151
|
Baquedano E, Burgos-Ramos E, Canelles S, González-Rodríguez A, Chowen JA, Argente J, Barrios V, Valverde AM, Frago LM. Increased oxidative stress and apoptosis in the hypothalamus of diabetic male mice in the insulin receptor substrate-2 knockout model. Dis Model Mech 2016; 9:573-83. [PMID: 27013528 PMCID: PMC4892662 DOI: 10.1242/dmm.023515] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/11/2016] [Indexed: 01/12/2023] Open
Abstract
Insulin receptor substrate-2-deficient (IRS2(-/-)) mice are considered a good model to study the development of diabetes because IRS proteins mediate the pleiotropic effects of insulin-like growth factor-I (IGF-I) and insulin on metabolism, mitogenesis and cell survival. The hypothalamus might play a key role in the early onset of diabetes, owing to its involvement in the control of glucose homeostasis and energy balance. Because some inflammatory markers are elevated in the hypothalamus of diabetic IRS2(-/-) mice, our aim was to analyze whether the diabetes associated with the absence of IRS2 results in hypothalamic injury and to analyze the intracellular mechanisms involved. Only diabetic IRS2(-/-) mice showed increased cell death and activation of caspase-8 and -3 in the hypothalamus. Regulators of apoptosis such as FADD, Bcl-2, Bcl-xL and p53 were also increased, whereas p-IκB and c-FLIPL were decreased. This was accompanied by increased levels of Nox-4 and catalase, enzymes involved in oxidative stress. In summary, the hypothalamus of diabetic IRS2(-/-) mice showed an increase in oxidative stress and inflammatory markers that finally resulted in cell death via substantial activation of the extrinsic apoptotic pathway. Conversely, non-diabetic IRS2(-/-) mice did not show cell death in the hypothalamus, possibly owing to an increase in the levels of circulating IGF-I and in the enhanced hypothalamic IGF-IR phosphorylation that would lead to the stimulation of survival pathways. In conclusion, diabetes in IRS2-deficient male mice is associated with increased oxidative stress and apoptosis in the hypothalamus.
Collapse
Affiliation(s)
- Eva Baquedano
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Emma Burgos-Ramos
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Sandra Canelles
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Agueda González-Rodríguez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid E-28029, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Julie A Chowen
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Jesús Argente
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Vicente Barrios
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Angela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid E-28029, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid E-28029, Spain
| | - Laura M Frago
- Department of Paediatrics, Universidad Autónoma de Madrid, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Av. Menéndez Pelayo, 65, Madrid 28009, Spain Instituto de Investigación Sanitaria Princesa, IIS-IP, Madrid E-28006, Spain Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid E-28029, Spain
| |
Collapse
|
152
|
Glycyrrhetic Acid Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in Vivo. Molecules 2016; 21:523. [PMID: 27110761 PMCID: PMC6273862 DOI: 10.3390/molecules21040523] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/27/2022] Open
Abstract
Glycyrrhizae Radix (GR) is a Korean traditional herb medicine that is widely used in clinical health care. Glycyrrhetic acid (GA) is an aglycone saponin extracted from GR that has anti-inflammatory, anti-cancer, and anti-viral effects. However, the anti-inflammatory effects of GA in colitis have not been reported. This study investigated the role of GA on ulcerative colitis in a dextran sulfate sodium (DSS)-induced mouse colitis model. DSS-treated mice displayed weight loss and shortened colon length compared with control mice. Mice administered GA showed less weight loss and longer colon length than the DSS-treated group. Interleukin (IL)-6, IL-1β, and tumor necrosis factor-alpha were decreased by GA treatment. GA treatment also reduced DSS-induced microscopic damage to colon tissue. GA regulates the phosphorylation of transcription factors including nuclear factor-kappa B (NF-κB) and IκB alpha, and regulates the expression of cycloxygenase-2 and prostaglandin E2. GA thus showed beneficial effects in a mouse model of colitis, implicating GA might be a useful herb-derived medicine in the treatment of ulcerative colitis.
Collapse
|
153
|
D'Ignazio L, Rocha S. Hypoxia Induced NF-κB. Cells 2016; 5:cells5010010. [PMID: 27005664 PMCID: PMC4810095 DOI: 10.3390/cells5010010] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/24/2022] Open
Abstract
As Nuclear Factor-κB (NF-κB) is a major transcription factor responding to cellular stress, it is perhaps not surprising that is activated by hypoxia, or decreased oxygen availability. However, how NF-κB becomes activated in hypoxia is still not completely understood. Several mechanisms have been proposed and this review will focus on the main findings highlighting the molecules that have been identified in the process of hypoxia induced NF-κB. In addition, we will discuss the role of NF-κB in the control of the cellular response to hypoxia.
Collapse
Affiliation(s)
- Laura D'Ignazio
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow street, Dundee DD1 5EH, UK.
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow street, Dundee DD1 5EH, UK.
| |
Collapse
|
154
|
Jang SE, Choi JR, Han MJ, Kim DH. The Preventive and Curative Effect of Cyanidin-3β-D-Glycoside and Its Metabolite Protocatechuic Acid Against TNBS-induced Colitis in Mice. ACTA ACUST UNITED AC 2016. [DOI: 10.20307/nps.2016.22.4.282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Se-Eun Jang
- Department of Food and Nutrition, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Jong-Ryul Choi
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Myung Joo Han
- Department of Food and Nutrition, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
155
|
Zhao M, Fan J, Liu Y, Yu Y, Xu J, Wen Q, Zhang J, Fu S, Wang B, Xiang L, Feng J, Wu J, Yang L. Oncogenic role of the TP53-induced glycolysis and apoptosis regulator in nasopharyngeal carcinoma through NF-κB pathway modulation. Int J Oncol 2015; 48:756-64. [PMID: 26691054 DOI: 10.3892/ijo.2015.3297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/02/2015] [Indexed: 11/06/2022] Open
Abstract
The TP53-induced glycolysis and apoptosis regulator (TIGAR) is a p53 target gene, which functions to suppress reactive oxygen species (ROS) damage and protect cells from apoptosis. In this study, we investigated the role of TIGAR in nasopharyngeal carcinoma (NPC) tumorigenesis. Imnunohistochemical analysis of the tissue specimens from nasopharyngeal carcinoma patients showed a higher expression level of TIGAR in tumor tissues, compared with normal nasopharyngeal epithelium. Knockdown of TIGAR by lentivirus-shRNA in CNE-2 or 5-8F cells resulted in decreased cell growth, colony formation, migration, invasion, and induced apoptosis. TIGAR overexpression exerted the opposite effects except for apoptosis reduction. In the xenograft tumor models, TIGAR knockdown reduced tumor growth rate and weight, whereas TIGAR overexpression showed the opposite effects. In addition, the NF-κB signaling pathway was decreased in TIGAR silenced cells. In conclusion, our data demonstrated that TIGAR acted as an oncogene in NPC tumorigenesis, and knockdown of TIGAR inhibited NPC tumor growth through the NF-κB pathway.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Juan Fan
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yong Liu
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yanxin Yu
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jinhui Xu
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qinglian Wen
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jianwen Zhang
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shaozhi Fu
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Biqiong Wang
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Xiang
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jing Feng
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jingbo Wu
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Linglin Yang
- Department of Oncology, The First Hospital of Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
156
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
157
|
D'Ignazio L, Bandarra D, Rocha S. NF-κB and HIF crosstalk in immune responses. FEBS J 2015; 283:413-24. [PMID: 26513405 PMCID: PMC4864946 DOI: 10.1111/febs.13578] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022]
Abstract
Hypoxia and inflammation have been associated with a number of pathological conditions, in particular inflammatory diseases. While hypoxia is mainly associated with the activation of hypoxia‐inducible factors (HIFs), inflammation activates the family of transcription factor called nuclear factor‐kappa B (NF‐κB). An extensive crosstalk between these two main molecular players involved in hypoxia and inflammation has been demonstrated. This crosstalk includes common activating stimuli, shared regulators and targets. In this review, we discuss the current understanding of the role of NF‐κB and HIF in the context of the immune response. We review the crosstalk between HIF and NF‐κB in the control of the immune response in different immune cell types including macrophages, neutrophils and B and T cells. Furthermore the importance of the molecular crosstalk between HIFs and NF‐κB for a variety of medical conditions will be discussed.
Collapse
Affiliation(s)
- Laura D'Ignazio
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| | - Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| |
Collapse
|
158
|
Dysregulation of the NF-κB pathway as a potential inducer of bipolar disorder. J Psychiatr Res 2015; 70:18-27. [PMID: 26424419 DOI: 10.1016/j.jpsychires.2015.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 11/20/2022]
Abstract
A century of investigations enhanced our understanding of bipolar disorder although it remains a complex multifactorial disorder with a mostly unknown pathophysiology and etiology. The role of the immune system in this disorder is one of the most controversial topics in genetic psychiatry. Though inflammation has been consistently reported in bipolar patients, it remains unclear how the immunologic process influences the disorder. One of the core components of the immune system is the NF-κB pathway, which plays an essential role in the development of innate and adaptive immunity. Remarkably, the NF-κB pathway received only little attention in bipolar studies, as opposed to studies of related psychiatric disorders where immune dysregulation has been proposed to explain the neurodegeneration in patient conditions. If immune dysregulation can also explains the neurodegeneration in bipolar disorder, it will underscore the role of the immune system in the chronicity and pathophysiology of the disorder and may promote personalized therapeutic strategies. This is the first review to summarize the current knowledge of the pathophysiological functions of NF-κB in bipolar disorder.
Collapse
|
159
|
Anisuzzaman ASM, Haque A, Rahman MA, Wang D, Fuchs JR, Hurwitz S, Liu Y, Sica G, Khuri FR, Chen ZG, Shin DM, Amin ARMR. Preclinical In Vitro, In Vivo, and Pharmacokinetic Evaluations of FLLL12 for the Prevention and Treatment of Head and Neck Cancers. Cancer Prev Res (Phila) 2015; 9:63-73. [PMID: 26511491 DOI: 10.1158/1940-6207.capr-15-0240] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/14/2015] [Indexed: 12/28/2022]
Abstract
Despite its high promise for cancer prevention and therapy, the potential utility of curcumin in cancer is compromised by its low bioavailability and weak potency. The purpose of the current study was to assess the in vitro and in vivo efficacy and pharmacokinetic parameters of the potent curcumin analogue FLLL12 in SCCHN and identify the mechanisms of its antitumor effect. IC50 values against a panel of one premalignant and eight malignant head and neck cancer cell lines as well as apoptosis assay results suggested that FLLL12 is 10- to 24-fold more potent than natural curcumin depending on the cell line and induces mitochondria-mediated apoptosis. In vivo efficacy (xenograft) and pharmacokinetic studies also suggested that FLLL12 is significantly more potent and has more favorable pharmacokinetic properties than curcumin. FLLL12 strongly inhibited the expression of p-EGFR, EGFR, p-AKT, AKT, Bcl-2, and Bid and increased the expression of Bim. Overexpression of constitutively active AKT or Bcl-2 or ablation of Bim or Bid significantly inhibited FLLL12-induced apoptosis. Further mechanistic studies revealed that FLLL12 regulated EGFR and AKT at transcriptional levels, whereas Bcl-2 was regulated at the translational level. Finally, FLLL12 strongly inhibited the AKT downstream targets mTOR and FOXO1a and 3a. Taken together, our results strongly suggest that FLLL12 is a potent curcumin analogue with more favorable pharmacokinetic properties that induces apoptosis of head and neck cancer cell lines by inhibition of survival proteins including EGFR, AKT, and Bcl-2 and increasing of the proapoptotic protein Bim.
Collapse
Affiliation(s)
- Abu Syed Md Anisuzzaman
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Abedul Haque
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Mohammad Aminur Rahman
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Dongsheng Wang
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - James R Fuchs
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, Ohio
| | - Selwyn Hurwitz
- Department of Pediatrics and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Yuan Liu
- Biostatistics & Bioinformatics Shared Resource at Winship Cancer Institute, Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Atlanta, Georgia
| | - Gabriel Sica
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Zhuo Georgia Chen
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Dong M Shin
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - A R M Ruhul Amin
- Department of Hematology and Medical Oncology and Winship Cancer Institute of Emory University, Atlanta, Georgia.
| |
Collapse
|
160
|
Proteomic Study to Survey the CIGB-552 Antitumor Effect. BIOMED RESEARCH INTERNATIONAL 2015; 2015:124082. [PMID: 26576414 PMCID: PMC4630370 DOI: 10.1155/2015/124082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/26/2015] [Indexed: 11/17/2022]
Abstract
CIGB-552 is a cell-penetrating peptide that exerts in vitro and in vivo antitumor effect on cancer cells. In the present work, the mechanism involved in such anticancer activity was studied using chemical proteomics and expression-based proteomics in culture cancer cell lines. CIGB-552 interacts with at least 55 proteins, as determined by chemical proteomics. A temporal differential proteomics based on iTRAQ quantification method was performed to identify CIGB-552 modulated proteins. The proteomic profile includes 72 differentially expressed proteins in response to CIGB-552 treatment. Proteins related to cell proliferation and apoptosis were identified by both approaches. In line with previous findings, proteomic data revealed that CIGB-552 triggers the inhibition of NF-κB signaling pathway. Furthermore, proteins related to cell invasion were differentially modulated by CIGB-552 treatment suggesting new potentialities of CIGB-552 as anticancer agent. Overall, the current study contributes to a better understanding of the antitumor action mechanism of CIGB-552.
Collapse
|
161
|
Baldea I, Olteanu DE, Bolfa P, Ion RM, Decea N, Cenariu M, Banciu M, Sesarman AV, Filip AG. Efficiency of photodynamic therapy on WM35 melanoma with synthetic porphyrins: Role of chemical structure, intracellular targeting and antioxidant defense. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 151:142-52. [DOI: 10.1016/j.jphotobiol.2015.07.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 01/10/2023]
|
162
|
BRENNENSTUHL HEIKO, ARMENTO ANGELA, BRACZYSNKI ANNEKRISTIN, MITTELBRONN MICHEL, NAUMANN ULRIKE. IκBζ, an atypical member of the inhibitor of nuclear factor kappa B family, is induced by γ-irradiation in glioma cells, regulating cytokine secretion and associated with poor prognosis. Int J Oncol 2015; 47:1971-80. [DOI: 10.3892/ijo.2015.3159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/03/2015] [Indexed: 11/06/2022] Open
|
163
|
Cojocneanu Petric R, Braicu C, Raduly L, Zanoaga O, Dragos N, Monroig P, Dumitrascu D, Berindan-Neagoe I. Phytochemicals modulate carcinogenic signaling pathways in breast and hormone-related cancers. Onco Targets Ther 2015; 8:2053-2066. [PMID: 26273208 PMCID: PMC4532173 DOI: 10.2147/ott.s83597] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Over the years, nutrition and environmental factors have been demonstrated to influence human health, specifically cancer. Owing to the fact that cancer is a leading cause of death worldwide, efforts are being made to elucidate molecular mechanisms that trigger or delay carcinogenesis. Phytochemicals, in particular, have been shown to modulate oncogenic processes through their antioxidant and anti-inflammatory activities and their ability to mimic the chemical structure and activity of hormones. These compounds can act not only by influencing oncogenic proteins, but also by modulating noncoding RNAs such as microRNAs and long noncoding RNAs. Although we are only beginning to understand the complete effects of many natural compounds, such as phytochemicals, researchers are motivated to combine these agents with traditional, chemo-based, or hormone-based therapies to fight against cancer. Since ongoing studies continue to prove effective, herein we exalt the importance of improving dietary choices as a chemo-preventive strategy.
Collapse
Affiliation(s)
- Roxana Cojocneanu Petric
- Department of Biology, Babes-Bolyai University, Cluj-Napoca, Romania
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Physiopathology, Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Cluj-Napoca, Romania
| | - Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Nicolae Dragos
- Department of Biology, Babes-Bolyai University, Cluj-Napoca, Romania
- Department of Taxonomy and Ecology, Institute of Biological Research, Cluj-Napoca, Romania
| | - Paloma Monroig
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Dumitrascu
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof Dr Ion Chiricuţă”, Cluj-Napoca, Romania
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
164
|
Zhang J, Cao L, Wang H, Cheng X, Wang L, Zhu L, Yan T, Xie Y, Wu Y, Zhao M, Ma S, Wu M, Wang G, Hao H. Ginsenosides Regulate PXR/NF-κB Signaling and Attenuate Dextran Sulfate Sodium-Induced Colitis. Drug Metab Dispos 2015; 43:1181-9. [PMID: 25986850 DOI: 10.1124/dmd.115.063800] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/15/2015] [Indexed: 02/13/2025] Open
Abstract
Pregnane X receptor (PXR) activation exhibits anti-inflammatory effects via repressing nuclear factor-κB (NF-κB); however, its overactivation may disrupt homeostasis of various enzymes and transporters. Here we found that ginsenosides restore PXR/NF-κB signaling in inflamed conditions without disrupting PXR function in normal conditions. The effects and mechanisms of ginsenosides in regulating PXR/NF-κB signals were determined both in vitro and in vivo. Ginsenosides significantly inhibited NF-κB activation and restored the expression of PXR target genes in tumor necrosis factor-α-stimulated LS174T cells. Despite not being PXR agonists, ginsenosides repressed NF-κB activation in a PXR-dependent manner. Ginsenosides significantly increased the physical association between PXR and the NF-κB p65 subunit and thereby decreased the nuclear translocation of p65. Ginsenoside Rb1 and compound K (CK) were major bioactive compounds in the regulating PXR/NF-κB signaling. Consistently, ginsenosides significantly attenuated dextran sulfate sodium-induced experimental colitis, which was associated with restored PXR/NF-κB signaling. This study indicates that ginsenosides may elicit anti-inflammatory effects via targeting PXR/NF-κB interaction without disrupting PXR function in healthy conditions. Ginsenoside Rb1 and CK may serve as leading compounds in the discovery of new drugs that target PXR/NF-κB interaction in therapy for inflammatory bowel disease.
Collapse
Affiliation(s)
- Jun Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Xuefang Cheng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Lin Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Lin Zhu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Tingting Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Yang Xie
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Yuzheng Wu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Sijing Ma
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Mengqiu Wu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (J.Z., L.C., H.W., X.C., L.W., L.Z., T.Y., Y.X., Y.W., M.Z., S.M., M.W., G.W., H.H.); and School of Pharmacy, Nanjing Medical University, Nanjing, China (J.Z.)
| |
Collapse
|
165
|
Liu D, Wang Y, Jia Z, Wang L, Wang J, Yang D, Song J, Wang S, Fan Z. Demethylation of IGFBP5 by Histone Demethylase KDM6B Promotes Mesenchymal Stem Cell-Mediated Periodontal Tissue Regeneration by Enhancing Osteogenic Differentiation and Anti-Inflammation Potentials. Stem Cells 2015; 33:2523-2536. [DOI: 10.1002/stem.2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Mesenchymal stem cell (MSC)-mediated periodontal tissue regeneration is considered a promising method for periodontitis treatment. The molecular mechanism underlying directed differentiation and anti-inflammatory actions remains unclear, thus limiting potential MSC application. We previously found that insulin-like growth factor binding protein 5 (IGFBP5) is highly expressed in dental tissue-derived MSCs compared with in non-dental tissue-derived MSCs. IGFBP5 is mainly involved in regulating biological activity of insulin-like growth factors, and its functions in human MSCs and tissue regeneration are unclear. In this study, we performed gain- and loss-of-function assays to test whether IGFBP5 could regulate the osteogenic differentiation and anti-inflammatory potential in MSCs. We found that IGFBP5 expression was upregulated upon osteogenic induction, and that IGFBP5 enhanced osteogenic differentiation in MSCs. We further showed that IGFBP5 prompted the anti-inflammation effect of MSCs via negative regulation of NFκB signaling. Depletion of the histone demethylase lysine (K)-specific demethylase 6B (KDM6B) downregulated IGFBP5 expression by increasing histone K27 methylation in the IGFBP5 promoter. Moreover, IGFBP5 expression in periodontal tissues was downregulated in individuals with periodontitis compared with in healthy people, and IGFBP5 enhanced MSC-mediated periodontal tissue regeneration and alleviated local inflammation in a swine model of periodontitis. In conclusion, our present results reveal a new function for IGFBP5, provide insight into the mechanism underlying the directed differentiation and anti-inflammation capacities of MSCs, and identify a potential target mediator for improving tissue regeneration. Stem Cells 2015;33:2523–2536
Collapse
Affiliation(s)
- Dayong Liu
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yuejun Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Zhi Jia
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Jinsong Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Biochemistry and Molecular Biology Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Dongmei Yang
- Department of Pediatrics Capital Medical University School of Stomatology, Beijing, China
| | - Jianqiu Song
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Biochemistry and Molecular Biology Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
166
|
Liu D, Wang Y, Jia Z, Wang L, Wang J, Yang D, Song J, Wang S, Fan Z. Demethylation of IGFBP5 by Histone Demethylase KDM6B Promotes Mesenchymal Stem Cell-Mediated Periodontal Tissue Regeneration by Enhancing Osteogenic Differentiation and Anti-Inflammation Potentials. Stem Cells 2015; 33:2523-2536. [DOI: 21.doi: 10.1002/stem.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
Abstract
Mesenchymal stem cell (MSC)-mediated periodontal tissue regeneration is considered a promising method for periodontitis treatment. The molecular mechanism underlying directed differentiation and anti-inflammatory actions remains unclear, thus limiting potential MSC application. We previously found that insulin-like growth factor binding protein 5 (IGFBP5) is highly expressed in dental tissue-derived MSCs compared with in non-dental tissue-derived MSCs. IGFBP5 is mainly involved in regulating biological activity of insulin-like growth factors, and its functions in human MSCs and tissue regeneration are unclear. In this study, we performed gain- and loss-of-function assays to test whether IGFBP5 could regulate the osteogenic differentiation and anti-inflammatory potential in MSCs. We found that IGFBP5 expression was upregulated upon osteogenic induction, and that IGFBP5 enhanced osteogenic differentiation in MSCs. We further showed that IGFBP5 prompted the anti-inflammation effect of MSCs via negative regulation of NFκB signaling. Depletion of the histone demethylase lysine (K)-specific demethylase 6B (KDM6B) downregulated IGFBP5 expression by increasing histone K27 methylation in the IGFBP5 promoter. Moreover, IGFBP5 expression in periodontal tissues was downregulated in individuals with periodontitis compared with in healthy people, and IGFBP5 enhanced MSC-mediated periodontal tissue regeneration and alleviated local inflammation in a swine model of periodontitis. In conclusion, our present results reveal a new function for IGFBP5, provide insight into the mechanism underlying the directed differentiation and anti-inflammation capacities of MSCs, and identify a potential target mediator for improving tissue regeneration. Stem Cells 2015;33:2523–2536
Collapse
Affiliation(s)
- Dayong Liu
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yuejun Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Zhi Jia
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Jinsong Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Biochemistry and Molecular Biology Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Dongmei Yang
- Department of Pediatrics Capital Medical University School of Stomatology, Beijing, China
| | - Jianqiu Song
- Department of Endodontics Tianjin Medical University School of Stomatology, Tianjin, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Biochemistry and Molecular Biology Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
167
|
Shan X, Tian LL, Zhang YM, Wang XQ, Yan Q, Liu JW. Ginsenoside Rg3 suppresses FUT4 expression through inhibiting NF-κB/p65 signaling pathway to promote melanoma cell death. Int J Oncol 2015; 47:701-9. [PMID: 26094873 PMCID: PMC6903900 DOI: 10.3892/ijo.2015.3057] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/22/2015] [Indexed: 12/21/2022] Open
Abstract
Abnormal glycosylation is catalyzed by the specific glycosyltransferases and correlates with tumor cell apoptosis. Increased fucosyltransferase IV (FUT4) is seen in many types of cancer, and manipulating FUT4 expression through specific signaling pathway inhibits cell growth and induces apoptosis. NF-κB is known playing a vital role to control cell growth and apoptosis. Ginsenoside Rg3 is an herbal medicine with strong antitumor activity through inhibiting tumor growth and promoting tumor cell death. However, whether Rg3-induced inhibition on tumor development involves reduced NF-κB signaling and FUT4 expression remains unknown. In the present study, we found that Rg3 suppressed FUT4 expression by abrogating the binding of NF-κB to FUT4 promoter through inhibiting the expression of signaling molecules of NF-κB pathway, reducing NF-κB DNA binding activity and NF-κB transcription activity. NF-κB inhibitor (Bay 11-7082) or knocking down p65 expression by p65 siRNA also led to a significant decreased FUT4 expression. In addition, Rg3 induced apoptosis by activating both extrinsic and intrinsic apoptotic pathways. Moreover, in a xenograft mouse model, Rg3 downregulated FUT4 and NF-κB/p65 expression and suppressed melanoma cell growth and induced apoptosis without any noticeable toxicity. In conclusion, Rg3 induces tumor cell apoptosis correlated with its inhibitory effect on NF-κB signaling pathway-mediated FUT4 expression. Results suggest Rg3 might be a novel therapy agent for melanoma treatment.
Collapse
Affiliation(s)
- Xiu Shan
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Li Li Tian
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yu Mei Zhang
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xiao Qi Wang
- Department of Dermatology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qiu Yan
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ji Wei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
168
|
The homeoprotein DLX4 stimulates NF-κB activation and CD44-mediated tumor-mesothelial cell interactions in ovarian cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2298-308. [PMID: 26067154 DOI: 10.1016/j.ajpath.2015.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/09/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
Ovarian cancers often highly express inflammatory cytokines and form implants throughout the peritoneal cavity. However, the mechanisms that drive inflammatory signaling and peritoneal metastasis of ovarian cancer are poorly understood. We previously identified that high expression of DLX4, a transcription factor encoded by a homeobox gene, is associated with reduced survival of ovarian cancer patients. In this study, we identified that DLX4 stimulates attachment of ovarian tumor cells to peritoneal mesothelial cells in vitro and increases the numbers of peritoneal implants in xenograft models. DLX4 induced expression of the cell surface molecule CD44 in ovarian tumor cells, and inhibition of CD44 abrogated the ability of DLX4 to stimulate tumor-mesothelial cell interactions. The induction of CD44 by DLX4 was attributed to increased activity of NF-κB that was stimulated by the inflammatory cytokine IL-1β, a transcriptional target of DLX4. The stimulatory effects of DLX4 on CD44 levels and tumor-mesothelial cell interactions were abrogated when IL-1β or NF-κB was inhibited in tumor cells. Furthermore, DLX4 expression levels strongly correlated with NF-κB activation and disease stage in clinical specimens of ovarian cancer. Collectively, these findings indicate that DLX4 induces CD44 by stimulating IL-1β-mediated NF-κB activity, thereby promoting peritoneal metastasis of ovarian cancer.
Collapse
|
169
|
Lee SY, Jeong JJ, Eun SH, Kim DH. Anti-inflammatory effects of ginsenoside Rg1 and its metabolites ginsenoside Rh1 and 20(S)-protopanaxatriol in mice with TNBS-induced colitis. Eur J Pharmacol 2015; 762:333-43. [PMID: 26054809 DOI: 10.1016/j.ejphar.2015.06.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/26/2015] [Accepted: 06/04/2015] [Indexed: 10/23/2022]
Abstract
Ginsenoside Rg1, one of the main constituents of Panax ginseng, exhibits anti-inflammatory effect. In a preliminary study, it was observed that ginsenoside Rg1 was metabolized to 20(S)-protopanaxtriol via ginsenosides Rh1 and F1 by gut microbiota. We further investigated the anti-inflammatory effects of ginsenoside Rg1 and its metabolites in vitro and in vivo. Ginsenosides Rg1, Rh1, and 20(S)-protopanaxtriol inhibited the activation of NF-κB activation, phosphorylation of transforming growth factor beta-activated kinase 1 and interleukin (IL)-1 receptor-associated kinase, and expression of tumor necrosis factor-α and IL-1β in lipopolysaccharide (LPS)-stimulated macrophages. They also inhibited the binding of LPS to toll-like receptor 4 on the macrophages. Orally administered ginsenoside Rg1, Rh1, or 20(S)-protopanaxtriol inhibited 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colon shortening, myeloperoxidase activity, and expression of IL-1β, IL-17, and tumor necrosis factor-α in mice with TNBS-induced colitis. They did not only inhibit TNBS-induced NF-κB activation, but also restored TNBS-induced Th17/Treg imbalance. They restored IL-10 and Foxp3 expression. Moreover, they inhibited Th17 cell differentiation in vitro. Of these metabolites, in vitro and in vivo anti-inflammatory effect of 20(S)-protopanaxtriol was the most potent, followed by Rh1. These findings suggest that ginsenoside Rg1 is metabolized to 20(S)-protopanaxtriol via ginsenosides Rh1 and F1 and these metabolites particularly 20(S)-protopanaxtriol, may ameliorate inflammatory disease such as colitis by inhibiting the binding of LPS to TLR4 on macrophages and restoring the Th17/Treg imbalance.
Collapse
Affiliation(s)
- Sang-Yun Lee
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Su-Hyeon Eun
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea.
| |
Collapse
|
170
|
Gibbs PEM, Miralem T, Maines MD. Biliverdin reductase: a target for cancer therapy? Front Pharmacol 2015; 6:119. [PMID: 26089799 PMCID: PMC4452799 DOI: 10.3389/fphar.2015.00119] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/20/2015] [Indexed: 12/30/2022] Open
Abstract
Biliverdin reductase (BVR) is a multifunctional protein that is the primary source of the potent antioxidant, bilirubin. BVR regulates activities/functions in the insulin/IGF-1/IRK/PI3K/MAPK pathways. Activation of certain kinases in these pathways is/are hallmark(s) of cancerous cells. The protein is a scaffold/bridge and intracellular transporter of kinases that regulate growth and proliferation of cells, including PKCs, ERK and Akt, and their targets including NF-κB, Elk1, HO-1, and iNOS. The scaffold and transport functions enable activated BVR to relocate from the cytosol to the nucleus or to the plasma membrane, depending on the activating stimulus. This enables the reductase to function in diverse signaling pathways. And, its expression at the transcript and protein levels are increased in human tumors and the infiltrating T-cells, monocytes and circulating lymphocytes, as well as the circulating and infiltrating macrophages. These functions suggest that the cytoprotective role of BVR may be permissive for cancer/tumor growth. In this review, we summarize the recent developments that define the pro-growth activities of BVR, particularly with respect to its input into the MAPK signaling pathway and present evidence that BVR-based peptides inhibit activation of protein kinases, including MEK, PKCδ, and ERK as well as downstream targets including Elk1 and iNOS, and thus offers a credible novel approach to reduce cancer cell proliferation.
Collapse
Affiliation(s)
- Peter E M Gibbs
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| | - Tihomir Miralem
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| | - Mahin D Maines
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| |
Collapse
|
171
|
Choi M, Jo H, Park HJ, Sateesh Kumar A, Lee J, Yun J, Kim Y, Han SB, Jung JK, Cho J, Lee K, Kwak JH, Lee H. Design, synthesis, and biological evaluation of benzofuran- and 2,3-dihydrobenzofuran-2-carboxylic acid N-(substituted)phenylamide derivatives as anticancer agents and inhibitors of NF-κB. Bioorg Med Chem Lett 2015; 25:2545-9. [DOI: 10.1016/j.bmcl.2015.04.050] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 04/06/2015] [Accepted: 04/17/2015] [Indexed: 01/02/2023]
|
172
|
Gjevestad GO, Holven KB, Ulven SM. Effects of Exercise on Gene Expression of Inflammatory Markers in Human Peripheral Blood Cells: A Systematic Review. CURRENT CARDIOVASCULAR RISK REPORTS 2015; 9:34. [PMID: 26005511 PMCID: PMC4439514 DOI: 10.1007/s12170-015-0463-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regular physical activity seems to be one of the most important contributors to prevent disease and promote health. Being physically active reduces the risk of developing chronic diseases such as cardiovascular disease, diabetes, and some types of cancers. The molecular mechanisms are however not fully elucidated. Depending on duration and intensity, exercise will cause disruption of muscle fibers triggering a temporary inflammatory response. This response may not only involve the muscle tissue, but also peripheral tissues such as white blood cells, which are important components of the immune system. The immune system plays a vital role in the development of atherosclerosis, thereby making white blood cells relevant to study when looking at molecular mechanisms induced by physical activity. In this review, we summarize the existing literature on exercise and gene expression in human white blood cells, and discuss these results in relation to inflammation and atherosclerosis.
Collapse
Affiliation(s)
| | - Kirsten B Holven
- University of Oslo, Oslo, Norway ; Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway
| | - Stine M Ulven
- Oslo and Akershus University College of Applied Sciences, Oslo, Norway
| |
Collapse
|
173
|
Clark R, Lee J, Lee SH. Synergistic anticancer activity of capsaicin and 3,3'-diindolylmethane in human colorectal cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:4297-4304. [PMID: 25876645 DOI: 10.1021/jf506098s] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. A promising area of cancer research is focused on chemoprevention by nutritional compounds. Epidemiological studies have shown a strong negative correlation between fruit, vegetable, and spice intake and rates of cancer. Although individual active compounds have demonstrated significant anticancer activity, an emerging area of research is focusing on the combination of multiple dietary compounds that act synergistically on cancer to exert greater effects. The current study evaluated the potential synergistic effects of capsaicin, an active compound from red chili peppers, in combination with 3,3'-diindolylmethane (DIM), from cruciferous vegetables. A synergistic induction of apoptosis and inhibition of cell proliferation was observed in human colorectal cancer cells treated with the combination of capsaicin and DIM. It was also observed that these two compounds activated transcriptional activity of NF-κB and p53 synergistically. Combination treatment stabilized nuclear p53 and up- or down-regulated expression of several target genes that are downstream of NF-κB and p53. The present study suggests capsaicin and DIM work synergistically to inhibit cell proliferation and induce apoptosis in colorectal cancer through modulating transcriptional activity of NF-κB, p53, and target genes associated with apoptosis.
Collapse
Affiliation(s)
- Ruth Clark
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| | - Jihye Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
174
|
Yun Y, Gao R, Yue H, Li G, Zhu N, Sang N. Synergistic effects of particulate matter (PM10) and SO2 on human non-small cell lung cancer A549 via ROS-mediated NF-κB activation. J Environ Sci (China) 2015; 31:146-53. [PMID: 25968268 DOI: 10.1016/j.jes.2014.09.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/02/2014] [Accepted: 09/06/2014] [Indexed: 05/21/2023]
Abstract
Since a real atmospheric scenario usually represents a system involving multiple pollutants, air pollution studies typically focused on describing adverse effects associated with exposure to individual pollutants cannot reflect actual health risk. Particulate matter (PM10) and sulfur dioxide (SO2) are two major pollutants derived from coal combustion processes and co-existing in coal-smoke air pollution, but their potentially synergistic toxicity remains elusive thus far. In this study, we investigated the cytotoxic responses of PM10 and SO2, singly and in binary mixtures, using human non-small cell lung cancer A549 cells, followed by clarifying the possible mechanisms for their interaction. The results indicated that the concomitant treatment of PM10 and SO2 at low concentrations led to synergistic injury in terms of cell survival and apoptosis occurrence, while PM10 and SO2 alone at the same concentrations did not cause damage to the cells. Also, radical oxygen species (ROS) production followed by nuclear factor kappa B (NF-κB) activation was involved in the above synergistic cytotoxicity, which was confirmed by the repression of the actions by an ROS inhibitor (NAC). This implies that assessment of health risk should consider the interactions between ambient PM and gaseous copollutants.
Collapse
Affiliation(s)
- Yang Yun
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China..
| | - Rui Gao
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Na Zhu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China..
| |
Collapse
|
175
|
Hanáková Z, Hošek J, Babula P, Dall'Acqua S, Václavík J, Šmejkal K. C-Geranylated Flavanones from Paulownia tomentosa Fruits as Potential Anti-inflammatory Compounds Acting via Inhibition of TNF-α Production. JOURNAL OF NATURAL PRODUCTS 2015; 78:850-863. [PMID: 25735399 DOI: 10.1021/acs.jnatprod.5b00005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Eleven new C-geranylated flavonoids, tomentodiplacones L, M, and N (1, 2, 10), tomentodiplacol B (3), 3',4'-O-dimethyl-5'-hydroxydiplacone (4), mimulones F, G, and H (5, 6, 7), paulowniones A (8) and B (9), tomentone (11), and 3',4',5'-trimethoxyflavanone (12), together with 11 known flavonoids (13-23), were isolated from fruits of Paulownia tomentosa. The structures of the compounds isolated were determined by spectroscopic data interpretation. The ability of compounds 1-23, together with the nonprenylated flavanones eriodictyol (24) and naringenin (25), to reduce the production of the pro-inflammatory cytokine TNF-α in THP-1 cells after bacterial lipopolysaccharide stimulation was evaluated using an in vitro screening test. The preliminary structure-activity relationships of these derivatives were also studied, and the correlation of their TNF-α inhibitory activity with their lipophilicity was investigated. The mechanism of action of compounds with significant antiphlogistic potential (4, 7, 10, 14, 22) was investigated. These compounds reduced both the secretion of TNF-α and the level of its corresponding mRNA. Compounds 4, 7, 10, 14, and 22 inhibited the nuclear translocation of NF-κB, which controls the expression of TNF-α, by blocking the degradation of IκB.
Collapse
Affiliation(s)
| | | | | | - Stefano Dall'Acqua
- §Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | | | | |
Collapse
|
176
|
Bradford JW, Baldwin AS. IKK/nuclear factor-kappaB and oncogenesis: roles in tumor-initiating cells and in the tumor microenvironment. Adv Cancer Res 2015; 121:125-145. [PMID: 24889530 DOI: 10.1016/b978-0-12-800249-0.00003-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The IKK/nuclear factor-kappaB pathway (NF-κB) is critical in proper immune function, cell survival, apoptosis, cellular proliferation, synaptic plasticity, and even memory. While NF-κB is crucial for both innate and adaptive immunity, defective regulation of this master transcriptional regulator is seen in a variety of diseases including autoimmune disease, neurodegenerative disease, and, important to this review, cancer. While NF-κB functions in cancer to promote a number of critical oncogenic functions, here we discuss the importance of the NF-κB signaling pathway in contributing to cancer through promotion of the tumor microenvironment and through maintenance/expansion of tumor-initiating cells, processes that appear to be functionally interrelated.
Collapse
Affiliation(s)
- Jennifer W Bradford
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.
| |
Collapse
|
177
|
Timosaponin AIII and its metabolite sarsasapogenin ameliorate colitis in mice by inhibiting NF-κB and MAPK activation and restoring Th17/Treg cell balance. Int Immunopharmacol 2015; 25:493-503. [DOI: 10.1016/j.intimp.2015.02.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 12/16/2022]
|
178
|
Singh V, Gupta D, Arora R. NF-kB as a key player in regulation of cellular radiation responses and identification of radiation countermeasures. Discoveries (Craiova) 2015; 3:e35. [PMID: 32309561 PMCID: PMC7159829 DOI: 10.15190/d.2015.27] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor (NF)-κB is a transcription factor that plays significant role in immunity, cellular survival and inhibition of apoptosis, through the induction of genetic networks. Depending on the stimulus and the cell type, the members of NF-κB related family (RelA, c-Rel, RelB, p50, and p52), forms different combinations of homo and hetero-dimers. The activated complexes (Es) translocate into the nucleus and bind to the 10bp κB site of promoter region of target genes in stimulus specific manner. In response to radiation, NF-κB is known to reduce cell death by promoting the expression of anti-apoptotic proteins and activation of cellular antioxidant defense system. Constitutive activation of NF-κB associated genes in tumour cells are known to enhance radiation resistance, whereas deletion in mice results in hypersensitivity to IR-induced GI damage. NF-κB is also known to regulate the production of a wide variety of cytokines and chemokines, which contribute in enhancing cell proliferation and tissue regeneration in various organs, such as the GI crypts stem cells, bone marrow etc., following exposure to IR. Several other cytokines are also known to exert potent pro-inflammatory effects that may contribute to the increase of tissue damage following exposure to ionizing radiation. Till date there are a series of molecules or group of compounds that have been evaluated for their radio-protective potential, and very few have reached clinical trials. The failure or less success of identified agents in humans could be due to their reduced radiation protection efficacy.
In this review we have considered activation of NF-κB as a potential marker in screening of radiation countermeasure agents (RCAs) and cellular radiation responses. Moreover, we have also focused on associated mechanisms of activation of NF-κB signaling and their specified family member activation with respect to stimuli. Furthermore, we have categorized their regulated gene expressions and their function in radiation response or modulation. In addition, we have discussed some recently developed radiation countermeasures in relation to NF-κB activation
Collapse
Affiliation(s)
- Vijay Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Damodar Gupta
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Rajesh Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
179
|
Deregulation of innate immune and inflammatory signaling in myelodysplastic syndromes. Leukemia 2015; 29:1458-69. [PMID: 25761935 DOI: 10.1038/leu.2015.69] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/31/2014] [Accepted: 01/25/2015] [Indexed: 12/18/2022]
Abstract
Myelodysplastic syndromes (MDSs) are a group of heterogeneous clonal hematologic malignancies that are characterized by defective bone marrow (BM) hematopoiesis and by the occurrence of intramedullary apoptosis. During the past decade, the identification of key genetic and epigenetic alterations in patients has improved our understanding of the pathophysiology of this disease. However, the specific molecular mechanisms leading to the pathogenesis of MDS have largely remained obscure. Recently, essential evidence supporting the direct role of innate immune abnormalities in MDS has been obtained, including the identification of multiple key regulators that are overexpressed or constitutively activated in BM hematopoietic stem and progenitor cells. Mounting experimental results indicate that the dysregulation of these molecules leads to abnormal hematopoiesis, unbalanced cell death and proliferation in patients' BM, and has an important role in the pathogenesis of MDS. Furthermore, there is compelling evidence that the deregulation of innate immune and inflammatory signaling also affects other cells from the immune system and the BM microenvironment, which establish aberrant associations with hematopoietic precursors and contribute to the MDS phenotype. Therefore, the deregulation of innate immune and inflammatory signaling should be considered as one of the driving forces in the pathogenesis of MDS. In this article, we review and update the advances in this field, summarizing the results from the most recent studies and discussing their clinical implications.
Collapse
|
180
|
Mukherjee N, Houston TJ, Cardenas E, Ghosh R. To be an ally or an adversary in bladder cancer: the NF-κB story has not unfolded. Carcinogenesis 2015; 36:299-306. [PMID: 25543121 PMCID: PMC4425835 DOI: 10.1093/carcin/bgu321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/09/2014] [Accepted: 12/21/2014] [Indexed: 12/20/2022] Open
Abstract
Signaling and regulation of transcription factor nuclear factor-kappaB (NF-κB) has been an area of extensive research since its first discovery nearly three decades ago. Members of the NF-κB family have been reported to critically mediate a multitude of responses in normal cells. Therefore, it is not surprising that NF-κB function can go awry and result in pathological conditions including cancer. Despite its critical importance, the functional role of NF-κB has not received the same attention in cancers of all tissue types. In the case of cancer of the urinary bladder, which is the second most common urologic cancer, the involvement of NF-κB in the development of superficial or muscle invasive disease and during cancer recurrence is rudimentary at best. Nuclear expression of p65/RelA is seen in bladder cancer patients and has been found to negatively affect survival of patients with superficial and muscle invasive disease. Despite these observations, the exact mechanism of NF-κB upregulation and function remains unknown. Furthermore, the emergence of a tumor suppressive role for NF-κB in recent years suggests that the family may play the role of a double-edged sword in cancer, which remains unexplored in bladder cancer. The challenge now is to delineate the increasing complexity of this pathway in the development and progression of bladder cancer. Here, we review key aspects of the current knowledge of signaling and regulation by the NF-κB family focusing on its controversial role in cancer and highlight the importance of studying NF-κB in bladder cancer in particular.
Collapse
Affiliation(s)
| | | | | | - Rita Ghosh
- Department of Urology, Department of Pharmacology, Department of Molecular Medicine and Cancer Therapy and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
181
|
Biddlestone J, Bandarra D, Rocha S. The role of hypoxia in inflammatory disease (review). Int J Mol Med 2015; 35:859-69. [PMID: 25625467 PMCID: PMC4356629 DOI: 10.3892/ijmm.2015.2079] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 01/27/2015] [Indexed: 02/06/2023] Open
Abstract
Mammals have developed evolutionarily conserved programs of transcriptional response to hypoxia and inflammation. These stimuli commonly occur together in vivo and there is significant crosstalk between the transcription factors that are classically understood to respond to either hypoxia or inflammation. This crosstalk can be used to modulate the overall response to environmental stress. Several common disease processes are characterised by aberrant transcriptional programs in response to environmental stress. In this review, we discuss the current understanding of the role of the hypoxia-responsive (hypoxia-inducible factor) and inflammatory (nuclear factor-κB) transcription factor families and their crosstalk in rheumatoid arthritis, inflammatory bowel disease and colorectal cancer, with relevance for future therapies for the management of these conditions.
Collapse
Affiliation(s)
- John Biddlestone
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| |
Collapse
|
182
|
SHI SHANSHAN, YUAN CHENXING, ZHUANG KAIZAN, LIANG GUIKAI, YAO ZHANGTING, WANG DUODUO, WENG QINJIE, CAO JI, LUO PEIHUA, ZHU HONG, DING LING, MA SHENGLIN. Resistance of SMMC-7721 hepatoma cells to etoposide in hypoxia is reversed by VEGF inhibitor. Mol Med Rep 2015; 11:3842-7. [DOI: 10.3892/mmr.2015.3217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022] Open
|
183
|
Safa M, Tavasoli B, Manafi R, Kiani F, Kashiri M, Ebrahimi S, Kazemi A. Indole-3-carbinol suppresses NF-κB activity and stimulates the p53 pathway in pre-B acute lymphoblastic leukemia cells. Tumour Biol 2015; 36:3919-30. [DOI: 10.1007/s13277-014-3035-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 12/30/2014] [Indexed: 01/07/2023] Open
|
184
|
Chung CY, Park YL, Kim N, Oh HH, Myung DS, Kim JS, Cho SB, Lee WS, Kim HS, Ahn BW, Joo YE. Rice prolamin extract ameliorates acute murine colitis by inhibiting nuclear factor-kappa B and modulating intestinal apoptosis and cell proliferation. Clin Exp Immunol 2015; 178:537-47. [PMID: 25098704 DOI: 10.1111/cei.12431] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2014] [Indexed: 12/19/2022] Open
Abstract
We investigated the impact of rice prolamin extract (RPE) on lipopolysaccharide (LPS)-induced nuclear factor (NF)-κB signalling in intestinal epithelial cells and macrophages, and determined the therapeutic efficacy of RPE in acute murine colitis. The effect of RPE on LPS-induced NF-κB signalling and proinflammatory gene expression was evaluated by reverse transcription-polymerase chain reaction (RT-PCR), Western blotting, immunofluorescence and electrophoretic mobility shift assay (EMSA). The in-vivo efficacy of RPE was assessed in mice with 3% dextran sulphate sodium (DSS)-induced colitis. Apoptotic and cellular proliferative activities were evaluated by immunostaining with cleaved caspase-3 and proliferating cell nuclear antigen (PCNA) antibodies. RPE inhibited LPS-induced expression of monocyte chemotactic protein (MCP)-1, interleukin (IL)-6 and tumour necrosis factor (TNF)-alpha and LPS-induced NF-κB signalling in intestinal epithelial cells and macrophages. RPE-fed, DSS-exposed mice showed less weight loss, longer colon length and lower histological score compared to control diet-fed, DSS-exposed mice. Immunostaining analysis revealed a significant decrease of cleaved caspase-3 positive cells in RPE-fed, DSS-exposed mice compared to DSS-exposed mice. Also, the number of PCNA-positive cells within intact colonic crypts decreased significantly in RPE-fed, DSS-exposed mice compared to control diet-fed, DSS-exposed mice. DSS-induced NF-κB signalling was inhibited by RPE. RPE ameliorates intestinal inflammation by inhibiting NF-κB activation and modulating intestinal apoptosis and cell proliferation in an acute murine colitis.
Collapse
Affiliation(s)
- C-Y Chung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Bandarra D, Biddlestone J, Mudie S, Müller HAJ, Rocha S. HIF-1α restricts NF-κB-dependent gene expression to control innate immunity signals. Dis Model Mech 2014; 8:169-81. [PMID: 25510503 PMCID: PMC4314782 DOI: 10.1242/dmm.017285] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hypoxia and inflammation are intimately linked. It is known that nuclear factor κB (NF-κB) regulates the hypoxia-inducible factor (HIF) system, but little is known about how HIF regulates NF-κB. Here, we show that HIF-1α represses NF-κB-dependent gene expression. HIF-1α depletion results in increased NF-κB transcriptional activity both in mammalian cells and in the model organism Drosophila melanogaster. HIF-1α depletion enhances the NF-κB response, and this required not only the TAK-IKK complex, but also CDK6. Loss of HIF-1α results in an increased angiogenic response in mammalian cancer cells and increased mortality in Drosophila following infection. These results indicate that HIF-1α is required to restrain the NF-κB response, and thus prevents excessive and damaging pro-inflammatory responses.
Collapse
Affiliation(s)
- Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - John Biddlestone
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sharon Mudie
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - H-Arno J Müller
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK.
| |
Collapse
|
186
|
Zhu HC, Qiu T, Dan C, Liu XH, Hu CH. Blockage of RelB expression by gene silencing enhances the radiosensitivity of androgen‑independent prostate cancer cells. Mol Med Rep 2014; 11:1167-73. [PMID: 25370388 DOI: 10.3892/mmr.2014.2857] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 08/29/2014] [Indexed: 11/05/2022] Open
Abstract
Levels of the nuclear factor‑kappa B (NF‑κB) alternative pathway member RelB have been shown to correlate with the effect of radiation therapy in prostate cancer. RelB expression was evaluated by immunohistochemistry in normal prostate, benign prostate hyperplasia and prostate cancer specimens. RM‑1 cells were pretreated with RelB siRNA prior to radiation therapy, and RelB expression in cytoplasmic and nuclear extracts was detected by real‑time polymerase chain reaction and western blot analysis. The apoptotic rates of experimental RM‑1 cell groups were assessed by flow cytometry. A clonogenic growth array was used to evaluate the radiosensitivity of RM‑1 cell groups. The NF‑κB family member RelB was expressed at a high level in prostate cancer specimens. Compared with irradiated control cells, RM‑1 cells transfected with RelB siRNA and treated with radiation therapy demonstrated a significant downregulation of RelB expression in the cytoplasm and nucleus. Notably, flow cytometry revealed that pretreatment of RM‑1 cells with RelB siRNA enhanced the apoptotic rate in response to radiation therapy compared with controls. Clonogenic growth assay results revealed enhanced radiosensitivity of RelB siRNA cells at various dosage points compared with control groups. Blockage of the alternative NF‑κB pathway via RelB silencing is a promising approach to enhance the radiosensitivity of prostate cancer.
Collapse
Affiliation(s)
- Heng-Cheng Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Qiu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chao Dan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chun-Hai Hu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
187
|
Kim KA, Jang SE, Jeong JJ, Yu DH, Han MJ, Kim DH. Doenjang, a Korean soybean paste, ameliorates TNBS-induced colitis in mice by suppressing gut microbial lipopolysaccharide production and NF-κB activation. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.09.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
188
|
Kumar G, Date OS, Kim KS, Manjunath R. Infection of human amniotic and endothelial cells by Japanese encephalitis virus: Increased expression of HLA-F. Virology 2014; 471-473:29-37. [PMID: 25461528 DOI: 10.1016/j.virol.2014.09.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/05/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022]
Abstract
Productive infection of human amniotic and endothelial cell lines with Japanese encephalitis virus (JEV) was established leading to the induction of NFκB and HLA-F, a non-classical MHC molecule. Induction of the HLA-F gene and protein in JEV-infected cells was shown to be NFκB dependent since it was blocked by inhibitors of NFκB activation. ShRNA targeting lentivirus-mediated stable knockdown of the p65 subunit of NFκB inhibited JEV-mediated induction of HLA-F both in the amniotic cell line, AV-3 as well as the human brain microendothelial cell line, HBMEC. The induction of HLA-F by treatment of AV-3 with TNF-α was also inhibited by ShRNA mediated knockdown of NFκB. TNF-α treatment of HEK293T cells that were transfected with reporter plasmids under the control of HLA-F enhancer A elements resulted in significant transactivation of the luciferase reporter gene. NFκB-mediated induction of HLA-F following JEV infection and TNF-α exposure is being suggested for the first time.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Onkar Sanjay Date
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Kwang Sik Kim
- Department of pediatric infectious diseases, John Hopkins university school of medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
189
|
Jang SE, Jeong JJ, Hyam SR, Han MJ, Kim DH. Ursolic acid isolated from the seed of Cornus officinalis ameliorates colitis in mice by inhibiting the binding of lipopolysaccharide to Toll-like receptor 4 on macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:9711-21. [PMID: 25213465 DOI: 10.1021/jf501487v] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ursolic acid, which was isolated from an ethanol extract of Cornus officinalis seed, potently inhibited nuclear factor κ light-chain enhancer of activated B cells (NF-κB) activation in lipopolysaccharide (LPS)-stimulated peritoneal macrophages. Therefore, we investigated the anti-inflammatory mechanism of ursolic acid in LPS-stimulated macrophages and colitic mice. Ursolic acid inhibited phosphorylation of interleukin 1 receptor-associated kinase (IRAK)1, TAK1, inhibitor of nuclear factor κB kinase subunit β (IKKβ), and IκBα as well as activation of NF-κB and MAPKs in LPS-stimulated macrophages. Ursolic acid suppressed LPS-stimulated interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, cyclooxygenase (COX)-2, and inducible NO synthetase (iNOS) expression as well as PGE2 and NO levels. Ursolic acid not only inhibited the Alexa Fluor 488-conjugated LPS-mediated shift of macrophages but also reduced the intensity of fluorescent LPS bound to the macrophages transiently transfected with or without MyD88 siRNA. However, ursolic acid did not suppress NF-κB activation in peptidoglycan-stimulated macrophages. Oral administration of ursolic acid significantly inhibited 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colon shortening and myeloperoxidase (MPO) activity in mice. Ursolic acid also suppressed TNBS-induced COX-2 and iNOS expression as well as NF-κB activation in colon tissues. Ursolic acid (20 mg/kg) also inhibited TNBS-induced IL-1β, IL-6, TNF-α by 93, 86, and 85%, respectively (p < 0.05). However, ursolic acid reversed TNBS-mediated downregulation of IL-10 expression to 79% of the normal control group (p < 0.05). On the basis of these findings, ursolic acid may ameliorate colitis by regulating NF-κB and MAPK signaling pathways via the inhibition of LPS binding to TLR4 on immune cells.
Collapse
Affiliation(s)
- Se-Eun Jang
- Department of Life and Nanopharmaceutical Sciences, ‡Department of Food and Nutrition, and §Department of Pharmacy, Kyung Hee University , Seoul 130-701, Korea
| | | | | | | | | |
Collapse
|
190
|
Tinospora cordifolia Induces Differentiation and Senescence Pathways in Neuroblastoma Cells. Mol Neurobiol 2014; 52:719-33. [PMID: 25280667 DOI: 10.1007/s12035-014-8892-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/03/2014] [Indexed: 01/06/2023]
Abstract
Children diagnosed with neuroblastomas often suffer from severe side as well as late effects of conventional treatments like chemotherapy and radiotherapy. Recent advances in understanding of molecular pathways involved in cellular differentiation and apoptosis have helped in the development of new therapeutic approach based on differentiation-based therapy of malignant tumours. Natural medicines with their holistic therapeutic approach are known to selectively eliminate cancer cells thus provide a better substitute for the conventional treatment modes. The current study was aimed to investigate the anti-cancer potential of aqueous ethanolic extract of Tinospora cordifolia (TCE) using IMR-32 human neuroblastoma cell line as a model system. TCE is highly recommended in Ayurveda for its general body and metal health-promoting properties. TCE treatment was seen to arrest the majority of cells in G0/G1 phase and modulated the expression of DNA clamp sliding protein (PCNA) and cyclin D1. Further, TCE-treated cells showed differentiation as revealed by their morphology and the expression of neuronal cell specific differentiation markers NF200, MAP-2 and NeuN in neuroblastoma cells. The differentiated phenotype was associated with induction of senescence and pro-apoptosis pathways by enhancing expression of senescence marker mortalin and Rel A subunit of nuclear factor kappa beta (NFkB) along with decreased expression of anti-apoptotic marker, Bcl-xl. TCE exhibited anti-metastatic activity and significantly reduced cell migration in the scratched area along with downregulation of neural cell adhesion molecule (NCAM) polysialylation and secretion of matrix metalloproteinases (MMPs). Our data suggest that crude extract or active phytochemicals from this plant may be a potential candidate for differentiation-based therapy of malignant neuroblastoma cells.
Collapse
|
191
|
Mirandola L, Nguyen DD, Rahman RL, Grizzi F, Yuefei Y, Figueroa JA, Jenkins MR, Cobos E, Chiriva-Internati M. Anti-galectin-3 therapy: a new chance for multiple myeloma and ovarian cancer? Int Rev Immunol 2014; 33:417-427. [PMID: 24801755 DOI: 10.3109/08830185.2014.911855] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 12/23/2022]
Abstract
Here we review the role of Galectins in the molecular pathogenesis of multiple myeloma and ovarian cancer, with a special focus on Glectin-3. Multiple myeloma is the second most common hematologic malignancy worldwide. Because the pathogenesis of multiple myeloma is still incompletely understood, there is no ultimately effective cure, and this cancer results fatal. Ovarian cancer is the most lethal gynecologic malignancy worldwide. Due to the lack of screening techniques for early detection, patients are mostly diagnosed with advanced disease, which results ultimately fatal. Multiple myeloma and ovarian cancer have different biologies, but they share a strong dependence on adhesion with extracellular matrix and other cells. Galectin-3 plays a key role in regulating such adhesive abilities of tumor cells. Here we discuss the outcomes and possible mechanism of action of a truncated, dominant negative form of Galectin-3, Galectin-3C, in these malignancies. Overall, we report that Galectin-3C is a promising new compound for effective adjuvant therapies in advanced, refractory multiple myeloma and ovarian cancer.
Collapse
Affiliation(s)
- Leonardo Mirandola
- 1Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Jeong JJ, Jang SE, Hyam SR, Han MJ, Kim DH. Mangiferin ameliorates colitis by inhibiting IRAK1 phosphorylation in NF-κB and MAPK pathways. Eur J Pharmacol 2014; 740:652-61. [DOI: 10.1016/j.ejphar.2014.06.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 12/27/2022]
|
193
|
Jeong JJ, Jang SE, Hyam SR, Han MJ, Kim DH. The Rhizome Mixture of Anemarrhena asphodeloides and Coptidis chinensis Ameliorates Acute and Chronic Colitis in Mice by Inhibiting the Binding of Lipopolysaccharide to TLR4 and IRAK1 Phosphorylation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:809083. [PMID: 25276218 PMCID: PMC4167948 DOI: 10.1155/2014/809083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 07/25/2014] [Accepted: 08/03/2014] [Indexed: 02/06/2023]
Abstract
In the previous study, the mixture of the rhizome of Anemarrhena asphodeloides (AA, family Liliaceae) and the rhizome of Coptidis chinensis (CC, family Ranunculaceae) (AC-mix) improved TNBS- or oxazolone-induced colitis in mice. Therefore, to investigate its anticolitic mechanism, we measured its effect in acute and chronic DSS-induced colitic mice and investigated its anti-inflammatory mechanism in peritoneal macrophages. AC-mix potently suppressed DSS-induced body weight loss, colon shortening, myeloperoxidase activity, and TNF-α, IL-1β, and IL-6 expressions in acute or chronic DSS-stimulated colitic mice. Among AC-mix ingredients, AA, CC, and their main constituents mangiferin and berberine potently inhibited the expression of proinflammatory cytokines TNF-α and IL-1β, as well as the activation of NF-κB in LPS-stimulated peritoneal macrophages. AA and mangiferin potently inhibited IRAK phosphorylation, but CC and berberine potently inhibited the binding of LPS to TLR4 on macrophages, as well as the phosphorylation of IRAK1. AC-mix potently inhibited IRAK phosphorylation and LPS binding to TLR4 on macrophages. Based on these findings, AC-mix may ameliorate colitis by the synergistic inhibition of IRAK phosphorylation and LPS binding to TLR4 on macrophages.
Collapse
Affiliation(s)
- Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Se-Eun Jang
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
- Department of Food and Nutrition, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Supriya R. Hyam
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Myung Joo Han
- Department of Food and Nutrition, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, College of Pharmacy, Kyung Hee University, 1 Hoegi, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| |
Collapse
|
194
|
O'Hara A, Simpson J, Morin P, Loveridge CJ, Williams AC, Novo SM, Stark LA. p300-mediated acetylation of COMMD1 regulates its stability, and the ubiquitylation and nucleolar translocation of the RelA NF-κB subunit. J Cell Sci 2014; 127:3659-65. [PMID: 25074812 PMCID: PMC4150058 DOI: 10.1242/jcs.149328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/20/2014] [Indexed: 12/02/2022] Open
Abstract
Nucleolar sequestration of the RelA subunit of nuclear factor (NF)-κB is an important mechanism for regulating NF-κB transcriptional activity. Ubiquitylation, facilitated by COMMD1 (also known as MURR1), acts as a crucial nucleolar-targeting signal for RelA, but how this ubiquitylation is regulated, and how it differs from cytokine-mediated ubiquitylation, which causes proteasomal degradation of RelA, is poorly understood. Here, we report a new role for p300 (also known as EP300) in controlling stimulus-specific ubiquitylation of RelA, through modulation of COMMD1. We show that p300 is required for stress-mediated ubiquitylation and nucleolar translocation of RelA, but that this effect is indirect. We also demonstrate that COMMD1 is acetylated by p300 and that acetylation protects COMMD1 from XIAP-mediated proteosomal degradation. Furthermore, we demonstrate that COMMD1 acetylation is enhanced by aspirin-mediated stress, and that this acetylation is absolutely required for the protein to bind RelA under these conditions. In contrast, tumour necrosis factor (TNF) has no effect on COMMD1 acetylation. Finally, we demonstrate these findings have relevance in a whole tissue setting. These data offer a new paradigm for the regulation of NF-κB transcriptional activity, and the multiple other pathways controlled by COMMD1.
Collapse
Affiliation(s)
- Andrew O'Hara
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - James Simpson
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Pierre Morin
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Carolyn J Loveridge
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Ann C Williams
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Sonia M Novo
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Lesley A Stark
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
195
|
Rodriguez MS, Egaña I, Lopitz-Otsoa F, Aillet F, Lopez-Mato MP, Dorronsoro A, Dorronroso A, Lobato-Gil S, Sutherland JD, Barrio R, Trigueros C, Lang V. The RING ubiquitin E3 RNF114 interacts with A20 and modulates NF-κB activity and T-cell activation. Cell Death Dis 2014; 5:e1399. [PMID: 25165885 PMCID: PMC4454333 DOI: 10.1038/cddis.2014.366] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/20/2014] [Accepted: 07/03/2014] [Indexed: 12/16/2022]
Abstract
Accurate regulation of nuclear factor-κB (NF-κB)
activity is crucial to prevent a variety of disorders including immune and
inflammatory diseases. Active NF-κB promotes
IκBα and A20 expression, important negative
regulatory molecules that control the NF-κB response. In this
study, using two-hybrid screening we identify the RING-type zinc-finger protein
114 (RNF114) as an A20-interacting factor. RNF114 interacts with A20 in T cells
and modulates A20 ubiquitylation. RNF114 acts as negative regulator of
NF-κB-dependent transcription, not only by stabilizing the
A20 protein but also IκBα. Importantly, we
demonstrate that in T cells, the effect of RNF114 is linked to the modulation of
T-cell activation and apoptosis but is independent of cell cycle regulation.
Altogether, our data indicate that RNF114 is a new partner of A2O involved in
the regulation of NF-κB activity that contributes to the control
of signaling pathways modulating T cell-mediated immune response.
Collapse
Affiliation(s)
- M S Rodriguez
- Cancer Unit, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | - I Egaña
- CIC bioGUNE, Derio, Bizkaia, Spain
| | | | - F Aillet
- Cancer Unit, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | - M P Lopez-Mato
- Cytometry and Advanced Optical Microscopy Core Facility, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | | | - A Dorronroso
- Hematological Diseases, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | - S Lobato-Gil
- Cancer Unit, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | | | - R Barrio
- CIC bioGUNE, Derio, Bizkaia, Spain
| | - C Trigueros
- Hematological Diseases, Inbiomed, San Sebastian, Gipuzkoa, Spain
| | - V Lang
- Cancer Unit, Inbiomed, San Sebastian, Gipuzkoa, Spain
| |
Collapse
|
196
|
Kessler K, Borges LF, Ho-Tin-Noé B, Jondeau G, Michel JB, Vranckx R. Angiogenesis and remodelling in human thoracic aortic aneurysms. Cardiovasc Res 2014; 104:147-59. [PMID: 25139748 DOI: 10.1093/cvr/cvu196] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Human thoracic aneurysm of the ascending aorta (TAA) is a chronic disease characterized by dilatation of the aortic wall, which can progress to vessel dissection and rupture. TAA has several aetiologies, but all forms present common features, including tissue remodelling. Here, we determined and characterized the angiogenic process associated with TAA and its relation with wall remodelling. METHODS AND RESULTS Immunostaining for blood vessels showed an increased density of microvessels originating from the adventitia in the external medial layer of TAA compared with healthy aortas. Proteomic array analysis of 55 angiogenic factors in medial and adventitial layers showed different expression profiles in both tissue compartments between aneurysmal and healthy aortas. Quantification by ELISA confirmed that all forms of TAA contained higher levels of several pro- and anti-angiogenic factors, including angiopoietin-1 and -2, fibroblast growth factor-acidic, and thrombospondin-1, than that of healthy aortas. However, all groups showed comparable levels of vascular endothelial growth factor-A. Quantitative RT-PCR demonstrated that angiopoietins were overexpressed in TAA media. Immunostaining and electron microscopy revealed that neovessels had defective endothelial junctions and poor mural cell coverage. This incomplete structure was associated with the accumulation of plasminogen and albumin in the media of TAA. CONCLUSION We describe, for the first time, leaky neovessel formation in TAA media in association with an imbalance of angiogenic factor levels. Although the initiating mechanisms of neo-angiogenesis in TAA and the potential aetiology-related differences remain to be determined, our results suggest that neo-angiogenesis could participate in TAA wall remodelling and weakening through deposition of blood-borne zymogens.
Collapse
Affiliation(s)
- Ketty Kessler
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France INSERM Unit 1148, Hôpital Xavier Bichat, Secteur Claude Bernard, 46 rue Henri Huchard, FR-75877 Paris cedex 18, France
| | - Luciano F Borges
- Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Benoît Ho-Tin-Noé
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France
| | - Guillaume Jondeau
- Centre National de Référence pour le syndrome de Marfan et apparentés, Hôpital Xavier Bichat, Paris, France
| | - Jean-Baptiste Michel
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France INSERM Unit 1148, Hôpital Xavier Bichat, Secteur Claude Bernard, 46 rue Henri Huchard, FR-75877 Paris cedex 18, France
| | - Roger Vranckx
- Univ Paris Diderot, Sorbonne Paris Cité, LVTS, UMR-S1148, F-75018 Paris, France
| |
Collapse
|
197
|
Routray S. Caveolin-1 in oral squamous cell carcinoma microenvironment: an overview. Tumour Biol 2014; 35:9487-95. [PMID: 25123270 DOI: 10.1007/s13277-014-2482-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 08/08/2014] [Indexed: 11/29/2022] Open
Abstract
Caveolin-1 plays an important role in the pathogenesis of oncogenic cell transformation, tumorigenesis, and metastasis. Increased expression of caveolin-1 in an array of tumors has confirmed its value in prognosis. It has been established that oxidative stress is the main cause for loss of stromal caveolin-1 via autophagy in the tumor microenvironment. In this overview, we attempt to abridge the relationship between caveolin-1 and oral squamous cell carcinoma, taking all the established theories into consideration.
Collapse
Affiliation(s)
- Samapika Routray
- Department of Oral Pathology & Microbiology, Institute of Dental Sciences, SOA University, Ghatikia, Sector 8, Bhubaneswar, 751003, India,
| |
Collapse
|
198
|
Sandhu R, Rein J, D'Arcy M, Herschkowitz JI, Hoadley KA, Troester MA. Overexpression of miR-146a in basal-like breast cancer cells confers enhanced tumorigenic potential in association with altered p53 status. Carcinogenesis 2014; 35:2567-75. [PMID: 25123132 DOI: 10.1093/carcin/bgu175] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The tumor suppressor p53 is the most frequently mutated gene in human cancers, mutated in 25-30% of breast cancers. However, mutation rates differ according to breast cancer subtype, being more prevalent in aggressive estrogen receptor-negative tumors and basal-like and HER2-amplified subtypes. This heterogeneity suggests that p53 may function differently across breast cancer subtypes. We used RNAi-mediated p53 knockdown (KD) and antagomir-mediated KD of microRNAs to study how gene expression and cellular response to p53 loss differ in luminal versus basal-like breast cancer. As expected, p53 loss caused downregulation of established p53 targets (e.g. p21 and miR-34 family) and increased proliferation in both luminal and basal-like cell lines. However, some p53-dependent changes were subtype specific, including expression of miR-134, miR-146a and miR-181b. To study the cellular response to miR-146a upregulation in p53-impaired basal-like lines, antagomir KD of miR-146a was performed. KD of miR-146a caused decreased proliferation and increased apoptosis, effectively ablating the effects of p53 loss. Furthermore, we found that miR-146a upregulation decreased NF-κB expression and downregulated the NF-κB-dependent extrinsic apoptotic pathway (including tumor necrosis factor, FADD and TRADD) and antagomir-mediated miR-146a KD restored expression of these components, suggesting a plausible mechanism for miR-146a-dependent cellular responses. These findings are relevant to human basal-like tumor progression in vivo, since miR-146a is highly expressed in p53 mutant basal-like breast cancers. These findings suggest that targeting miR-146a expression may have value for altering the aggressiveness of p53 mutant basal-like tumors.
Collapse
Affiliation(s)
- Rupninder Sandhu
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
| | - Jessica Rein
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Monica D'Arcy
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason I Herschkowitz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA and
| | | | - Melissa A Troester
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
199
|
EGF receptor uses SOS1 to drive constitutive activation of NFκB in cancer cells. Proc Natl Acad Sci U S A 2014; 111:11721-6. [PMID: 25071181 DOI: 10.1073/pnas.1412390111] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activation of nuclear factor κB (NFκB) is a central event in the responses of normal cells to inflammatory signals, and the abnormal constitutive activation of NFκB is important for the survival of most cancer cells. In nonmalignant human cells, EGF stimulates robust activation of NFκB. The kinase activity of the EGF receptor (EGFR) is required, because the potent and specific inhibitor erlotinib blocks the response. Down-regulating EGFR expression or inhibiting EGFR with erlotinib impairs constitutive NFκB activation in several different types of cancer cells and, conversely, increased activation of NFκB leads to erlotinib resistance in these cells. We conclude that EGF is an important mediator of NFκB activation in cancer cells. To explore the mechanism, we selected an erlotinib-resistant cell line in which the guanine nucleotide exchange factor Son of Sevenless 1 (SOS1), well known to be important for EGF-dependent signaling to MAP kinases, is overexpressed. Increased expression of SOS1 increases NFκB activation in several different types of cancer cells, and ablation of SOS1 inhibits EGF-induced NFκB activation in these cells, indicating that SOS1 is a functional component of the pathway connecting EGFR to NFκB activation. Importantly, the guanine nucleotide exchange activity of SOS1 is not required for NFκB activation.
Collapse
|
200
|
Dimauro I, Grasso L, Fittipaldi S, Fantini C, Mercatelli N, Racca S, Geuna S, Di Gianfrancesco A, Caporossi D, Pigozzi F, Borrione P. Platelet-rich plasma and skeletal muscle healing: a molecular analysis of the early phases of the regeneration process in an experimental animal model. PLoS One 2014; 9:e102993. [PMID: 25054279 PMCID: PMC4108405 DOI: 10.1371/journal.pone.0102993] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/24/2014] [Indexed: 12/27/2022] Open
Abstract
Platelet-rich plasma (PRP) has received increasing interest in applied medicine, being widely used in clinical practice with the aim of stimulating tissue healing. Despite the reported clinical success, there is still a lack of knowledge when considering the biological mechanisms at the base of the activity of PRP during the process of muscle healing. The aim of the present study was to verify whether the local delivery of PRP modulates specific molecular events involved in the early stages of the muscle regeneration process. The right flexor sublimis muscle of anesthetized Wistar rats was mechanically injured and either treated with PRP or received no treatment. At day 2 and 5 after surgery, the animals were sacrificed and the muscle samples evaluated at molecular levels. PRP treatment increased significantly the mRNA level of the pro-inflammatory cytokines IL-1β, and TGF-β1. This phenomenon induced an increased expression at mRNA and/or protein levels of several myogenic regulatory factors such as MyoD1, Myf5 and Pax7, as well as the muscular isoform of insulin-like growth factor1 (IGF-1Eb). No effect was detected with respect to VEGF-A expression. In addition, PRP application modulated the expression of miR-133a together with its known target serum response factor (SRF); increased the phosphorylation of αB-cristallin, with a significant improvement in several apoptotic parameters (NF-κB-p65 and caspase 3), indexes of augmented cell survival. The results of the present study indicates that the effect of PRP in skeletal muscle injury repair is due both to the modulation of the molecular mediators of the inflammatory and myogenic pathways, and to the control of secondary pathways such as those regulated by myomiRNAs and heat shock proteins, which contribute to proper and effective tissue regeneration.
Collapse
Affiliation(s)
- Ivan Dimauro
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Loredana Grasso
- Unit of Internal Medicine, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Simona Fittipaldi
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Cristina Fantini
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Neri Mercatelli
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Silvia Racca
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Di Gianfrancesco
- Unit of Internal Medicine, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Daniela Caporossi
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
- * E-mail:
| | - Fabio Pigozzi
- Unit of Internal Medicine, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Paolo Borrione
- Unit of Internal Medicine, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| |
Collapse
|