1
|
Lv R, Zhao Y, Wang X, He Y, Dong N, Min X, Liu X, Yu Q, Yuan K, Yue H, Yin Q. GLP-1 analogue liraglutide attenuates CIH-induced cognitive deficits by inhibiting oxidative stress, neuroinflammation, and apoptosis via the Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Int Immunopharmacol 2024; 142:113222. [PMID: 39321702 DOI: 10.1016/j.intimp.2024.113222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Obstructive sleep apnea (OSA) is a common clinical condition linked to cognitive impairment, mainly characterized by chronic intermittent hypoxia (CIH). GLP-1 receptor agonist, known for promoting insulin secretion and reducing glucose levels, has demonstrated neuroprotective effects in various experimental models such as stroke, Alzheimer's disease, and Parkinson's disease. This study aims to investigate the potential role and mechanisms of the GLP-1 receptor agonist liraglutide in ameliorating OSA-induced cognitive deficits. CIH exposure, a well-established and mature OSA pathological model, was used both in vitro and in vivo. In vitro, CIH significantly activated oxidative stress, inflammation, and apoptosis in SH-SY5Y cells. Liraglutide enhanced the nuclear translocation of Nrf2, activating its downstream pathways, thereby mitigating CIH-induced injury in SH-SY5Y cells. Additionally, liraglutide modulated the MAPK/NF-κB signaling pathway, reducing the expression of inflammatory factors and proteins. In vivo, we subjected mice to an intermittent hypoxia incubator to mimic the pathogenesis of human OSA. The Morris water maze test revealed that CIH exposure substantially impaired spatial memory. Subsequent western blot analyses and histopathological examinations indicated that liraglutide could activate the Nrf2/HO-1 axis and inhibit the MAPK/NF-κB signaling pathway, thereby alleviating OSA-associated cognitive dysfunction in mice. These findings suggest that GLP-1 receptor agonists may offer a promising preventive strategy for OSA-associated cognitive impairment. By refining these findings, we provide new insights into GLP-1's protective mechanisms in combating cognitive deficits associated with CIH, underscoring its potential as a therapeutic agent for conditions linked to OSA.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiangzhen Min
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xueying Liu
- Jinan Third People's Hospital, Jinan, Shandong 250132, China
| | - Qin Yu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| |
Collapse
|
2
|
Basova LV, Riley T, Franklin D, Delorme-Walker V, Lim WL, Grant I, Letendre SL, Iudicello JE, Cherner M, Ellis RJ, Marcondes MCG. Identifying methamphetamine use predictors in HIV infection: Immune-dopaminergic signatures in peripheral leukocytes and the role of COMT genotype. Brain Behav Immun Health 2024; 42:100873. [PMID: 39430881 PMCID: PMC11490913 DOI: 10.1016/j.bbih.2024.100873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/24/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
The pursuit of translational biomarkers is complex due to the heterogeneous human pathophysiology, but critical for disease diagnosis, prognosis, monitoring therapeutic efficacy, and for patient stratification. In HIV-associated neurocognitive impairment (NCI), biomarkers that delineate the trajectory of neuropathogenesis and neurocognitive sequelae are critical, particularly considering confounders such as substance use, including Methamphetamine (METH). METH use is a significant health concern among persons living with HIV (PWH), aggravating cognitive deficits and neuroinflammation despite of antiretrovirals, introducing elements in the microenvironment that are fundamentally differerent in relation to non-METH users, such as high levels of dopamine (DA) affecting HIV-innate immune targets. Yet, current biomarkers do not detect these differences. We hypothesized that predefined DA-induced signatures detectable in peripheral blood leukocytes, can distinguish HIV+ METH users compared to HIV-negative or PWH that are non METH users. The elevated expression of CD8A, CREBBP, CCL5, and combinations of dopaminergic pathway transcripts clustered METH users with detectable CSF viral load and major depressive disorder (MDD), indicating neuroimmune-mechanistic links. Cathecol-o-methyltransferase (COMT) gene polymorphisms affecting DA metabolism improved the identification of PWH using METH with biomarkers. The results indicate that underlying immunedopaminergic mechanisms provide signatures and genotypes that can identify PWH that are METH users and their attributes.
Collapse
Affiliation(s)
- Liana V. Basova
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
| | - Tera Riley
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
- National Institute for Drug Abuse, Summer Internship, 2023, USA
| | - Donald Franklin
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | | | - Wei Ling Lim
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
| | - Igor Grant
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Scott L. Letendre
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Jennifer E. Iudicello
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Mariana Cherner
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Ronald J. Ellis
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | | |
Collapse
|
3
|
Kazemifard N, Farmani M, Baradaran Ghavami S, Kazemi M, Shahrokh S, Asadzadeh Aghdaei H, Zali M. A prediction of the CRNDE role by modulating NF-κB pathway in inflammatory bowel disease (IBD). Biochem Biophys Rep 2024; 38:101731. [PMID: 38766384 PMCID: PMC11101873 DOI: 10.1016/j.bbrep.2024.101731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) regulate multiple pathways and cellular mechanisms. Recent research has emphasized their involvement in the pathogenesis of complex diseases, such as Inflammatory Bowel Disease (IBD) which is characterized by chronic inflammation of the intestines. The two most common types of IBD are ulcerative colitis and Crohn's disease. CRNDE lncRNA was initially detected in colorectal cancer (CRC) and found to be involved in the tumorigenesis pathways. Further studies revealed the role of CRNDE in activating inflammation and promoting the release of inflammatory cytokines. This study utilizes the RNA-seq data analysis and bioinformatics tools to clarify the role of CRNDE in the IBD pathogenesis and confirms its expression in inflamed HT-29 and Caco-2 cell lines and also colonic and blood samples of UC patients and controls ex vivo. Based on our results, CRNDE was significantly upregulated in IBD samples compared to controls in RNA-seq data analysis and Real-time PCR of inflamed HT-29 cell line and colonic biopsies from UC patients. Additionally, predicted that its expression is positively correlated with the pro-inflammatory cytokines production. CRNDE interactions was investigated with several inflammation-related miRNAs and regulatory proteins computationally. Thus, CRNDE upregulation in the colon of IBD patients could be involved in IBD pathogenesis by promoting inflammatory pathways and targeting anti-inflammatory miRNAs.
Collapse
Affiliation(s)
- Nesa Kazemifard
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Farmani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Baradaran Ghavami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Reproductive Sciences and Sexual Health Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shabnam Shahrokh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Nemoto H, Sakai D, Watson D, Masuda K. Nuclear Factor-κB Decoy Oligodeoxynucleotide Attenuates Cartilage Resorption In Vitro. Bioengineering (Basel) 2024; 11:46. [PMID: 38247922 PMCID: PMC10813736 DOI: 10.3390/bioengineering11010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Cartilage harvest and transplantation is a common surgery using costal, auricular, and septal cartilage for craniofacial reconstruction. However, absorption and warping of the cartilage grafts can occur due to inflammatory factors associated with wound healing. Transcription factor nuclear factor-κB (NF-κB) is activated by the various stimulation such as interleukin-1 (IL-1), and plays a central role in the transactivation of this inflammatory cytokine gene. Inhibition of NF-κB may have anti-inflammatory effects. The aim of this study was to explore the potential of an NF-κB decoy oligodeoxynucleotide (Decoy) as a chondroprotective agent. MATERIALS AND METHODS Safe and efficacious concentrations of Decoy were assessed using rabbit nasal septal chondrocytes (rNSChs) and assays for cytotoxicity, proteoglycan (PG) synthesis, and PG turnover were carried out. The efficacious concentration of Decoy determined from the rNSChs was then applied to human nasal septal cartilage (hNSC) in vitro and analyzed for PG turnover, the levels of inflammatory markers, and catabolic enzymes in explant-conditioned culture medium. RESULTS Over the range of Decoy conditions and concentrations, no inhibition of PG synthesis or cytotoxicity was observed. Decoy at 10 μM effectively inhibited PG degradation in the hNSC explant, prolonging PG half-life by 63% and decreasing matrix metalloprotease 3 (MMP-3) by 70.7% (p = 0.027). CONCLUSIONS Decoy may be considered a novel chondroprotective therapeutic agent in cartilage transplantation due to its ability to inhibit cartilage degradation due to inflammation cytokines.
Collapse
Affiliation(s)
- Hitoshi Nemoto
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA
- Department of Plastic Surgery, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA; (D.S.); (K.M.)
- Department of Orthopaedic Surgery, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan
| | - Deborah Watson
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA
| | - Koichi Masuda
- Department of Orthopaedic Surgery, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA; (D.S.); (K.M.)
| |
Collapse
|
5
|
Yuan M, Feng Y, Zhao M, Xu T, Li L, Guo K, Hou D. Identification and verification of genes associated with hypoxia microenvironment in Alzheimer's disease. Sci Rep 2023; 13:16252. [PMID: 37759083 PMCID: PMC10533856 DOI: 10.1038/s41598-023-43595-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
As the incidence of Alzheimer's disease (AD) increases year by year, more people begin to study this disease. In recent years, many studies on reactive oxygen species (ROS), neuroinflammation, autophagy, and other fields have confirmed that hypoxia is closely related to AD. However, no researchers have used bioinformatics methods to study the relationship between AD and hypoxia. Therefore, our study aimed to screen the role of hypoxia-related genes in AD and clarify their diagnostic significance. A total of 7681 differentially expressed genes (DEGs) were identified in GSE33000 by differential expression analysis and cluster analysis. Weighted gene co-expression network analysis (WGCNA) was used to detect 9 modules and 205 hub genes with high correlation coefficients. Next, machine learning algorithms were applied to 205 hub genes and four key genes were selected. Through the verification of external dataset and quantitative real-time PCR (qRT-PCR), the AD diagnostic model was established by ANTXR2, BDNF and NFKBIA. The bioinformatics analysis results suggest that hypoxia-related genes may increase the risk of AD. However, more in-depth studies are still needed to investigate their association, this article would guide the insights and directions for further research.
Collapse
Affiliation(s)
- Mingyang Yuan
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Yanjin Feng
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Mingri Zhao
- School of Life Sciences, Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, 410000, China
| | - Ting Xu
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Liuhong Li
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Ke Guo
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Deren Hou
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China.
| |
Collapse
|
6
|
Mulligan EA, Tudhope SJ, Hunter JE, Clift AEG, Elliott SL, Summerfield GP, Wallis J, Pepper CJ, Durkacz B, Veuger S, Willmore E. Expression and Activity of the NF-κB Subunits in Chronic Lymphocytic Leukaemia: A Role for RelB and Non-Canonical Signalling. Cancers (Basel) 2023; 15:4736. [PMID: 37835430 PMCID: PMC10571822 DOI: 10.3390/cancers15194736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Canonical NF-κB signalling by p65 (RelA) confers chemo-resistance and poor survival in chronic lymphocytic leukaemia (CLL). The role of non-canonical NF-κB signalling (leading to RelB and p52 subunit activation) in CLL is less understood, but given its importance in other B-cell tumour types, we theorised that RelB and p52 may also contribute to the pathology of CLL. METHODS DNA binding activity of all five NF-kB subunits, p65, p50, RelB, p52, and c-Rel, was quantified using ELISA and correlated to ex vivo chemoresistance, CD40L-stimulated signalling (to mimic the lymph node microenvironment), and clinical data. RESULTS Importantly, we show for the first time that high basal levels of RelB DNA binding correlate with nuclear RelB protein expression and are associated with del(11q), ATM dysfunction, unmutated IGHV genes, and shorter survival. High levels of nuclear p65 are prevalent in del(17p) cases (including treatment-naïve patients) and also correlate with the outcome. CD40L-stimulation resulted in rapid RelB activation, phosphorylation and processing of p100, and subsequent CLL cell proliferation. CONCLUSIONS These data highlight a role for RelB in driving CLL cell tumour growth in a subset of patients and therefore strategies designed to inhibit non-canonical NF-κB signalling represent a novel approach that will have therapeutic benefit in CLL.
Collapse
Affiliation(s)
- Evan A. Mulligan
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Susan J. Tudhope
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jill E. Hunter
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Arabella E. G. Clift
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Sarah L. Elliott
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | | | - Jonathan Wallis
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Chris J. Pepper
- Medical Research Building, Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9PX, UK
| | - Barabara Durkacz
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Stephany Veuger
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE7 7XA, UK
| | - Elaine Willmore
- Cancer Research UK Drug Discovery Unit, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
7
|
Nicolò S, Antonelli A, Tanturli M, Baccani I, Bonaiuto C, Castronovo G, Rossolini GM, Mattiuz G, Torcia MG. Bacterial Species from Vaginal Microbiota Differently Affect the Production of the E6 and E7 Oncoproteins and of p53 and p-Rb Oncosuppressors in HPV16-Infected Cells. Int J Mol Sci 2023; 24:ijms24087173. [PMID: 37108333 PMCID: PMC10138431 DOI: 10.3390/ijms24087173] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Vaginal dysbiosis is characterized by a decrease in the relative abundance of Lactobacillus species in favor of other species. This condition facilitates infections by sexually transmitted pathogens including high risk (HR)-human papilloma viruses (HPVs) involved in the development of cervical cancer. Some vaginal dysbiosis bacteria contribute to the neoplastic progression by inducing chronic inflammation and directly activating molecular pathways involved in carcinogenesis. In this study, SiHa cells, an HPV-16-transformed epithelial cell line, were exposed to different representative vaginal microbial communities. The expression of the HPV oncogenes E6 and E7 and the production of relative oncoproteins was evaluated. The results showed that Lactobacillus crispatus and Lactobacillus gasseri modulated the basal expression of the E6 and E7 genes of SiHa cells and the production of the E6 and E7 oncoproteins. Vaginal dysbiosis bacteria had contrasting effects on E6/E7 gene expression and protein production. The expression of the E6 and E7 genes and the production of the relative oncoproteins was increased by strains of Gardnerella vaginalis and, to a lesser extent, by Megasphaera micronuciformis. In contrast, Prevotella bivia decreased the expression of oncogenes and the production of the E7 protein. A decreased amount of p53 and pRb was found in the cultures of SiHa cells with M. micronuciformis, and accordingly, in the same cultures, a higher percentage of cells progressed to the S-phase of the cell cycle compared to the untreated or Lactobacillus-stimulated cultures. These data confirm that L. crispatus represents the most protective component of the vaginal microbiota against neoplastic progression of HR-HPV infected cells, while M. micronuciformis and, to a lesser extent, G. vaginalis may directly interfere in the oncogenic process, inducing or maintaining the production of viral oncoproteins.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Alberto Antonelli
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50139 Florence, Italy
| | - Michele Tanturli
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Ilaria Baccani
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50139 Florence, Italy
| | - Chiara Bonaiuto
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50139 Florence, Italy
| | - Giuseppe Castronovo
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Gian Maria Rossolini
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50139 Florence, Italy
| | - G Mattiuz
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - M G Torcia
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
8
|
Deka K, Li Y. Transcriptional Regulation during Aberrant Activation of NF-κB Signalling in Cancer. Cells 2023; 12:788. [PMID: 36899924 PMCID: PMC10001244 DOI: 10.3390/cells12050788] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The NF-κB signalling pathway is a major signalling cascade involved in the regulation of inflammation and innate immunity. It is also increasingly recognised as a crucial player in many steps of cancer initiation and progression. The five members of the NF-κB family of transcription factors are activated through two major signalling pathways, the canonical and non-canonical pathways. The canonical NF-κB pathway is prevalently activated in various human malignancies as well as inflammation-related disease conditions. Meanwhile, the significance of non-canonical NF-κB pathway in disease pathogenesis is also increasingly recognized in recent studies. In this review, we discuss the double-edged role of the NF-κB pathway in inflammation and cancer, which depends on the severity and extent of the inflammatory response. We also discuss the intrinsic factors, including selected driver mutations, and extrinsic factors, such as tumour microenvironment and epigenetic modifiers, driving aberrant activation of NF-κB in multiple cancer types. We further provide insights into the importance of the interaction of NF-κB pathway components with various macromolecules to its role in transcriptional regulation in cancer. Finally, we provide a perspective on the potential role of aberrant NF-κB activation in altering the chromatin landscape to support oncogenic development.
Collapse
Affiliation(s)
- Kamalakshi Deka
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore 138673, Singapore
| |
Collapse
|
9
|
Spoerl S, Erber R, Gerken M, Taxis J, Ludwig N, Nieberle F, Biermann N, Geppert CI, Ettl T, Hartmann A, Beckhove P, Reichert TE, Spanier G, Spoerl S. A20 as a Potential New Tool in Predicting Recurrence and Patient's Survival in Oral Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15030675. [PMID: 36765630 PMCID: PMC9913673 DOI: 10.3390/cancers15030675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
A20, known as a potent inhibitor of NF-κB signaling, has been characterized in numerous clinical as well as preclinical studies. Recently, especially in various malignant diseases, the prognostic and therapeutic relevance of A20 was investigated. In oral squamous cell carcinoma (OSCC) however, the characterization of A20 is uncharted territory. We analyzed a tissue microarray (TMA) of 229 surgically-treated OSCC patients (2003-2013). Immunohistochemical (IHC) stainings were performed for A20 and CD3; additionally, standard haematoxylin-eosin staining was applied. IHC findings were correlated with a comprehensive dataset, comprising clinical and pathohistological information. A20 expression was analyzed in tumor cells as well as in tumor infiltrating lymphocytes (TILs) and correlated with the overall survival (OS) and recurrence-free survival (RFS) using uni- and multivariable Cox regression. The median follow-up time was 10.9 years and the A20 expression was significantly decreased in CD3+ TILs compared to mucosa-infiltrating lymphocytes (MILs). In the Kaplan-Meier analyses, higher A20 expression in TILs was correlated with better OS (p = 0.017) and RFS (p = 0.020). In the multivariable survival analysis, A20 overexpression correlated with improved OS (HR: 0.582; 95% CI 0.388-0.873, p = 0.009) and RFS (HR 0.605; 95% CI 0.411-0.889, p = 0.011). Our results indicate a novel prognostic role for A20 in OSCC. Due to its elevated expression in TILs, further research is highly desirable, which therefore could offer new therapeutic opportunities for patients suffering from OSCC.
Collapse
Affiliation(s)
- Steffen Spoerl
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-944-6340; Fax: +49-941-944-6342
| | - Ramona Erber
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
| | - Michael Gerken
- Tumor Center, Institute for Quality Management and Health Services Research, University of Regensburg, 93053 Regensburg, Germany
| | - Juergen Taxis
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nils Ludwig
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Felix Nieberle
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Niklas Biermann
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Carol Immanuel Geppert
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
| | - Tobias Ettl
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
| | - Philipp Beckhove
- Division of Interventional Immunology, Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Torsten E. Reichert
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gerrit Spanier
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Silvia Spoerl
- Department of Internal Medicine 5—Haematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
10
|
Zhuo C, Ruan Q, Zhao X, Shen Y, Lin R. CXCL1 promotes colon cancer progression through activation of NF-κB/P300 signaling pathway. Biol Direct 2022; 17:34. [PMID: 36434686 PMCID: PMC9701058 DOI: 10.1186/s13062-022-00348-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The upregulated expression of CXCL1 has been validated in colorectal cancer patients. As a potential biotherapeutic target for colorectal cancer, the mechanism by which CXCL1 affects the development of colorectal cancer is not clear. METHODS Expression data of CXCL1 in colorectal cancer were obtained from the GEO database and verified using the GEPIA database and the TIMER 2.0 database. Knockout and overexpression of CXCL1 in colorectal cancer cells by CRISPR/Cas and "Sleeping Beauty" transposon-mediated gene editing techniques. Cell biological function was demonstrated by CCK-8, transwell chamber and Colony formation assay. RT-qPCR and Western Blot assays measured RNA and protein expression. Protein localization and expression were measured by immunohistochemistry and immunofluorescence. RESULTS Bioinformatics analysis showed significant overexpression of CXCL1 in the colorectal cancer tissues compared to normal human tissues, and identified CXCL1 as a potential therapeutic target for colorectal cancer. We demonstrate that CXCL1 promotes the proliferation and migration of colon cancer cells and has a facilitative effect on tumor angiogenesis. Furthermore, CXCL1 elevation promoted the migration of M2-tumor associated macrophages (TAMs) while disrupting the aggregation of CD4+ and CD8+ T cells at tumor sites. Mechanistic studies suggested that CXCL1 activates the NF-κB pathway. In the in vivo colon cancer transplantation tumor model, treatment with the P300 inhibitor C646 significantly inhibited the growth of CXCL1-overexpressing colon cancer. CONCLUSION CXCL1 promotes colon cancer development through activation of NF-κB/P300, and that CXCL1-based therapy is a potential novel strategy to prevent colon cancer development.
Collapse
Affiliation(s)
- Changhua Zhuo
- grid.415110.00000 0004 0605 1140Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014 People’s Republic of China ,grid.411604.60000 0001 0130 6528Fuzhou University, College of Chemistry, Fuzhou, 350108 People’s Republic of China ,Fujian Key Laboratory of Translational Cancer Medicine and Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian 350014 People’s Republic of China
| | - Qiang Ruan
- grid.411604.60000 0001 0130 6528Fuzhou University, College of Chemistry, Fuzhou, 350108 People’s Republic of China
| | - Xiangqian Zhao
- grid.411503.20000 0000 9271 2478Fujian Normal University Qishan Campus, College of Life Science, Biomedical Research Center of South China, Fuzhou, 350117 People’s Republic of China
| | - Yangkun Shen
- grid.411503.20000 0000 9271 2478Fujian Normal University Qishan Campus, College of Life Science, Biomedical Research Center of South China, Fuzhou, 350117 People’s Republic of China
| | - Ruirong Lin
- grid.415110.00000 0004 0605 1140Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014 People’s Republic of China ,grid.411604.60000 0001 0130 6528Fuzhou University, College of Chemistry, Fuzhou, 350108 People’s Republic of China ,Fujian Key Laboratory of Translational Cancer Medicine and Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian 350014 People’s Republic of China
| |
Collapse
|
11
|
Wang Y, Liu Z, Ma J, Xv Q, Gao H, Yin H, Yan G, Jiang X, Yu W. Lycopene attenuates the inflammation and apoptosis in aristolochic acid nephropathy by targeting the Nrf2 antioxidant system. Redox Biol 2022; 57:102494. [PMID: 36198206 PMCID: PMC9530962 DOI: 10.1016/j.redox.2022.102494] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 10/31/2022] Open
Abstract
Lycopene (LYC) is a carotenoid, has antioxidant properties. This study investigated whether lycopene attenuates aristolochic acids (AAs) -induced chronic kidney disease. In this experiment, lycopene was used to intervene C57BL/6 mice with renal injury induced by aristolochic acid exposure. The histomorphological changes and serological parameters of the kidney were measured in order to assess the alleviating effect of lycopene on renal injury in aristolochic acid nephropathy. In vitro and in vivo experiments were carried out to verify the main mechanism of action and drug targets of lycopene in improving aristolochic acid nephropathy (AAN) and by various experimental methods such as ELISA, immunohistochemistry, immunofluorescence, Western-blot and qRT-PCR. The results showed that oxidative stress injury was induced in the kidney of mice after AAI exposure, resulting in inflammatory response and tubular epithelial cell apoptosis. The results showed that the Nrf2/HO-1 antioxidant signaling pathway was inhibited after AAI exposure. AAI induces oxidative stress injury in the kidney, which ultimately leads to inflammation and tubular epithelial cell apoptosis. After LYC intervened in the body, it activated Nrf2 nuclear translocation and its downstream HO-1 and NQO1 antioxidant signaling pathways. LYC inhibited ROS production by renal tubular epithelial cells, and alleviated mitochondrial damage. LYC further modulated the TNF-α/NF-κB signaling cascade, thereby reduced the accumulation of inflammatory factors in the renal interstitium. Moreover, LYC was able to up-regulate the expression of Bcl-2, down-regulate Bax expression and inhibit the activation of cleaved forms of Caspase-9 and Caspase-3, which finally attenuated the apoptosis of the mitochondrial pathway induced by AAI exposure. It was concluded that lycopene was able to activate the Nrf2 antioxidant signaling pathway to maintain the homeostasis of renal oxidative stress and ultimately attenuated renal inflammatory response and apoptosis. These results suggested that lycopene can be used as a drug to relieve AAN.
Collapse
Affiliation(s)
- Yu Wang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Zhihui Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jun Ma
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Qingyang Xv
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hongxin Gao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hang Yin
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ge Yan
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China; Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
12
|
Deng Y, Feng Y, Lv Z, He J, Chen X, Wang C, Yuan M, Xu T, Gao W, Chen D, Zhu H, Hou D. Machine learning models identify ferroptosis-related genes as potential diagnostic biomarkers for Alzheimer’s disease. Front Aging Neurosci 2022; 14:994130. [PMID: 36262887 PMCID: PMC9575464 DOI: 10.3389/fnagi.2022.994130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex, and multifactorial neurodegenerative disease. Previous studies have revealed that oxidative stress, synaptic toxicity, autophagy, and neuroinflammation play crucial roles in the progress of AD, however, its pathogenesis is still unclear. Recent researches have indicated that ferroptosis, an iron-dependent programmed cell death, might be involved in the pathogenesis of AD. Therefore, we aim to screen correlative ferroptosis-related genes (FRGs) in the progress of AD to clarify insights into the diagnostic value. Interestingly, we identified eight FRGs were significantly differentially expressed in AD patients. 10,044 differentially expressed genes (DEGs) were finally identified by differential expression analysis. The following step was investigating the function of DEGs using gene set enrichment analysis (GSEA). Weight gene correlation analysis was performed to explore ten modules and 104 hub genes. Subsequently, based on machine learning algorithms, we constructed diagnostic classifiers to select characteristic genes. Through the multivariable logistic regression analysis, five features (RAF1, NFKBIA, MOV10L1, IQGAP1, FOXO1) were then validated, which composed a diagnostic model of AD. Thus, our findings not only developed genetic diagnostics strategy, but set a direction for further study of the disease pathogenesis and therapy targets.
Collapse
Affiliation(s)
- Yanyao Deng
- Department of Rehabilitation, The First Hospital of Changsha, Changsha, China
| | - Yanjin Feng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Lv
- Department of Neurosurgery, The First People’s Hospital of Chenzhou, Chenzhou, China
| | - Jinli He
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xun Chen
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chen Wang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyang Yuan
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ting Xu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenzhe Gao
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongjie Chen
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Zhu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Deren Hou
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Deren Hou,
| |
Collapse
|
13
|
Zhang T, Ding C, Chen H, Zhao J, Chen Z, Chen B, Mao K, Hao Y, Roulis M, Xu H, Kluger Y, Zou Q, Ye Y, Zhan M, Flavell RA, Li HB. m 6A mRNA modification maintains colonic epithelial cell homeostasis via NF-κB-mediated antiapoptotic pathway. SCIENCE ADVANCES 2022; 8:eabl5723. [PMID: 35333576 PMCID: PMC8956260 DOI: 10.1126/sciadv.abl5723] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/02/2022] [Indexed: 05/26/2023]
Abstract
Colonic mucosal barrier dysfunction is one of the major causes of inflammatory bowel disease (IBD). However, the mechanisms underlying mucosal barrier dysfunction are poorly understood. N6-methyladenosine (m6A) mRNA modification is an important modulator of epitranscriptional regulation of gene expression, participating in multiple physiological and pathological processes. However, the function of m6A modification in colonic epithelial cells and stem cells is unknown. Here, we show that m6A modification is essential for maintaining the homeostatic self-renewal in colonic stem cells. Specific deletion of the methyltransferase 14 (Mettl14) gene in mouse colon resulted in colonic stem cell apoptosis, causing mucosal barrier dysfunction and severe colitis. Mechanistically, we revealed that Mettl14 restricted colonic epithelial cell death by regulating the stability of Nfkbia mRNA and modulating the NF-κB pathway. Our results identified a previously unidentified role for m6A modification in colonic epithelial cells and stem cells, suggesting that m6A modification may be a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Ting Zhang
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Renji Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Chenbo Ding
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huifang Chen
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Zhao
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| | - Zhejun Chen
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Baiwen Chen
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kaiqiong Mao
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yajuan Hao
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Manolis Roulis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| | - Hao Xu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| | - Qiang Zou
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meixiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital of Jinan University, Zhuhai, Guangdong 519000, China
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Jiao Tong University School of Medicine–Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520-8055, USA
| |
Collapse
|
14
|
Xu W, Wang B, Gao Y, Cai Y, Zhang J, Wu Z, Wei J, Guo C, Yuan C. Alkaloids exhibit a meaningful function as anticancer agents by restraining cellular signaling pathways. Mini Rev Med Chem 2021; 22:968-983. [PMID: 34620048 DOI: 10.2174/1389557521666211007114935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/07/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
Alkaloids are nitrogen-containing organic compounds widely found in natural products, which play an essential role in clinical treatment. Cellular signaling pathways in tumors are a series of enzymatic reaction pathways that convert extracellular signals into intracellular signals to produce biological effects. The ordered function of cell signaling pathways is essential for tumor cell proliferation, differentiation, and programmed death. This review describes the antitumor progression mediated by various alkaloids after inhibiting classical signaling pathways; related studies are systematically retrieved and collected through PubMed. We selected the four currently most popular pathways for discussion and introduced the molecular mechanisms mediated by alkaloids in different signaling pathways, including the NF-kB signaling pathway, PI3K/AKT signaling pathway, MAPK signaling pathway, and P53 signaling pathway. The research progress of alkaloids related to tumor signal transduction pathways and the realization of alkaloids as cancer prevention drugs by targeting signal pathways remains.
Collapse
Affiliation(s)
- Wen Xu
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Bei Wang
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yisong Gao
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yuxuan Cai
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Jiali Zhang
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Zhiyin Wu
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Jiameng Wei
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Chong Guo
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| |
Collapse
|
15
|
|
16
|
Chu J, Gao J, Wang J, Li L, Chen G, Dang J, Wang Z, Jin Z, Liu X. Mechanism of hydrogen on cervical cancer suppression revealed by high‑throughput RNA sequencing. Oncol Rep 2021; 46:141. [PMID: 34080660 PMCID: PMC8165587 DOI: 10.3892/or.2021.8092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/14/2021] [Indexed: 12/09/2022] Open
Abstract
Cervical cancer is considered one of the diseases with the highest mortality among women and with limited treatment options. Hydrogen (H2) inhalation has been reported to have a variety of tumor-suppressive effects, but the exact mechanism remains unclear. In the present study, HeLa cervical cancer cells and HaCaT keratinocytes treated with H2, and a HeLa xenograft mouse model subjected to H2 inhalation were established. TUNEL, Cell Counting Kit-8 and Ki67 staining assays were used to detect cell apoptosis and proliferation. Oxidative stress was determined according to the levels of reactive oxygen species, malondialdehyde and superoxide dismutase. Tumor growth was recorded every 3 days, and the excised tumors were stained with hematoxylin and eosin. High-throughput RNA sequencing and subsequent Gene Ontology (GO) enrichment analysis were performed in HeLa-treated and un-treated HeLa cells. The expression of hypoxia-inducible factor (HIF)-1α and NF-κB p65 was verified by western blotting, immunohistochemistry and reverse transcription-quantitative PCR. The results revealed an increased apoptosis rate, and reduced cell proliferation and oxidative stress in H2-treated HeLa cells but not in HaCaT cells. Similarly, decreased tumor growth and cell proliferation, and enhanced cell apoptosis were observed in H2-treated HeLa tumors. RNA sequencing and GO analysis suggest that downregulated HIF1A (HIF-1α mRNA) and RelA (NF-κB p65) levels, and reduced NF-κB signaling were associated with the antitumor effect of H2. Finally, decreased HIF-1α and NF-κB p65 expression both at the transcriptional and translational levels were observed in H2-treated HeLa cells and in HeLa-derived tumors. In conclusion, the present study reveals a novel mechanism of H2 against cervical cancer, which may serve as a potential therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Jing Chu
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Jinghai Gao
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Jing Wang
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Lingling Li
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Guoqiang Chen
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Jianhong Dang
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Zhifeng Wang
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Zhijun Jin
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| | - Xiaojun Liu
- Department of Obstetrics and Gynaecology, Changzheng Hospital, Naval Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
17
|
Zeng Q, Cai X, Cao Y, Zhou C, Yu L, Chen J. Preparation, characterization, and pharmacodynamic study on deep second degree burns of total flavonoids composite phospholipids liposome gel of Oxytropis falcata Bunge. Drug Dev Ind Pharm 2020; 46:2000-2009. [PMID: 33095085 DOI: 10.1080/03639045.2020.1841787] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Wound healing is the treatment problem after deep second degree (II°) burns. The p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor-κB/inhibitory factor-κB (NF-κB/IκB) signal pathways play significant role in angiogenesis and wound repair after burns.This study aimed to investigate the preparation, characterization and pharmacodynamics of the total flavonoids composite phospholipids liposome of Oxytropis falcata Bunge (TFOFB-CPL) on deep II° burns to research its biological activity and underlying mechanism. The TFOFB-CPL was prepared by thin-film dispersion method and the preparation process was optimized via central composite design. The TFOFB-CPL was then characterized by using particle size, polydispersity indexes (PDIs), zeta potential, encapsulation efficiency (EE) and morphology. Moerover, in vitro transdermal test and in vivo pharmacodynamic study included wound healing rate, hematoxylin-eosin (HE) staining, masson staning, western blotting and RT-PCR. The results showed that the therapeutic effects of TFOFB-CPL gel on deep II° burns, especially during wound healing were significant. TFOFB-CPL gel has a sustained-release effect during the treatment of deep II° burns with forming drug depot in the dermis layer. The wound healing rate of TFOFB-CPL gel group was near positive group and better than the other groups. TFOFB-CPL gel could promote the growth of epidermis, skin appendages, fibrovascular and collagen fibers, and had obvious anti-inflammatory effects. Moreover, TFOFB-CPL gel inhibited the activation of p38MAPK and the degradation of IκBα, and promoted the neonatal wounds during the early stage. Therefore, TFOFB-CPL gel could be considered as a novel preparation for treating deep II° burns.
Collapse
Affiliation(s)
- Qiping Zeng
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Xiaohui Cai
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Yixiang Cao
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Chengfang Zhou
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Le Yu
- Department of Pathology, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Jinshan Chen
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| |
Collapse
|
18
|
Legge DN, Chambers AC, Parker CT, Timms P, Collard TJ, Williams AC. The role of B-Cell Lymphoma-3 (BCL-3) in enabling the hallmarks of cancer: implications for the treatment of colorectal carcinogenesis. Carcinogenesis 2020; 41:249-256. [PMID: 31930327 PMCID: PMC7221501 DOI: 10.1093/carcin/bgaa003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/29/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
With its identification as a proto-oncogene in chronic lymphocytic leukaemia and central role in regulating NF-κB signalling, it is perhaps not surprising that there have been an increasing number of studies in recent years investigating the role of BCL-3 (B-Cell Chronic Lymphocytic Leukaemia/Lymphoma-3) in a wide range of human cancers. Importantly, this work has begun to shed light on our mechanistic understanding of the function of BCL-3 in tumour promotion and progression. Here, we summarize the current understanding of BCL-3 function in relation to the characteristics or traits associated with tumourigenesis, termed ‘Hallmarks of Cancer’. With the focus on colorectal cancer, a major cause of cancer related mortality in the UK, we describe the evidence that potentially explains why increased BCL-3 expression is associated with poor prognosis in colorectal cancer. As well as promoting tumour cell proliferation, survival, invasion and metastasis, a key emerging function of this proto-oncogene is the regulation of the tumour response to inflammation. We suggest that BCL-3 represents an exciting new route for targeting the Hallmarks of Cancer; in particular by limiting the impact of the enabling hallmarks of tumour promoting inflammation and cell plasticity. As BCL-3 has been reported to promote the stem-like potential of cancer cells, we suggest that targeting BCL-3 could increase the tumour response to conventional treatment, reduce the chance of relapse and hence improve the prognosis for cancer patients.
Collapse
Affiliation(s)
- Danny N Legge
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - Adam C Chambers
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - Christopher T Parker
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - Penny Timms
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - Tracey J Collard
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - Ann C Williams
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, UK
| |
Collapse
|
19
|
Bellucci A, Bubacco L, Longhena F, Parrella E, Faustini G, Porrini V, Bono F, Missale C, Pizzi M. Nuclear Factor-κB Dysregulation and α-Synuclein Pathology: Critical Interplay in the Pathogenesis of Parkinson's Disease. Front Aging Neurosci 2020; 12:68. [PMID: 32265684 PMCID: PMC7105602 DOI: 10.3389/fnagi.2020.00068] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
The loss of dopaminergic neurons of the nigrostriatal system underlies the onset of the typical motor symptoms of Parkinson's disease (PD). Lewy bodies (LB) and Lewy neurites (LN), proteinaceous inclusions mainly composed of insoluble α-synuclein (α-syn) fibrils are key neuropathological hallmarks of the brain of affected patients. Compelling evidence supports that in the early prodromal phases of PD, synaptic terminal and axonal alterations initiate and drive a retrograde degeneration process culminating with the loss of nigral dopaminergic neurons. This notwithstanding, the molecular triggers remain to be fully elucidated. Although it has been shown that α-syn fibrillary aggregation can induce early synaptic and axonal impairment and cause nigrostriatal degeneration, we still ignore how and why α-syn fibrillation begins. Nuclear factor-κB (NF-κB) transcription factors, key regulators of inflammation and apoptosis, are involved in the brain programming of systemic aging as well as in the pathogenesis of several neurodegenerative diseases. The NF-κB family of factors consists of five different subunits (c-Rel, p65/RelA, p50, RelB, and p52), which combine to form transcriptionally active dimers. Different findings point out a role of RelA in PD. Interestingly, the nuclear content of RelA is abnormally increased in nigral dopamine (DA) neurons and glial cells of PD patients. Inhibition of RelA exert neuroprotection against (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) MPTP and 1-methyl-4-phenylpyridinium (MPP+) toxicity, suggesting that this factor decreases neuronal resilience. Conversely, the c-Rel subunit can exert neuroprotective actions. We recently described that mice deficient for c-Rel develop a PD-like motor and non-motor phenotype characterized by progressive brain α-syn accumulation and early synaptic changes preceding the frank loss of nigrostriatal neurons. This evidence supports that dysregulations in this transcription factors may be involved in the onset of PD. This review highlights observations supporting a possible interplay between NF-κB dysregulation and α-syn pathology in PD, with the aim to disclose novel potential mechanisms involved in the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padua, Padua, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Edoardo Parrella
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Bono
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Missale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
20
|
Liu X, Sun J, Yuan P, Shou K, Zhou Y, Gao W, She J, Hu J, Yang J, Yang J. Mfn2 inhibits proliferation and cell-cycle in Hela cells via Ras-NF-κB signal pathway. Cancer Cell Int 2019; 19:197. [PMID: 31384172 PMCID: PMC6664827 DOI: 10.1186/s12935-019-0916-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/19/2019] [Indexed: 01/02/2023] Open
Abstract
Background Mitofusin 2 (Mfn2) is outer membrane protein, as the inhibitor of Ras protein. This study aimed to investigate the effect of Mfn2 on cell proliferation, and cell-cycle in Hela cervical carcinoma cell lines. Methods After treated with Adv-mfn2 or Adv-control for 48 h and 60 h, the RNA and protein of Mfn2 in Hela cells were detected by qRT-PCR and western blot. The immunofluorescence assay was performed to observe the expression and sub-location of Mfn2 in Hela cells. The flow cytometry was performed to detect the cell cycle of Hela cells, while western blots were performed to observe the Ras-NF-κB signal pathway. Then, the xenografted cervix carcinoma mouse model was used to confirm the effect of Mfn2 in Hela cells in vivo and the expression of Ras-NF-κB signaling pathway in vivo. Results In immunofluorescence detection, Mfn2 was located in cytoplasmic, not in the nucleus. In addition, Mfn2 inhibited cell proliferation of Hela cells through reducing PCNA protein expression. Mfn2 induced arrest in G0/G1 phase of the cell cycle in Hela cells. Meanwhile, Mfn2 reduced Cyclin D1 protein expression. Moreover, Mfn2 decreased the Ras signal pathway proteins such as Myc, NF-κB p65, STAT3 in a dose-dependent manner. Then, the in vivo experiment also confirmed that Mfn2 could inhibit the tumor growth, and depress the Cyclin D1, Ras, Myc, NF-κB p65, Erk1/2 and mTOR protein expression. Conclusions Mfn2 could significantly inhibit cell proliferation in Hela cells. It might be acted as an potential anti-cancer target through inducing cell cycle arrest in human cervical carcinoma cells.
Collapse
Affiliation(s)
- Xiaowen Liu
- 1Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical College, China Three Gorges University, Yichang, China.,2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Jun Sun
- 3Department of Biochemistry and Molecular Biology, Basic Medical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yuan
- 3Department of Biochemistry and Molecular Biology, Basic Medical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kangquan Shou
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Yuanhong Zhou
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Wenqi Gao
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Jin She
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Jun Hu
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Jun Yang
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| | - Jian Yang
- 2Institute of Cardiovascular Research & Department of Center Experiment Laboratory, the First College of Clinical Medical Science, China Three Gorges University, Yichang, 44300 China
| |
Collapse
|
21
|
Prasad AS, Bao B. Molecular Mechanisms of Zinc as a Pro-Antioxidant Mediator: Clinical Therapeutic Implications. Antioxidants (Basel) 2019; 8:antiox8060164. [PMID: 31174269 PMCID: PMC6617024 DOI: 10.3390/antiox8060164] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/21/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
The essentiality of zinc as a trace mineral in human health has been recognized for over five decades. Zinc deficiency, caused by diet, genetic defects, or diseases, can cause growth retardation, delayed sexual maturation, depressed immune response, and abnormal cognitive functions in humans. Zinc supplementation in zinc-deficient individuals can overcome or attenuate these abnormalities, suggesting zinc is an essential micro-nutrient in the body. A large number of in vitro and in vivo experimental studies indicate that zinc deficiency also causes apoptosis, cellular dysfunction, deoxyribonucleic acid (DNA) damage, and depressed immune response. Oxidative stress, due to the imbalance of reactive oxygen species (ROS) production and detoxification in the anti-oxidant defense system of the body, along with subsequent chronic inflammation, is believed to be associated with many chronic degenerative diseases such as diabetes, heart diseases, cancers, alcohol-related disease, macular degenerative disease, and neuro-pathogenesis. A large number of experimental studies including cell culture, animal, and human clinical studies have provided supportive evidence showing that zinc acts as an anti-oxidative stress agent by inhibition of oxidation of macro-molecules such as (DNA)/ribonucleic acid (RNA) and proteins as well as inhibition of inflammatory response, eventually resulting in the down-regulation of (ROS) production and the improvement of human health. In this article, we will discuss the molecular mechanisms of zinc as an anti-oxidative stress agent or mediator in the body. We will also discuss the applications of zinc supplementation as an anti-oxidative stress agent or mediator in human health and disease.
Collapse
Affiliation(s)
- Ananda S Prasad
- Department of Oncology, School of Medicine, Wayne State University and Karmanos Cancer Center, Detroit, MI 48201, USA.
| | - Bin Bao
- Department of Oncology, School of Medicine, Wayne State University and Karmanos Cancer Center, Detroit, MI 48201, USA.
| |
Collapse
|
22
|
Induction of the Epithelial-to-Mesenchymal Transition of Human Colorectal Cancer by Human TNF-β (Lymphotoxin) and its Reversal by Resveratrol. Nutrients 2019; 11:nu11030704. [PMID: 30917533 PMCID: PMC6471988 DOI: 10.3390/nu11030704] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Tumor necrosis factor-beta (TNF-β), as an inflammatory mediator that has been shown to promote tumorigenesis, induces NF-κB. Natural multi-targeted agent resveratrol in turn shows anti-inflammatory and anti-cancer properties. Epithelial-to-mesenchymal transition (EMT) allows cancer cells to turn into a motile state with invasive capacities and is associated with metastasis and development of cancer stem cells (CSC). However, TNF-β-induced EMT and the anti-invasion mechanism of resveratrol on CRC are not yet completely understood. Methods: We investigated the underlying molecular mechanisms of resveratrol on TNF-β/TNF-βR-induced EMT and migration of CRC cells (HCT116, RKO, SW480) in monolayer or 3D alginate cultures. Results: TNF-β, similar to TNF-α, induced significant cell proliferation, morphological change, from an epithelial to a spindle-like mesenchymal shape with the formation of filopodia and lamellipodia associated with the expression of EMT parameters (elevated vimentin and slug, reduced E-cadherin), increased migration/invasion, and formation of CSC in all CRC cells. Interestingly, these effects were dramatically decreased in the presence of resveratrol or anti-TNF-βR with TNF-β co-treatment, inducing biochemical changes to the mesenchymal-epithelial transition (MET), with a planar cell surface and suppressed formation of CSC cells. This was associated with a significant increase in apoptosis. Furthermore, we found that resveratrol suppressed TNF-β-induced NF-κB and NF-κB-regulated gene biomarkers associated with growth, proliferation, and invasion. Finally, TNF-βR interacts directly with focal adhesion kinase (FAK) and NF-κB. Conclusion: These results suggest that resveratrol down-regulates TNF-β/TNF-βR-induced EMT, at least in part via specific suppression of NF-κΒ and FAK in CRC cells.
Collapse
|
23
|
Fouani L, Kovacevic Z, Richardson DR. Targeting Oncogenic Nuclear Factor Kappa B Signaling with Redox-Active Agents for Cancer Treatment. Antioxid Redox Signal 2019; 30:1096-1123. [PMID: 29161883 DOI: 10.1089/ars.2017.7387] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Nuclear factor kappa B (NF-κB) signaling is essential under physiologically relevant conditions. However, aberrant activation of this pathway plays a pertinent role in tumorigenesis and contributes to resistance. Recent Advances: The importance of the NF-κB pathway means that its targeting must be specific to avoid side effects. For many currently used therapeutics and those under development, the ability to generate reactive oxygen species (ROS) is a promising strategy. CRITICAL ISSUES As cancer cells exhibit greater ROS levels than their normal counterparts, they are more sensitive to additional ROS, which may be a potential therapeutic niche. It is known that ROS are involved in (i) the activation of NF-κB signaling, when in sublethal amounts; and (ii) high levels induce cytotoxicity resulting in apoptosis. Indeed, ROS-induced cytotoxicity is valuable for its capabilities in killing cancer cells, but establishing the potency of ROS for effective inhibition of NF-κB signaling is necessary. Indeed, some cancer treatments, currently used, activate NF-κB and may stimulate oncogenesis and confer resistance. FUTURE DIRECTIONS Thus, combinatorial approaches using ROS-generating agents alongside conventional therapeutics may prove an effective tactic to reduce NF-κB activity to kill cancer cells. One strategy is the use of thiosemicarbazones, which form redox-active metal complexes that generate high ROS levels to deliver potent antitumor activity. These agents also upregulate the metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), which functions as an NF-κB signaling inhibitor. It is proposed that targeting NF-κB signaling may proffer a new therapeutic niche to improve the efficacy of anticancer regimens.
Collapse
Affiliation(s)
- Leyla Fouani
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
24
|
Myeloid disorders after autoimmune disease. Best Pract Res Clin Haematol 2019; 32:74-88. [PMID: 30927978 DOI: 10.1016/j.beha.2019.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/02/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022]
Abstract
Autoimmune diseases (ADs) are associated with an increased risk not only of lymphoproliferative disorders but also of myeloid malignancies. The excess risk of myelodysplastic syndromes and/or acute myeloid leukemia is observed across several AD types, including systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disorders, multiple sclerosis, among others. The risk of developing myeloid neoplasms (MNs) is dependent on several variables, including the specific AD type, chronicity and severity of the AD, type and duration of exposure of disease modifying anti-rheumatic drugs or cytotoxics/immunosuppressives, and genetic predisposition risk. Putative triggering factors linking AD to elevated MN risk include AD-directed medications, shared genetic susceptibilities between the two disease entities, and chronic immune stimulation or bone marrow infiltration by the AD. Molecular mechanisms underpinning leukemogenesis remain largely speculative and warrant further investigation. Leukemias arising in patients with AD are not always 'therapy-related' in that MNs may develop in certain AD subtypes even among patients with no prior therapy exposure. Only a few studies have attempted to determine factors associated with MN development in AD but failed to demonstrate consistent characteristic clinical or paraclinical features. These reports have failed to demonstrate a clear correlation between individual agent exposure and subsequent leukemia development due to the low rates of therapy exposure compounded by the rarity of MN occurrence. Notwithstanding, the leukemogenic potential is best documented with agents such as azathioprine, cyclophosphamide, and mitoxantrone; this risk of MN development does not appear to be shared by biologic approaches such as anti-tumor necrosis factors-alpha inhibitors. In this article, we discuss plausible biologic mechanisms underlying MN pathogenesis in AD and review the data available on the development of MNs in patients with AD.
Collapse
|
25
|
Knockdown of PHF5A Inhibits Migration and Invasion of HCC Cells via Downregulating NF- κB Signaling. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1621854. [PMID: 30766880 PMCID: PMC6350539 DOI: 10.1155/2019/1621854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/29/2018] [Accepted: 12/23/2018] [Indexed: 02/07/2023]
Abstract
Background Inflammation is the major risk factor for the progression of hepatocellular carcinoma (HCC), and the nuclear factor-κB (NF-κB) signaling plays the central role in the inflammation process. However, the activated mechanism of NF-κB signaling in HCC is unclear. Methods The expression of PHF5A is examined by qPCR, western blotting, and immunohistochemistry (IHC) assay. The potential of PHF5A (PHD-finger domain protein 5a) for migration and invasion is examined by wound healing and Transwell assay. Luciferase reporter assay, western blotting, and qPCR were applied to explore the mechanism by which PHF5A is involved in progression of HCC. Results PHF5A was significantly upregulated in HCC tissues and cells. Downregulation of PHF5A inhibits the migration and invasion of HCC cells. Further study demonstrated that PHF5A is implicated in HCC progression through NF-κB signaling. In addition, blocking the NF-κB signaling can weaken the stimulatory effect of PHF5A on migration and invasion of HCC cells. Conclusion PHF5A expression is upregulated in HCC tissues, and depletion of PHF5A inhibits the migration and invasion of HCC cells. Further experiments demonstrated that PHF5A is implicated in NF-κB signaling and knockdown of PHF5A downregulates the activity of NF-κB pathway to inhibit the tumor progression. The above results provide the evidence that PHF5A plays an indispensable role in progressive effect of NF-κB pathway in HCC and may be a novel therapeutic target of HCC.
Collapse
|
26
|
Ubiquitination and SUMOylation in the chronic inflammatory tumor microenvironment. Biochim Biophys Acta Rev Cancer 2018; 1870:165-175. [DOI: 10.1016/j.bbcan.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022]
|
27
|
Liang N, Sang Y, Liu W, Yu W, Wang X. Anti-Inflammatory Effects of Gingerol on Lipopolysaccharide-Stimulated RAW 264.7 Cells by Inhibiting NF-κB Signaling Pathway. Inflammation 2018; 41:835-845. [PMID: 29508185 DOI: 10.1007/s10753-018-0737-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gingerol was the main functional substance of Zingiberaceous plant which has been known as traditional medicine for thousands of years. The purpose of this experiment was to explore anti-inflammatory effects of gingerol and study the possible mechanism in lipopolysaccharide (LPS)-stimulated RAW246.7 cells. The cells were treated with 10 μg/mL LPS and 300, 200, 100, and 50 μg/mL gingerol for 24 h. The cytotoxicity of gingerol was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-zoliumbromide (MTT) method. Nitric oxide (NO) production was observed using Griess assays. Prostaglandin E2 (PGE2) and pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 have been analyzed by ELISA. Real-time PCR was used to detect the mRNA expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), IL-6, and IL-1β in LPS-induced RAW246.7 cells. Nuclear transcription factor kappa-B (NF-κB) signaling pathway-related proteins have been assessed by western blot assays. The determination of MTT showed that cell viability was not significantly affected by up to 300 μg/mL gingerol. Compared with LPS group, 50, 100, 200, and 300 μg/mL gingerol can inhibit the production of NO and the inhibitory rate was 10.4, 29.1, 58.9, and 62.4%, respectively. The results indicated gingerol existed anti-inflammatory effect. In addition, gingerol also observably inhibited LPS-induced TNF-α, IL-1β, IL-6, and PGE2 (p < 0.01) expression and secretion in a dose-dependent manner. At the genetic level, after the intervention of gingerol, mRNA transcriptions of iNOS, COX-2, IL-6, and IL-1β were all decreased. The protein expressions of iNOS, NF-κB, p-p65, and p-IκB were significantly increased in LPS-induced cells, while these changes were reversed by the treatment with gingerol. This study suggested that gingerol exerts its anti-inflammatory activities in LPS-induced macrophages which can inhibit the production of inflammatory cytokines by targeting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Na Liang
- Faculty of Food Science and Technology, Agricultural University of Hebei, Baoding, 071001, People's Republic of China
| | - Yaxin Sang
- Faculty of Food Science and Technology, Agricultural University of Hebei, Baoding, 071001, People's Republic of China
- Hebei Research Center of Primary Products Processing Technology, Baoding, 071001, People's Republic of China
| | - Weihua Liu
- Faculty of Food Science and Technology, Agricultural University of Hebei, Baoding, 071001, People's Republic of China
- Hebei Research Center of Primary Products Processing Technology, Baoding, 071001, People's Republic of China
| | - Wenlong Yu
- Faculty of Food Science and Technology, Agricultural University of Hebei, Baoding, 071001, People's Republic of China
| | - Xianghong Wang
- Faculty of Food Science and Technology, Agricultural University of Hebei, Baoding, 071001, People's Republic of China.
- Hebei Research Center of Primary Products Processing Technology, Baoding, 071001, People's Republic of China.
| |
Collapse
|
28
|
Momen-Heravi F, Bala S. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogram monocytes via NF-κB pathway. Oncotarget 2018; 9:34838-34854. [PMID: 30410681 PMCID: PMC6205181 DOI: 10.18632/oncotarget.26208] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/21/2018] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) are carriers of different biomacromolecules that participate in cellular signaling and disease pathogenesis. Although it has been shown that EVs can play an active role in cellular communication and different stages of cancer progression, the role of EVs in oral squamous cell carcinoma (OSCC) cancer pathogenesis, especially in the crosstalk of cancer cells with immune cells is unknown. Here, we present a detailed analysis of findings regarding the profile of EVs in OSCC and the role of EVs and associated miRNAs in the crosstalk of malignant cells with monocytes. We demonstrate that EVs are detectable in significantly higher quantities in the plasma of patients with OSCC. Oncogenic miRNAs (such as miR-21, miR-27) were detectable in high quantities in the circulating EVs and plasma of patients with OSCC. EVs isolated from the circulation of OSCC patients and OSCC cell lines showed comparable miRNA signature, indicating the tumor origin of EVs in the circulation of patients with OSCC. Danger signals such as LPS and ethanol increased the production of EVs. EVs were taken up by monocytes after co-culture. Mechanistically, uptake of EVs derived from oral cancer cells by monocytes caused activation of the inflammatory pathway, NF-κB activation, and establishment of a pro-inflammatory and pro-tumorigenic milieu marked by increased levels of IL-6, CCL2, PEG2 and MMP9 levels. Series of experiments involving the introduction of exogenous oncogenic miR-21 mimic induced a similar pro-inflammatory and pro-tumorigenic profile in monocytes. Inhibiting miR-21 function in monocytes attenuated the pro-inflammatory phenotype of monocytes after EV challenge. These results indicate the role of EV-associated miR-21 in modulating the immune response in monocytes.
Collapse
Affiliation(s)
- Fatemeh Momen-Heravi
- Division of Periodontics, Section of Oral and Diagnostic Sciences, College of Dental Medicine, Columbia University, New York, NY, USA
| | - Shashi Bala
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
29
|
Zhang H, Zhang D, Li H, Yan H, Zhang Z, Zhou C, Chen Q, Ye Z, Hang C. Biphasic activation of nuclear factor-κB and expression of p65 and c-Rel following traumatic neuronal injury. Int J Mol Med 2018; 41:3203-3210. [PMID: 29568960 PMCID: PMC5881643 DOI: 10.3892/ijmm.2018.3567] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/22/2018] [Indexed: 01/28/2023] Open
Abstract
The transcription factor nuclear factor-κB (NF-κB) has been shown to function as a key regulator of cell death or survival in neuronal cells. Previous studies indicate that the biphasic activation of NF-κB occurs following experimental neonatal hypoxia-ischemia and subarachnoid hemorrhage. However, the comprehensive understanding of NF-κB activity following traumatic brain injury (TBI) is incomplete. In the current study, an in vitro model of TBI was designed to investigate the NF-κB activity and expression of p65 and c-Rel subunits following traumatic neuronal injury. Primary cultured neurons were assigned to control and transected groups. NF-κB activity was detected by electrophoretic mobility shift assay. Western blotting and immunofluorescence were used to investigate the expression and distribution of p65 and c-Rel. Reverse transcription-quantitative polymerase chain reaction was performed to assess the downstream genes of NF-κB. Lactate dehydrogenase (LDH) quantification and trypan blue staining were used to estimate the neuronal injury. Double peaks of elevated NF-κB activity were observed at 1 and 24 h following transection. The expression levels of downstream genes exhibited similar changes. The protein levels of p65 also presented double peaks while c-Rel was elevated significantly in the late stage. The results of the trypan blue staining and LDH leakage assays indicated there was no sustained neuronal injury during the late peak of NF-κB activity. In conclusion, biphasic activation of NF-κB is induced following experimental traumatic neuronal injury. The elevation of p65 and c-Rel levels at different time periods suggests that within a single neuron, NF-κB may participate in different pathophysiological processes.
Collapse
Affiliation(s)
- Huasheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Dingding Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Hua Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Huiying Yan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Zihuan Zhang
- Department of Neurosurgery, Zhongdu Hospital, Bengbu, Anhui 233004, P.R. China
| | - Chenhui Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiang Chen
- Department of Neurosurgery, Southern Medical University (Guangzhou), Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Zhennan Ye
- Department of Neurosurgery, Southern Medical University (Guangzhou), Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Chunhua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
30
|
Savarese AM, Lasek AW. Transcriptional Regulators as Targets for Alcohol Pharmacotherapies. Handb Exp Pharmacol 2018; 248:505-533. [PMID: 29594350 PMCID: PMC6242703 DOI: 10.1007/164_2018_101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing brain disease that currently afflicts over 15 million adults in the United States. Despite its prevalence, there are only three FDA-approved medications for AUD treatment, all of which show limited efficacy. Because of their ability to alter expression of a large number of genes, often with great cell-type and brain-region specificity, transcription factors and epigenetic modifiers serve as promising new targets for the development of AUD treatments aimed at the neural circuitry that underlies chronic alcohol abuse. In this chapter, we will discuss transcriptional regulators that can be targeted pharmacologically and have shown some efficacy in attenuating alcohol consumption when targeted. Specifically, the transcription factors cyclic AMP-responsive element binding protein (CREB), peroxisome proliferator-activated receptors (PPARs), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and glucocorticoid receptor (GR), as well as the epigenetic enzymes, the DNA methyltransferases (DNMTs) and histone deacetylases (HDACs), will be discussed.
Collapse
Affiliation(s)
| | - Amy W. Lasek
- Department of Psychiatry, University of Illinois at Chicago,Corresponding author: 1601 West Taylor Street, MC 912, Chicago, IL 60612, Tel: (312) 355-1593,
| |
Collapse
|
31
|
Tilborghs S, Corthouts J, Verhoeven Y, Arias D, Rolfo C, Trinh XB, van Dam PA. The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit Rev Oncol Hematol 2017; 120:141-150. [PMID: 29198328 DOI: 10.1016/j.critrevonc.2017.11.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/01/2017] [Indexed: 12/27/2022] Open
Abstract
Background The Nuclear Factor kappaB (NF-kB) family consists of transcription factors that play a complex and essential role in the regulation of immune responses and inflammation. NF-kB has recently generated considerable interest as it has been implicated in human cancer initiation, progression and resistance to treatment. In the present comprehensive review the different aspects of NF-kB signaling in the carcinogenesis of cancer of the uterine cervix are discussed. NF-kB functions as part of a network, which determines the pattern of its effects on the expression of several other genes (such as crosstalks with reactive oxygen species, p53, STAT3 and miRNAS) and thus its function. Activation of NF-kB triggered by a HPV infection is playing an important role in the innate and adaptive immune response of the host. The virus induces down regulation of NF-kB to liquidate the inhibitory activity for its replication triggered by the immune system leading a status of persistant HPV infection. During the progression to high grade intraepithelial neoplasia and cervical cancer NF-KB becomes constitutionally activated again. Mutations in NF-kB genes are rare in solid tumors but mutations of upstream signaling molecules such as RAS, EGFR, PGF, HER2 have been implicated in elevated NF-kB signaling. NF-kB can stimulate transcription of proliferation regulating genes (eg. cyclin D1 and c-myc), genes involved in metastasis, VEGF dependent angiogenesis and cell immortality by telomerase. NF-kB activation can also induce the expression of activation-induced cytodine deaminase (AID) and the APOBEC proteins, providing a mechanistic link between the NF-kB pathway and mutagenic characteristic of cervical cancer. Inhibition of NF-kB has the potential to be used to reverse resistance to radiotherapy and systemic anti-cancer medication, but currently no clinicaly active NF-kB targeting strategies are available.
Collapse
Affiliation(s)
- Sam Tilborghs
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium
| | - Jerome Corthouts
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium
| | - Yannick Verhoeven
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium
| | - David Arias
- Phase I - Early Clinical Trials Unit & Center for Oncological Research (CORE), Antwerp University, Belgium
| | - Christian Rolfo
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium; Phase I - Early Clinical Trials Unit & Center for Oncological Research (CORE), Antwerp University, Belgium
| | - Xuan Bich Trinh
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium; Gynecologic Oncology Unit, Antwerp University Hospital & Centre of Oncologic Research (CORE), Antwerp University, Belgium
| | - Peter A van Dam
- Multidisciplinary Oncologic Centre Antwerp (MOCA) Antwerp University Hospital, Edegem, Belgium; Gynecologic Oncology Unit, Antwerp University Hospital & Centre of Oncologic Research (CORE), Antwerp University, Belgium.
| |
Collapse
|
32
|
Gudkov AV, Komarova EA. p53 and the Carcinogenicity of Chronic Inflammation. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026161. [PMID: 27549311 DOI: 10.1101/cshperspect.a026161] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is a major cancer predisposition factor. Constitutive activation of the inflammation-driving NF-κB pathway commonly observed in cancer or developed in normal tissues because of persistent infections or endogenous tissue irritating factors, including products of secretion by senescent cells accumulating with age, markedly represses p53 functions. In its turn, p53 acts as a suppressor of inflammation helping to keep it within safe limits. The antagonistic relationship between p53 and NF-κB is controlled by multiple mechanisms and reflects cardinal differences in organismal responses to intrinsic and extrinsic cell stresses driven by these two transcription factors, respectively. This provides an opportunity for developing drugs to treat diseases associated with inappropriate activity of either p53 or NF-κB through targeting the opposing pathway. Several drug candidates of this kind are currently in clinical testing. These include anticancer small molecules capable of simultaneous suppression of p53 and activation of NF-κB and NF-κB-activating biologics that counteract p53-mediated pathologies associated with systemic genotoxic stresses such as acute radiation syndrome and side effects of cancer treatment.
Collapse
Affiliation(s)
- Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Elena A Komarova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263
| |
Collapse
|
33
|
Saeed MT, Ahmad J, Kanwal S, Holowatyj AN, Sheikh IA, Zafar Paracha R, Shafi A, Siddiqa A, Bibi Z, Khan M, Ali A. Formal modeling and analysis of the hexosamine biosynthetic pathway: role of O-linked N-acetylglucosamine transferase in oncogenesis and cancer progression. PeerJ 2016; 4:e2348. [PMID: 27703839 PMCID: PMC5047222 DOI: 10.7717/peerj.2348] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/19/2016] [Indexed: 12/21/2022] Open
Abstract
The alteration of glucose metabolism, through increased uptake of glucose and glutamine addiction, is essential to cancer cell growth and invasion. Increased flux of glucose through the Hexosamine Biosynthetic Pathway (HBP) drives increased cellular O-GlcNAcylation (hyper-O-GlcNAcylation) and contributes to cancer progression by regulating key oncogenes. However, the association between hyper-O-GlcNAcylation and activation of these oncogenes remains poorly characterized. Here, we implement a qualitative modeling framework to analyze the role of the Biological Regulatory Network in HBP activation and its potential effects on key oncogenes. Experimental observations are encoded in a temporal language format and model checking is applied to infer the model parameters and qualitative model construction. Using this model, we discover step-wise genetic alterations that promote cancer development and invasion due to an increase in glycolytic flux, and reveal critical trajectories involved in cancer progression. We compute delay constraints to reveal important associations between the production and degradation rates of proteins. O-linked N-acetylglucosamine transferase (OGT), an enzyme used for addition of O-GlcNAc during O-GlcNAcylation, is identified as a key regulator to promote oncogenesis in a feedback mechanism through the stabilization of c-Myc. Silencing of the OGT and c-Myc loop decreases glycolytic flux and leads to programmed cell death. Results of network analyses also identify a significant cycle that highlights the role of p53-Mdm2 circuit oscillations in cancer recovery and homeostasis. Together, our findings suggest that the OGT and c-Myc feedback loop is critical in tumor progression, and targeting these mediators may provide a mechanism-based therapeutic approach to regulate hyper-O-GlcNAcylation in human cancer.
Collapse
Affiliation(s)
- Muhammad Tariq Saeed
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Jamil Ahmad
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, Pakistan; School of Computer Science and IT, Stratford University, VA, United States
| | - Shahzina Kanwal
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou , China
| | - Andreana N Holowatyj
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute , Detroit , MI , United States
| | - Iftikhar A Sheikh
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Rehan Zafar Paracha
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Aamir Shafi
- School of Electrical Engineering and Computer Science (SEECS), National University of Sciences and Technology (NUST), Islamabad, Pakistan; College of Computer Science and Information Technology, University of Dammam, Al Khobar, Kingdom of Saudi Arabia
| | - Amnah Siddiqa
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Zurah Bibi
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Mukaram Khan
- Research Centre for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Amjad Ali
- Atta-ur-Rehman School of Applied Bio-science (ASAB), National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| |
Collapse
|
34
|
Hussein RM, Elsirafy OM, Wahba YS, Kawy HSA, Hasanin AH, Hamam GG. Theophylline, an old drug with multi-faceted effects: Its potential benefits in immunological liver injury in rats. Life Sci 2015; 136:100-7. [DOI: 10.1016/j.lfs.2015.06.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/26/2015] [Accepted: 06/29/2015] [Indexed: 01/06/2023]
|
35
|
Niu X, Zhang H, Li W, Wang Y, Mu Q, Wang X, He Z, Yao H. Protective effect of cavidine on acetic acid-induced murine colitis via regulating antioxidant, cytokine profile and NF-κB signal transduction pathways. Chem Biol Interact 2015; 239:34-45. [PMID: 26102009 DOI: 10.1016/j.cbi.2015.06.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 12/30/2022]
Abstract
Ulcerative colitis is an inflammatory disorder characterized by neutrophils infiltration, oxidative stress, upregulation of pro-inflammatory mediators and cytokines. Cavidine possesses anti-inflammatory activity and has been used to treat various inflammatory diseases but its effect on ulcerative colitis has not been previously explored. The present study aims to evaluate the effect of cavidine on acetic acid-induced ulcerative colitis in mice. Colitis mice induced by intra-rectal acetic acid (5%, v/v) administration received cavidine (1, 5 and 10mg/kg, i.g) or sulfasalazine (500mg/kg, i.g) for seven consecutive days. After euthanized by cervical dislocation, colonic segments of mice were excised for clinical, macroscopic, biochemical and histopathological examinations. Results suggested treatment with cavidine significantly decreased mortality rate, body weight loss, disease activity index (DAI), wet colon weight, macroscopic and histological score when compared with that of acetic acid-induced controls. In addition, administration of cavidine effectively modulated expressions of MPO, GSH, SOD and MDA. Furthermore cavidine inhibited the level of TNF-α and IL-6 in the serum and colon tissue in response to the regulation of p65 NF-κB protein expression. All these results indicated cavidine exerts marked protective effect in experimental colitis, possibly by regulating the expression of oxygen metabolites, NF-κB and subsequent pro-inflammatory cytokines production.
Collapse
Affiliation(s)
- Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Hailin Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Yu Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Qingli Mu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiumei Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Zehong He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Huan Yao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| |
Collapse
|
36
|
McGrath KCY, Hill MD, McRobb LS, Heather AK. The androgen receptor drives the sex-specific expression of vascular cell adhesion molecule-1 in endothelial cells but not lipid metabolism genes in monocyte-derived macrophages. Horm Mol Biol Clin Investig 2015; 2:203-9. [PMID: 25961193 DOI: 10.1515/hmbci.2010.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2009] [Accepted: 01/12/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND Anecdotal evidence suggests that male sex hormones are proatherogenic. We hypothesized that the male sex hormone receptor, the androgen receptor (AR), acts as a molecular switch in sex-specific inflammatory signaling in vascular cells. MATERIALS AND METHODS AR expression in human umbilical vein endothelial cells (HUVECs), human monocyte-derived macrophages (MDMs) or HeLa cells was modulated by transfection with AR siRNA or human AR cDNA expression vector. Activity and expression levels were measured by luciferase reporter assays, Western blotting or real-time PCR analysis. RESULTS AR knockdown reduced expression of vascular cell adhesion molecule-1 (VCAM-1) in genetically male HUVECs. Conversely, AR upregulation in genetically female HUVECs induced VCAM-1 expression and increased dihydrotestosterone-stimulated monocyte adhesion. Co-transfection of an AR expression vector with VCAM-1 or NF-κB-reporter vectors into phenotypically female, AR-negative HeLa cells confirmed AR regulation of VCAM-1 expression as well as AR activation of NF-κB. AR upregulation was not sufficient to increase ICAM-1 levels in female HUVECs or lipoprotein metabolism gene expression in female MDMs, despite AR knockdown limiting expression in their male counterparts. CONCLUSIONS AR acts as a molecular switch to induce VCAM-1 expression. Low AR levels in female HUVECs limit NF-κB/VCAM-1 induction and monocyte adhesion and could contribute to the gender bias in cardiovascular disease. Unidentified factors in female cells limit induction of other proatherogenic genes not primarily regulated by NF-κB.
Collapse
|
37
|
Li F, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. NF-κB in cancer therapy. Arch Toxicol 2015; 89:711-31. [PMID: 25690730 DOI: 10.1007/s00204-015-1470-4] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor kappa B (NF-κB) has attracted increasing attention in the field of cancer research from last few decades. Aberrant activation of this transcription factor is frequently encountered in a variety of solid tumors and hematological malignancies. NF-κB family members and their regulated genes have been linked to malignant transformation, tumor cell proliferation, survival, angiogenesis, invasion/metastasis, and therapeutic resistance. In this review, we highlight the diverse molecular mechanism(s) by which the NF-κB pathway is constitutively activated in different types of human cancers, and the potential role of various oncogenic genes regulated by this transcription factor in cancer development and progression. Additionally, various pharmacological approaches employed to target the deregulated NF-κB signaling pathway, and their possible therapeutic potential in cancer therapy is also discussed briefly.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, Cancer Science Institute, National University of Singapore, Singapore, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Pratheeshkumar P, Son YO, Divya SP, Roy RV, Hitron JA, Wang L, Kim D, Dai J, Asha P, Zhang Z, Wang Y, Shi X. Luteolin inhibits Cr(VI)-induced malignant cell transformation of human lung epithelial cells by targeting ROS mediated multiple cell signaling pathways. Toxicol Appl Pharmacol 2014; 281:230-41. [PMID: 25448439 DOI: 10.1016/j.taap.2014.10.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/22/2014] [Accepted: 10/14/2014] [Indexed: 12/27/2022]
Abstract
Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with the incidence of lung cancer. Inhibition of metal induced carcinogenesis by a dietary antioxidant is a novel approach. Luteolin, a natural dietary flavonoid found in fruits and vegetables, possesses potent antioxidant and anti-inflammatory activity. We found that short term exposure of human bronchial epithelial cells (BEAS-2B) to Cr(VI) (5μM) showed a drastic increase in ROS generation, NADPH oxidase (NOX) activation, lipid peroxidation, and glutathione depletion, which were significantly inhibited by the treatment with luteolin in a dose dependent manner. Treatment with luteolin decreased AP-1, HIF-1α, COX-2, and iNOS promoter activity induced by Cr(VI) in BEAS-2B cells. In addition, luteolin protected BEAS-2B cells from malignant transformation induced by chronic Cr(VI) exposure. Moreover, luteolin also inhibited the production of pro-inflammatory cytokines (IL-1β, IL-6, IL-8, TNF-α) and VEGF in chronic Cr(VI) exposed BEAS-2B cells. Western blot analysis showed that luteolin inhibited multiple gene products linked to survival (Akt, Fak, Bcl-2, Bcl-xL), inflammation (MAPK, NF-κB, COX-2, STAT-3, iNOS, TNF-α) and angiogenesis (HIF-1α, VEGF, MMP-9) in chronic Cr(VI) exposed BEAS-2B cells. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of luteolin showed reduced tumor incidence compared to Cr(VI) alone treated group. Overexpression of catalase (CAT) or SOD2, eliminated Cr(VI)-induced malignant transformation. Overall, our results indicate that luteolin protects BEAS-2B cells from Cr(VI)-induced carcinogenesis by scavenging ROS and modulating multiple cell signaling mechanisms that are linked to ROS. Luteolin, therefore, serves as a potential chemopreventive agent against Cr(VI)-induced carcinogenesis.
Collapse
Affiliation(s)
- Poyil Pratheeshkumar
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Sasidharan Padmaja Divya
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Ram Vinod Roy
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Donghern Kim
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Jin Dai
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Padmaja Asha
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Cochin, India
| | - Zhuo Zhang
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xianglin Shi
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| |
Collapse
|
39
|
Pratheeshkumar P, Son YO, Wang X, Divya SP, Joseph B, Hitron JA, Wang L, Kim D, Yin Y, Roy RV, Lu J, Zhang Z, Wang Y, Shi X. Cyanidin-3-glucoside inhibits UVB-induced oxidative damage and inflammation by regulating MAP kinase and NF-κB signaling pathways in SKH-1 hairless mice skin. Toxicol Appl Pharmacol 2014; 280:127-37. [PMID: 25062774 PMCID: PMC4330564 DOI: 10.1016/j.taap.2014.06.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/24/2014] [Accepted: 06/29/2014] [Indexed: 12/17/2022]
Abstract
Skin cancer is one of the most commonly diagnosed cancers in the United States. Exposure to ultraviolet-B (UVB) radiation induces inflammation and photocarcinogenesis in mammalian skin. Cyanidin-3-glucoside (C3G), a member of the anthocyanin family, is present in various vegetables and fruits especially in edible berries, and displays potent antioxidant and anticarcinogenic properties. In this study, we have assessed the in vivo effects of C3G on UVB irradiation induced chronic inflammatory responses in SKH-1 hairless mice, a well-established model for UVB-induced skin carcinogenesis. Here, we show that C3G inhibited UVB-induced skin damage and inflammation in SKH-1 hairless mice. Our results indicate that C3G inhibited glutathione depletion, lipid peroxidation and myeloperoxidation in mouse skin by chronic UVB exposure. C3G significantly decreased the production of UVB-induced pro-inflammatory cytokines, such as IL-6 and TNF-α, associated with cutaneous inflammation. Likewise, UVB-induced inflammatory responses were diminished by C3G as observed by a remarkable reduction in the levels of phosphorylated MAP kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, C3G also decreased UVB-induced cyclooxygenase-2 (COX-2), PGE2 and iNOS levels, which are well-known key mediators of inflammation and cancer. Treatment with C3G inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mice skin. Immunofluorescence assay revealed that topical application of C3G inhibited the expression of 8-hydroxy-2'-deoxyguanosine, proliferating cell nuclear antigen, and cyclin D1 in chronic UVB exposed mouse skin. Collectively, these data indicates that C3G can provide substantial protection against the adverse effects of UVB radiation by modulating UVB-induced MAP kinase and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Poyil Pratheeshkumar
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Xin Wang
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Sasidharan Padmaja Divya
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Binoy Joseph
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Donghern Kim
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yuanqin Yin
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Cancer Institute, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Ram Vinod Roy
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Jian Lu
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhuo Zhang
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xianglin Shi
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| |
Collapse
|
40
|
Biphasic activation of nuclear factor kappa B and expression of p65 and c-Rel after traumatic brain injury in rats. Inflamm Res 2013; 63:109-15. [DOI: 10.1007/s00011-013-0677-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 10/01/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022] Open
|
41
|
Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 2013; 12:86. [PMID: 23915189 PMCID: PMC3750319 DOI: 10.1186/1476-4598-12-86] [Citation(s) in RCA: 2427] [Impact Index Per Article: 202.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/30/2013] [Indexed: 02/07/2023] Open
Abstract
The NF-κB family of transcription factors has an essential role in inflammation and innate immunity. Furthermore, NF-κB is increasingly recognized as a crucial player in many steps of cancer initiation and progression. During these latter processes NF-κB cooperates with multiple other signaling molecules and pathways. Prominent nodes of crosstalk are mediated by other transcription factors such as STAT3 and p53 or the ETS related gene ERG. These transcription factors either directly interact with NF-κB subunits or affect NF-κB target genes. Crosstalk can also occur through different kinases, such as GSK3-β, p38, or PI3K, which modulate NF-κB transcriptional activity or affect upstream signaling pathways. Other classes of molecules that act as nodes of crosstalk are reactive oxygen species and miRNAs. In this review, we provide an overview of the most relevant modes of crosstalk and cooperativity between NF-κB and other signaling molecules during inflammation and cancer.
Collapse
Affiliation(s)
- Bastian Hoesel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Johannes A Schmid
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| |
Collapse
|
42
|
Bao B, Azmi AS, Ali S, Ahmad A, Li Y, Banerjee S, Kong D, Sarkar FH. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1826:272-96. [PMID: 22579961 PMCID: PMC3788359 DOI: 10.1016/j.bbcan.2012.04.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/25/2012] [Accepted: 04/28/2012] [Indexed: 12/13/2022]
Abstract
Hypoxia is one of the fundamental biological phenomena that are intricately associated with the development and aggressiveness of a variety of solid tumors. Hypoxia-inducible factors (HIF) function as a master transcription factor, which regulates hypoxia responsive genes and has been recognized to play critical roles in tumor invasion, metastasis, and chemo-radiation resistance, and contributes to increased cell proliferation, survival, angiogenesis and metastasis. Therefore, tumor hypoxia with deregulated expression of HIF and its biological consequence lead to poor prognosis of patients diagnosed with solid tumors, resulting in higher mortality, suggesting that understanding of the molecular relationship of hypoxia with other cellular features of tumor aggressiveness would be invaluable for developing newer targeted therapy for solid tumors. It has been well recognized that cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) phenotypic cells are associated with therapeutic resistance and contribute to aggressive tumor growth, invasion, metastasis and believed to be the cause of tumor recurrence. Interestingly, hypoxia and HIF signaling pathway are known to play an important role in the regulation and sustenance of CSCs and EMT phenotype. However, the molecular relationship between HIF signaling pathway with the biology of CSCs and EMT remains unclear although NF-κB, PI3K/Akt/mTOR, Notch, Wnt/β-catenin, and Hedgehog signaling pathways have been recognized as important regulators of CSCs and EMT. In this article, we will discuss the state of our knowledge on the role of HIF-hypoxia signaling pathway and its kinship with CSCs and EMT within the tumor microenvironment. We will also discuss the potential role of hypoxia-induced microRNAs (miRNAs) in tumor development and aggressiveness, and finally discuss the potential effects of nutraceuticals on the biology of CSCs and EMT in the context of tumor hypoxia.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Asfar S. Azmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Shadan Ali
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Yiwei Li
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
43
|
Lentsch AB. Regulatory mechanisms of injury and repair after hepatic ischemia/reperfusion. SCIENTIFICA 2012; 2012:513192. [PMID: 24278708 PMCID: PMC3820555 DOI: 10.6064/2012/513192] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/12/2012] [Indexed: 06/02/2023]
Abstract
Hepatic ischemia/reperfusion injury is an important complication of liver surgery and transplantation. The mechanisms of this injury as well as the subsequent reparative and regenerative processes have been the subject of thorough study. In this paper, we discuss the complex and coordinated responses leading to parenchymal damage after liver ischemia/reperfusion as well as the manner in which the liver clears damaged cells and regenerates functional mass.
Collapse
Affiliation(s)
- Alex B. Lentsch
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267-0558, USA
| |
Collapse
|
44
|
Qiu M, Chen Y, Song S, Song H, Chu Y, Yuan Z, Cheng L, Zheng D, Chen Z, Wu Z. Poly (4-styrenesulfonic acid-co-maleic acid) is an entry inhibitor against both HIV-1 and HSV infections - potential as a dual functional microbicide. Antiviral Res 2012; 96:138-47. [PMID: 22940611 DOI: 10.1016/j.antiviral.2012.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 08/09/2012] [Accepted: 08/14/2012] [Indexed: 11/29/2022]
Abstract
Genital herpes is one of the most prevalent sexually transmitted diseases (STD) caused by herpes simplex viruses type 1 and 2 (HSV-1 and -2). HSV is considered as a major risk factor in human immunodeficiency virus type-1 (HIV-1) infection and rapid progression to acquired immunodeficiency syndrome (AIDS). Here, we reported the finding of a polymer of styrenesulfonic acid and maleic acid (PSM) which exhibited antiviral activity with low cytotoxicity. PSM exhibited in vitro inhibitory activity against HIV-1 pseudovirus and HSV-1 and -2. In vivo efficacy of PSM against HSV-2 (G) was also investigated. We found that both 1% and 5% PSM gels protected mice from HSV-2 vaginal infection and disease progression significantly. Mechanistic analysis demonstrated that PSM was likely an entry inhibitor that disrupted viral attachment to the target cells. In particular, PSM disrupted gp120 binding to CD4 by interacting with the gp120 V3-loop and the CD4-binding site. The in vitro cytotoxicity studies showed that PSM did not stimulate NF-κB activation and up-regulation of proinflammatory cytokine IL-1β and IL-8 in vaginal epithelial cells. In addition, PSM also showed low adverse effect on the growth of vaginal Lactobacillus strains. PSM is, therefore, a novel viral entry inhibitor and a potential microbicide candidate against both HIV-1 and HSV.
Collapse
Affiliation(s)
- Min Qiu
- Center for Public Health Research, School of Medicine, Nanjing University, Nanjing, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bao B, Ahmad A, Li Y, Azmi AS, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSCs within the tumor microenvironment for cancer therapy: a potential role of mesenchymal stem cells. Expert Opin Ther Targets 2012; 16:1041-54. [DOI: 10.1517/14728222.2012.714774] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
46
|
Chen YF, Pottala JV, Weltman NY, Ge X, Savinova OV, Gerdes AM. Regulation of gene expression with thyroid hormone in rats with myocardial infarction. PLoS One 2012; 7:e40161. [PMID: 22870193 PMCID: PMC3411604 DOI: 10.1371/journal.pone.0040161] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 06/01/2012] [Indexed: 11/19/2022] Open
Abstract
Introduction The expression of hundreds of genes is altered in response to left ventricular (LV) remodeling following large transmural myocardial infarction (MI). Thyroid hormone (TH) improves LV remodeling and cardiac performance after MI. However, the molecular basis is unknown. Methods MI was produced by ligation of the left anterior descending coronary artery in female SD rats. Rats were divided into the following groups: (1) Sham MI, (2) MI, and (3) MI+T4 treatment (T4 pellet 3.3 mg, 60 days release, implanted subcutaneously immediately following MI). Four weeks after surgery, total RNA was isolated from LV non-infarcted areas for microarray analysis using the Illumina RatRef-12 Expression BeadChip Platform. Results Signals were detected in 13,188 genes (out of 22,523), of which the expression of 154 genes were decreased and the expression of 200 genes were increased in MI rats compared with Sham MI rats (false discovery rate (FDR) <0.05). Compared to MI rats, T4 treatment decreased expression of 27 genes and increased expression of 28 genes. In particular, 6 genes down-regulated by MI and 12 genes up-regulated by MI were reversed by T4. Most of the 55 genes altered by T4 treatment are in the category of molecular function under binding (24) and biological processes which includes immune system process (9), multi-organism process (5) and biological regulation (19) nonexclusively. Conclusions These results suggest that altered expression of genes for molecular function and biological process may be involved in the beneficial effects of thyroid hormone treatment following MI in rats.
Collapse
Affiliation(s)
- Yue-Feng Chen
- Department of Biomedical Sciences, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury, New York, United States of America.
| | | | | | | | | | | |
Collapse
|
47
|
Contrasting effect of recombinant human erythropoietin on breast cancer cell response to cisplatin induced cytotoxicity. Radiol Oncol 2012; 46:213-25. [PMID: 23077460 PMCID: PMC3472952 DOI: 10.2478/v10019-012-0037-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 05/18/2012] [Indexed: 12/11/2022] Open
Abstract
Background Human recombinant erythropoietin (rHuEpo) that is used for the treatment of the chemotherapy-induced anaemia in cancer patients was shown to cause detrimental effects on the course of disease due to increased adverse events inflicting patient’s survival, potentially related to rHuEpo-induced cancer progression. In this study, we elucidate the effect of rHuEpo administration on breast cancer cell proliferation and gene expression after cisplatin (cDDP) induced cytotoxicity. Materials and methods Two breast carcinoma models, MCF-7 and MDA-MB-231 cell lines, were used differing in oestrogen (ER) and progesterone (PR) receptors and p53 status. Cells were cultured with or without rHuEpo for 24 h or 9 weeks and their growth characteristics after cDDP treatment were assessed together with expression of genes involved in the p53-signaling pathway. Results Short-term exposure of breast cancer cells to rHuEpo lowers their proliferation and reduces cDDP cytotoxic potency. In contrast, long-term exposure of MCF-7 cells to rHuEpo increases proliferation and predisposes MCF-7 cells to cDDP cytotoxicity, but has no effect on MDA-MB-231 cells. MDA-MB-231 cells show altered level of ERK phosphorylation, indicating involvement of MAPK signalling pathway. Gene expression analysis of p53-dependent genes and bcl-2 gene family members confirmed differences between long and short-term rHuEpo effects, indicating the most prominent changes in BCL2 and BAD expression. Conclusions Proliferation and survival characteristics of MCF-7 cells are reversely modulated by the length of the rHuEpo exposure. On the other hand, MDA-MB-231 cells are almost irresponsive to long-term rHuEpo, supposedly due to the mutated p53 and ER(+)/PR(−) status. The p53 and ER/PR status may predict tumour response on rHuEpo and cDDP treatment.
Collapse
|
48
|
Gancz D, Lusthaus M, Fishelson Z. A role for the NF-κB pathway in cell protection from complement-dependent cytotoxicity. THE JOURNAL OF IMMUNOLOGY 2012; 189:860-6. [PMID: 22685314 DOI: 10.4049/jimmunol.1103451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nucleated cells are equipped with several mechanisms that support their resistance to complement-dependent cytotoxicity (CDC). The role of the NF-κB pathway in cell protection from CDC was examined. Elevated sensitivity to CDC was demonstrated in cells lacking the p65 subunit of NF-κB or the IκB kinases IKKα or IKKβ, and in cells treated with p65 small interfering RNA. Pretreatment with the IKK inhibitor PS-1145 also enhanced CDC of wild-type cells (WT) but not of p65(-/-) cells. Furthermore, reconstitution of p65 into p65(-/-) cells and overexpression of p65 in WT cells lowered their sensitivity to CDC. The postulated effect of p65 on the JNK-mediated death-signaling pathway activated by complement was examined. p65 small interfering RNA enhanced CDC in WT cells but not in cells lacking JNK. JNK phosphorylation induced by complement was more pronounced in p65(-/-) cells than in WT cells. The results indicate that the NF-κB pathway mediates cell resistance to CDC, possibly by suppressing JNK-dependent programmed necrotic cell death.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | |
Collapse
|
49
|
Bao B, Thakur A, Li Y, Ahmad A, Azmi AS, Banerjee S, Kong D, Ali S, Lum LG, Sarkar FH. The immunological contribution of NF-κB within the tumor microenvironment: a potential protective role of zinc as an anti-tumor agent. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1825:160-72. [PMID: 22155217 PMCID: PMC3811120 DOI: 10.1016/j.bbcan.2011.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/14/2011] [Accepted: 11/19/2011] [Indexed: 12/16/2022]
Abstract
Over decades, cancer treatment has been mainly focused on targeting cancer cells and not much attention to host tumor microenvironment. Recent advances suggest that the tumor microenvironment requires in-depth investigation for understanding the interactions between tumor cell biology and immunobiology in order to optimize therapeutic approaches. Tumor microenvironment consists of cancer cells and tumor associated reactive fibroblasts, infiltrating non-cancer cells, secreted soluble factors or molecules, and non-cellular support materials. Tumor associated host immune cells such as Th(1), Th(2), Th17, regulatory cells, dendritic cells, macrophages, and myeloid-derived suppressor cells are major components of the tumor microenvironment. Accumulating evidence suggests that these tumor associated immune cells may play important roles in cancer development and progression. However, the exact functions of these cells in the tumor microenvironment are poorly understood. In the tumor microenvironment, NF-κB plays an important role in cancer development and progression because this is a major transcription factor which regulates immune functions within the tumor microenvironment. In this review, we will focus our discussion on the immunological contribution of NF-κB in tumor associated host immune cells within the tumor microenvironment. We will also discuss the potential protective role of zinc, a well-known immune response mediator, in the regulation of these immune cells and cancer cells in the tumor microenvironment especially because zinc could be useful for conditioning the tumor microenvironment toward innovative cancer therapy.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Archana Thakur
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Yiwei Li
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Asfar S. Azmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Shadan Ali
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Lawrence G. Lum
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Immunology and Microbiology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
50
|
Lactobacillus acidophilus induces cytokine and chemokine production via NF-κB and p38 mitogen-activated protein kinase signaling pathways in intestinal epithelial cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:603-8. [PMID: 22357649 DOI: 10.1128/cvi.05617-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Intestinal epithelial cells can respond to certain bacteria by producing an array of cytokines and chemokines which are associated with host immune responses. Lactobacillus acidophilus NCFM is a characterized probiotic, originally isolated from human feces. This study aimed to test the ability of L. acidophilus NCFM to stimulate cytokine and chemokine production in intestinal epithelial cells and to elucidate the mechanisms involved in their upregulation. In experiments using intestinal epithelial cell lines and mouse models, we observed that L. acidophilus NCFM could rapidly but transiently upregulate a number of effector genes encoding cytokines and chemokines such as interleukin 1α (IL-1α), IL-1β, CCL2, and CCL20 and that cytokines showed lower expression levels with L. acidophilus NCFM treatment than chemokines. Moreover, L. acidophilus NCFM could activate a pathogen-associated molecular pattern receptor, Toll-like receptor 2 (TLR2), in intestinal epithelial cell lines. The phosphorylation of NF-κB p65 and p38 mitogen-activated protein kinase (MAPK) in intestinal epithelial cell lines was also enhanced by L. acidophilus NCFM. Furthermore, inhibitors of NF-κB (pyrrolidine dithiocarbamate [PDTC]) and p38 MAPK (SB203580) significantly reduced cytokine and chemokine production in the intestinal epithelial cell lines stimulated by L. acidophilus NCFM, suggesting that both NF-κB and p38 MAPK signaling pathways were important for the production of cytokines and chemokines induced by L. acidophilus NCFM.
Collapse
|