in nucleus pulposus cells by activating autophagy. J Cell Physiol 2018; 234:11631-11645. [PMID: 30515797 DOI: 10.1002/jcp.27820] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/06/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Lu Chen
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
- Surgery Research Center, Institute of Clinical Sciences, School of Medicine, Southeast University Nanjing China
| | - Lei Liu
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
| | - Zhi‐Yang Xie
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
| | - Feng Wang
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
| | - Lei Zhu
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
| | - Cong Zhang
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
- Surgery Research Center, Institute of Clinical Sciences, School of Medicine, Southeast University Nanjing China
| | - Pan Fan
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
- Surgery Research Center, Institute of Clinical Sciences, School of Medicine, Southeast University Nanjing China
| | - Arjun Sinkemani
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
- Surgery Research Center, Institute of Clinical Sciences, School of Medicine, Southeast University Nanjing China
| | - Xin Hong
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
| | - Xiao‐Tao Wu
- Department of Spine Surgery Zhongda Hospital, School of Medicine, Southeast University Nanjing China
- Surgery Research Center, Institute of Clinical Sciences, School of Medicine, Southeast University Nanjing China
| |
Collapse
|
141
|
Okamoto M, Hidaka A, Toyama M, Baba M. Galectin-3 is involved in HIV-1 expression through NF-κB activation and associated with Tat in latently infected cells. Virus Res 2018; 260:86-93. [PMID: 30481548 DOI: 10.1016/j.virusres.2018.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/12/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
Abstract
Galectin-3 (Gal-3) is involved in many biological processes and pathogenesis of diseases in part through nuclear factor (NF)-κB activation. We demonstrated that Gal-3 expression was significantly induced by tumor necrosis factor (TNF)-α or phorbol 12-myristate 13-acetate in OM-10.1 and ACH-2 cells, which are considered as a model of HIV-1 latently infected cells. The expression of Gal-3 was also associated with their viral production. However, the induction of Gal-3 by TNF-α was not observed in their uninfected parental cells. Knockdown of Gal-3 resulted in the suppression of NF-κB activation and HIV-1 replication in the latently infected cells. The expression level of Gal-3 was highly correlated with that of HIV-1 Tat in the latently infected cells stimulated with TNF-α. Furthermore, colocalization and possible interaction of Gal-3 and Tat were observed in the stimulated cells. These results suggent that Gal-3 expression is closely correlated with HIV-1 expression in latently infected cells through NF-κB activation and the interaction with Tat.
Collapse
Affiliation(s)
- Mika Okamoto
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Akemi Hidaka
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Masaaki Toyama
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Masanori Baba
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Kagoshima University, Kagoshima, 890-8544, Japan.
| |
Collapse
|
142
|
Chen L, Xie ZY, Liu L, Zhu L, Wang F, Fan P, Sinkemani A, Zhang C, Hong X, Wu XT. Nuclear factor-kappa B-dependent X-box binding protein 1 signalling promotes the proliferation of nucleus pulposus cells under tumour necrosis factor alpha stimulation. Cell Prolif 2018; 52:e12542. [PMID: 30430692 PMCID: PMC6496019 DOI: 10.1111/cpr.12542] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/27/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Tumour necrosis factor alpha (TNF-α) expressed by nucleus pulposus cells (NPCs) plays a critical role in intervertebral disc (IVD) degeneration. A key unfolded protein response (UPR) component, X-box binding protein 1 (XBP1) and nuclear factor-kappa B (NF-κB) are essential for cell survival and proliferation. The aim of our study was to elucidate the roles of XBP1 and NF-κB in IVD degeneration (IDD). MATERIALS AND METHODS Rat NPCs were cultured with TNF-α in the presence or absence of XBP1 and NF-κB-p65 small interfering RNA. The associated genes and proteins were evaluated through quantitative real-time PCR, Western blot analyses and immunofluorescence staining to monitor UPR and NF-κB signalling and identify the regulatory mechanism of p65 by XBP1. Cell counting kit-8 assay, cell cycle analysis and related gene and protein expression were performed to examine the proliferation of NPCs. RESULTS The acute exposure of TNF-α accelerated the proliferation of rat NPCs by activating the UPR/XBP1 pathway. XBP1 signalling favoured the phosphorylation and nuclear translocation of p65 subunit of NF-κB. The activation of NF-κB in the later phase also enhanced NPC proliferation. CONCLUSIONS Unfolded protein response reinforces the survival and proliferation of NPCs under TNF-α stimulation by activating the XBP1 pathway, and NF-κB serves as a vital mediator in these events. The XBP1 signalling of UPR can be a novel therapeutic target in IDD.
Collapse
Affiliation(s)
- Lu Chen
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhi-Yang Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lei Liu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lei Zhu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Pan Fan
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Arjun Sinkemani
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Cong Zhang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xin Hong
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiao-Tao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
143
|
Dynamic changes of proteasome and protective effect of bortezomib, a proteasome inhibitor, in mice with acute pancreatitis. Biochem Biophys Res Commun 2018; 505:126-133. [DOI: 10.1016/j.bbrc.2018.09.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
|
144
|
Dash AK, Nayak D, Hussain N, Mintoo MJ, Bano S, Katoch A, Mondhe DM, Goswami A, Mukherjee D. Synthesis and Investigation of the Role of Benzopyran Dihydropyrimidinone Hybrids in Cell Proliferation, Migration and Tumor Growth. Anticancer Agents Med Chem 2018; 19:276-288. [PMID: 30179143 DOI: 10.2174/1871520618666180903101422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/06/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cancer is the second leading cause of mortality worldwide after heart diseases, and lung cancer is the topmost cause of all cancer-related deaths in both sexes. Dihydropyrimidinones (DHPMs) are medicinally important class of molecules with diverse pharmacological activities including anticancer activity. The present study focuses on the molecular hybridization of novel Benzopyran with Dihydropyrimidinone and evaluation of the resulting hybrids for cancer cell proliferation, migration and tumor growth. METHODS We have synthesized a focused library of dihydropyrimidinone benzopyran hybrids (compounds 1-11) by joining the aromatic as well as pyran portions of the benzopyran core with dihydropyrimidinone. All the synthesized hybrid molecules were evaluated for their cytotoxic activities against a panel of four human cancer cell lines of diverse tissue origin, viz: A549 (lung carcinoma), MCF7 (mammary gland adenocarcinoma), HCT-116 (colorectal carcinoma), and PANC-1 (pancreatic duct carcinoma) with the help of MTT cell viability assay. A structure-activity relationship was made on the basis of IC50 values of different hybrids. Effect on cell proliferation was examined through colony formation assay, reactive oxygen species generation and mitochondrial membrane potential studies. Wound healing assays and cell scattering assays were employed to check the effect on cell migration. Western blotting experiments were performed to find out the molecular mechanism of action and anti-tumor studies were carried out to evaluate the in vivo efficacy of the selected lead molecule. RESULTS Two types of novel hybrids were synthesized efficiently from benzopyran aldehydes, ethylacetoacetate and urea under heteropolyacid catalysis. Compound 3 was found to be the most potent hybrid among the synthesized compounds with consistent cytotoxic activities against four human cancer cell lines (IC50 values: 0.139 - 2.32 μM). Compound 3 strongly inhibited proliferation abilities of A549 cells in colony formation assay. Compound 3 exerted oxidative stress-mediated mitochondrial dysfunction, in which mitochondrial reactive oxygen species (ROS) generation as a mechanism of its anti-proliferative effects was analysed. Further, the molecule abrogated migration and cell scattering properties of aggressive PANC-1 cells. Mechanistic studies revealed that compound 3 modulated NF-kB expression and its downstream oncogenic proteins involved in cancer cell proliferation and invasion. Finally, compound 3 confirmed its in vivo anti-tumor efficacy; there observed 41.87% tumor growth inhibition at a dose of 30 mg/kg/body weight against a mouse model of Ehrlich solid tumor. CONCLUSION Our study unravels a potential anticancer lead (compound 3) from DHPMs that have opened up new research avenues for the development of promising anticancer therapeutic agents.
Collapse
Affiliation(s)
- Ashutosh K Dash
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Schoolof pharmaceutical sciences, Shoolini University of Biotechnology and Management Sciences, Bajhol, PO Sultanpur, Distt. Solan-173229 (HP), India
| | - Debasis Nayak
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Nazar Hussain
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Mubashir J Mintoo
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sumera Bano
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Archana Katoch
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Dilip M Mondhe
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Anindya Goswami
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Debaraj Mukherjee
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India
| |
Collapse
|
145
|
Hou Y, Liang H, Rao E, Zheng W, Huang X, Deng L, Zhang Y, Yu X, Xu M, Mauceri H, Arina A, Weichselbaum RR, Fu YX. Non-canonical NF-κB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy. Immunity 2018; 49:490-503.e4. [PMID: 30170810 DOI: 10.1016/j.immuni.2018.07.008] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/23/2018] [Accepted: 07/17/2018] [Indexed: 12/18/2022]
Abstract
The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.
Collapse
Affiliation(s)
- Yuzhu Hou
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Hua Liang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Enyu Rao
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA; Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenxin Zheng
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaona Huang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Liufu Deng
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA; Shanghai Institute of Immunology; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, China
| | - Yuan Zhang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Xinshuang Yu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Meng Xu
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Helena Mauceri
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Ainhoa Arina
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA.
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235-9072, USA.
| |
Collapse
|
146
|
Guo J, Cheng Y. RETRACTED: MicroRNA-1247 inhibits lipopolysaccharides-induced acute pneumonia in A549 cells via targeting CC chemokine ligand 16. Biomed Pharmacother 2018; 104:60-68. [DOI: 10.1016/j.biopha.2018.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
|
147
|
Xu B, Lian S, Li SZ, Guo JR, Wang JF, Wang D, Zhang LP, Yang HM. GABAB receptor mediate hippocampal neuroinflammation in adolescent male and female mice after cold expose. Brain Res Bull 2018; 142:163-175. [PMID: 30031816 DOI: 10.1016/j.brainresbull.2018.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 07/04/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
Stress induces many non-specific inflammatory responses in the mouse brain, especially during adolescence. Although the impact of stress on the brain has long been reported, the effects of cold stress on hippocampal neuroinflammation in adolescent mice are not well understood; furthermore, whether these effects are gender specific are also not well established. Adolescent male and female C57BL/6 mice were exposed to 4 °C temperatures for 12 h, after which behavior was assessed using the open field test. Using western blotting and immunohistochemistry we also assessed glial cell numbers and microglial activation, as well as inflammatory cytokine levels and related protein expression levels. We found that in mice subjected to cold stress: 1) There were significant behavioral changes; 2) neuronal nuclei densities were smaller and total cell numbers were significantly decreased; 3) nuclear factor (NF)-κB and phosphorylated AKT were upregulated; 4) pro-inflammatory cytokines such as interleukin-6 and tumor necrosis factor-α were also upregulated; and 5) microglia were activated, while glial fibrillary acid protein and ionized calcium-binding adapter molecule 1 protein expression increased. Taken together, these results indicate that cold stress induces pro-inflammatory cytokine upregulation that leads to neuroinflammation and neuronal apoptosis in the hippocampi of adolescent mice. We believe that these effects are influenced by a GABAB/Rap1B/AKT/NF-κB pathway. Finally, male mice were more sensitive to the effects of cold stress than were female mice.
Collapse
Affiliation(s)
- Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jian-Fa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Di Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Li-Ping Zhang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| | - Huan-Min Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| |
Collapse
|
148
|
Peng M, Wang Y, Qiang L, Xu Y, Li C, Li T, Zhou X, Xiao M, Wang J. Interleukin-35 Inhibits TNF-α-Induced Osteoclastogenesis and Promotes Apoptosis via Shifting the Activation From TNF Receptor-Associated Death Domain (TRADD)-TRAF2 to TRADD-Fas-Associated Death Domain by JAK1/STAT1. Front Immunol 2018; 9:1417. [PMID: 30061878 PMCID: PMC6054960 DOI: 10.3389/fimmu.2018.01417] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/06/2018] [Indexed: 12/27/2022] Open
Abstract
Over-activated osteoclasts derived from myeloid or peripheral blood monocytes by inflammatory cytokines results in osteoporosis, osteoarthritis, and other bone erosion-related diseases. Interleukin 35 (IL-35) is a novel anti-inflammatory and immunosuppressive factor. This study investigated the effect of IL-35 on TNF-α-induced osteoclastogenesis. In the presence of IL-35, this process was detected by Tartrate-Resistant Acid Phosphatase (TRAP) staining, F-actin staining, and bone resorption assays. The effects of IL-35 on TNF-α-induced apoptosis were demonstrated by TUNEL staining, cell viability assays, and flow cytometry. Moreover, a microarray was performed to detect the effect of IL-35 on TNF-α-activated phosphatase kinase. The effect of IL-35 on the TNF-α-mediated activation of NF-κB, MAPK, TRAF2, RIP1, Fas-associated death domain (FADD), and caspase3 was further investigated. In addition, a murine calvarial osteolysis model was established via the subcutaneous injection of TNF-α onto the calvaria, and histological analysis was subsequently performed. As a result, IL-35 inhibited TNF-α-induced osteoclast formation and bone resorption in vitro and osteolysis calvaria in vivo. NFATc1, c-fos, and TRAP were downregulated by IL-35 through the inhibition of NF-κB and MAPK, during which JAK1/STAT1 was activated. Moreover, based on TUNEL staining and flow cytometry, IL-35 was shown to enhance TNF-α-induced osteoclast apoptosis. Meanwhile, FADD and cleaved-caspase 3 were increased in cells treated with TNF-α and IL-35, whereas the DNA-binding activity of NF-κB was increased in TNF-α-treated cells, but was decreased in cells treated with both TNF-α and IL-35. In conclusion, IL-35 inhibits TNF-α-induced osteoclastogenesis and promotes apoptosis by activating JAK1/STAT1 and shifting activation from TNF receptor-associated death domain (TRADD)-TRAF2/RIP1-NF-κB to TRADD-FADD-caspase 3 signaling.
Collapse
Affiliation(s)
- Mingzheng Peng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanguo Wang
- Department of Orthopedic-Spine Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, China
| | - Lei Qiang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| | - Yan Xu
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| | - Cuidi Li
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Zhou
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Xiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
149
|
Hesari A, Ghasemi F, Salarinia R, Biglari H, Tabar Molla Hassan A, Abdoli V, Mirzaei H. Effects of curcumin on NF-κB, AP-1, and Wnt/β-catenin signaling pathway in hepatitis B virus infection. J Cell Biochem 2018; 119:7898-7904. [PMID: 29923222 DOI: 10.1002/jcb.26829] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/28/2018] [Indexed: 12/30/2022]
Abstract
Curcumin is a yellow-orange powder derived from the Curcuma longa plant. Curcumin has been used extensively in traditional medicine for centuries. This component is non-toxic and shown different therapeutic properties such as anti-inflammatory, anti-cancer, antiviral, anti-bacterial, anti-fungal, anti-parasites, and anti-oxidant. Hepatitis B virus (HBV) is a small DNA member of the genus Orthohepadnavirus (Hepadnaviridae family) which is a highly contagious blood-borne viral pathogen. HBV infection is a major public health problem with 2 billion people infected throughout the world and 350 million suffering from chronic HBV infection. Increasing evidence indicated that curcumin as a natural product could be employed in the treatment of HBV patients. It has been showed that curcumin exerts its therapeutic effects on HBV patients via targeting a variety of cellular and molecular pathways such as Wnt/β-catenin, Ap1, STAT3, MAPK, and NF-κB signaling. Here, we summarized the therapeutic effects of curcumin on patients who infected with HBV. Moreover, we highlighted main signaling pathways (eg, NF-κB, AP1, and Wnt/β-catenin signaling) which affected by curcumin in HBV infections.
Collapse
Affiliation(s)
- AmirReza Hesari
- Faculty of Medicine, Department of Biotechnology, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Faculty of Medicine, Department of Biotechnology, Arak University of Medical Sciences, Arak, Iran
| | - Reza Salarinia
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamed Biglari
- Department of Environmental Health Engineering, School of Public Health, Gonabad University of Medical Sciences, Gonabad, Iran
| | | | - Vali Abdoli
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Hamed Mirzaei
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
150
|
Quercetin inhibits prostate cancer by attenuating cell survival and inhibiting anti-apoptotic pathways. World J Surg Oncol 2018; 16:108. [PMID: 29898731 PMCID: PMC6001031 DOI: 10.1186/s12957-018-1400-z] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023] Open
Abstract
Background Despite recent advances in diagnosis and treatment, prostate cancer (PCa) remains the leading cause of cancer-related deaths in men. Current treatments offered in the clinics are often toxic and have severe side effects. Hence, to treat and manage PCa, new agents with fewer side effects or having potential to reduce side effects of conventional therapy are needed. In this study, we show anti-cancer effects of quercetin, an abundant bioflavonoid commonly used to treat prostatitis, and defined quercetin-induced cellular and molecular changes leading to PCa cell death. Methods Cell viability was assessed using MTT. Cell death mode, mitochondrial outer membrane potential, and oxidative stress levels were determined by flow cytometry using Annexin V-7 AAD dual staining kit, JC-1 dye, and ROS detection kit, respectively. Antibody microarray and western blot were used to delineate the molecular changes induced by quercetin. Results PCa cells treated with various concentrations of quercetin showed time- and dose-dependent decrease in cell viability compared to controls, without affecting normal prostate epithelial cells. Quercetin led to apoptotic and necrotic cell death in PCa cells by affecting the mitochondrial integrity and disturbing the ROS homeostasis depending upon the genetic makeup and oxidative status of the cells. LNCaP and PC-3 cells that have an oxidative cellular environment showed ROS quenching after quercetin treatment while DU-145 showed rise in ROS levels despite having a highly reductive environment. Opposing effects of quercetin were also observed on the pro-survival pathways of PCa cells. PCa cells with mutated p53 (DU-145) and increased ROS showed significant reduction in the activation of pro-survival Akt pathway while Raf/MEK were activated in response to quercetin. PC-3 cells lacking p53 and PTEN with reduced ROS levels showed significant activation of Akt and NF-κB pathway. Although some of these changes are commonly associated with oncogenic response, the cumulative effect of these alterations is PCa cell death. Conclusions Our results demonstrated quercetin exerts its anti-cancer effects by modulating ROS, Akt, and NF-κB pathways. Quercetin could be used as a chemopreventive option as well as in combination with chemotherapeutic drugs to improve clinical outcomes of PCa patients.
Collapse
|