1
|
Ansari U, Nadora D, Alam M, Wen J, Asad S, Lui F. Influence of dietary patterns in the pathophysiology of Huntington's Disease: A literature review. AIMS Neurosci 2024; 11:63-75. [PMID: 38988882 PMCID: PMC11230857 DOI: 10.3934/neuroscience.2024005] [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: 01/01/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 07/12/2024] Open
Abstract
Huntington's disease (HD), a rare autosomal dominant neurodegenerative disease, causes the gradual deterioration of neurons in the basal ganglia, specifically in the striatum. HD displays a wide range of symptoms, from motor disturbances such as chorea, dystonia, and bradykinesia to more debilitating symptoms such as cognitive decline, behavioral abnormalities, and psychiatric disturbances. Current research suggests the potential use of dietary interventions as viable strategies for slowing the progression of HD. Most notably, the Mediterranean, vegan, carnivore, paleo, and ketogenic diets have gained attention due to their hypothesized impact on neuroprotection and symptomatic modulation in various neurodegenerative disorders. Despite substantial nutritional differences among these diets, they share a fundamental premise-that dietary factors have an influential impact in modifying pertinent biological pathways linked to neurodegeneration. Understanding the intricate interactions between these dietary regimens and HD pathogenesis could open avenues for personalized interventions tailored to the individual's specific needs and genetic background. Ultimately, elucidating the multifaceted effects of these diets on HD offers a promising framework for developing comprehensive therapeutic approaches that integrate dietary strategies with conventional treatments.
Collapse
Affiliation(s)
- Ubaid Ansari
- California Northstate University College of Medicine, USA
| | - Dawnica Nadora
- California Northstate University College of Medicine, USA
| | - Meraj Alam
- California Northstate University College of Medicine, USA
| | - Jimmy Wen
- California Northstate University College of Medicine, USA
| | - Shaheryar Asad
- California Northstate University College of Medicine, USA
| | - Forshing Lui
- California Northstate University College of Medicine, USA
| |
Collapse
|
2
|
Li Y, Xu H, Zhou L, Zhang Y, Yu W, Li S, Gao J. Bifidobacterium breve Protects the Intestinal Epithelium and Mitigates Inflammation in Colitis via Regulating the Gut Microbiota-Cholic Acid Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3572-3583. [PMID: 38334304 DOI: 10.1021/acs.jafc.3c08527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
In this study, we aimed to explore the protective effects of Bifidobacterium in colitis mice and the potential mechanisms. Results showed that Bifidobacterium breve (B. breve) effectively colonized the intestinal tract and alleviated colitis symptoms by reducing the disease activity index. Moreover, B. breve mitigated intestinal epithelial cell damage, inhibited the pro-inflammatory factors, and upregulated tight junction (TJ)-proteins. Gut microbiota and metabolome analysis found that B. breve boosted bile acid-regulating genera (such as Bifidobacterium and Clostridium sensu stricto 1), which promoted bile acid deconjugation in the intestine. Notably, cholic acid (CA) was closely associated with the expression levels of inflammatory factors and TJ-proteins (p < 0.05). Our in vitro cell experiments further confirmed that CA (20.24 ± 4.53 pg/mL) contributed to the inhibition of lipopolysaccharide-induced tumor necrosis factor-α expression (49.32 ± 5.27 pg/mL) and enhanced the expression of TJ-proteins (Occludin and Claudin-1) and MUC2. This study suggested that B. breve could be a probiotic candidate for use in infant foods.
Collapse
Affiliation(s)
- Yaqian Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Hongtao Xu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Liuyang Zhou
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yuwei Zhang
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Wenqing Yu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Shubo Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jie Gao
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| |
Collapse
|
3
|
Chong ZX, Yong CY, Ong AHK, Yeap SK, Ho WY. Deciphering the roles of aryl hydrocarbon receptor (AHR) in regulating carcinogenesis. Toxicology 2023; 495:153596. [PMID: 37480978 DOI: 10.1016/j.tox.2023.153596] [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: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent receptor that belongs to the superfamily of basic helix-loop-helix (bHLH) transcription factors. The activation of the canonical AHR signaling pathway is known to induce the expression of cytochrome P450 enzymes, facilitating the detoxification metabolism in the human body. Additionally, AHR could interact with various signaling pathways such as epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), hypoxia-inducible factor-1α (HIF-1α), nuclear factor ekappa B (NF-κβ), estrogen receptor (ER), and androgen receptor (AR) signaling pathways. Over the past 30 years, several studies have reported that various chemical, physical, or biological agents, such as tobacco, hydrocarbon compounds, industrial and agricultural chemical wastes, drugs, UV, viruses, and other toxins, could affect AHR expression or activity, promoting cancer development. Thus, it is valuable to overview how these factors regulate AHR-mediated carcinogenesis. Current findings have reported that many compounds could act as AHR ligands to drive the expressions of AHR-target genes, such as CYP1A1, CYP1B1, MMPs, and AXL, and other targets that exert a pro-proliferation or anti-apoptotic effect, like XIAP. Furthermore, some other physical and chemical agents, such as UV and 3-methylcholanthrene, could promote AHR signaling activities, increasing the signaling activities of a few oncogenic pathways, such as the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways. Understanding how various factors regulate AHR-mediated carcinogenesis processes helps clinicians and scientists plan personalized therapeutic strategies to improve anti-cancer treatment efficacy. As many studies that have reported the roles of AHR in regulating carcinogenesis are preclinical or observational clinical studies that did not explore the detailed mechanisms of how different chemical, physical, or biological agents promote AHR-mediated carcinogenesis processes, future studies should focus on conducting large-scale and functional studies to unravel the underlying mechanism of how AHR interacts with different factors in regulating carcinogenesis processes.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Chean Yeah Yong
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia
| | - Alan Han Kiat Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
4
|
Villéger R, Chulkina M, Mifflin RC, Markov NS, Trieu J, Sinha M, Johnson P, Saada JI, Adegboyega PA, Luxon BA, Beswick EJ, Powell DW, Pinchuk IV. Loss of alcohol dehydrogenase 1B in cancer-associated fibroblasts: contribution to the increase of tumor-promoting IL-6 in colon cancer. Br J Cancer 2023; 128:537-548. [PMID: 36482184 PMCID: PMC9938173 DOI: 10.1038/s41416-022-02066-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/24/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Increases in IL-6 by cancer-associated fibroblasts (CAFs) contribute to colon cancer progression, but the mechanisms involved in the increase of this tumor-promoting cytokine are unknown. The aim of this study was to identify novel targets involved in the dysregulation of IL-6 expression by CAFs in colon cancer. METHODS Colonic normal (N), hyperplastic, tubular adenoma, adenocarcinoma tissues, and tissue-derived myo-/fibroblasts (MFs) were used in these studies. RESULTS Transcriptomic analysis demonstrated a striking decrease in alcohol dehydrogenase 1B (ADH1B) expression, a gene potentially involved in IL-6 dysregulation in CAFs. ADH1B expression was downregulated in approximately 50% of studied tubular adenomas and all T1-4 colon tumors, but not in hyperplastic polyps. ADH1B metabolizes alcohols, including retinol (RO), and is involved in the generation of all-trans retinoic acid (atRA). LPS-induced IL-6 production was inhibited by either RO or its byproduct atRA in N-MFs, but only atRA was effective in CAFs. Silencing ADH1B in N-MFs significantly upregulated LPS-induced IL-6 similar to those observed in CAFs and lead to the loss of RO inhibitory effect on inducible IL-6 expression. CONCLUSION Our data identify ADH1B as a novel potential mesenchymal tumor suppressor, which plays a critical role in ADH1B/retinoid-mediated regulation of tumor-promoting IL-6.
Collapse
Affiliation(s)
- Romain Villéger
- Laboratoire Ecologie and Biologie des Interactions, UMR CNRS 7267, Université de Poitiers, Poitiers, France
| | - Marina Chulkina
- Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Randy C Mifflin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UTMB, Galveston, TX, 77555, USA
| | - Nikolay S Markov
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Judy Trieu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UTMB, Galveston, TX, 77555, USA
| | - Mala Sinha
- Institute for Translational Sciences, UTMB, Galveston, TX, 77555, USA
| | - Paul Johnson
- Department of Surgery, UTMB, Galveston, TX, 77555, USA
| | - Jamal I Saada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UTMB, Galveston, TX, 77555, USA
| | - Patrick A Adegboyega
- Department of Pathology, St. Louis University School of Medicine, St. Louis, MO, 63106, USA
| | - Bruce A Luxon
- Institute for Translational Sciences, UTMB, Galveston, TX, 77555, USA
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Don W Powell
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UTMB, Galveston, TX, 77555, USA
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84132, USA
- Department of Neuroscience and Cell Biology, UTMB, Galveston, TX, 77555, USA
| | - Irina V Pinchuk
- Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
5
|
Ogundepo S, Chiamaka AM, Olatinwo M, Adepoju D, Aladesanmi MT, Celestine UO, Ali KC, Umezinwa OJ, Olasore J, Alausa A. The role of diosgenin in crohn’s disease. CLINICAL PHYTOSCIENCE 2022. [DOI: 10.1186/s40816-022-00338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractInflammatory bowel disease (IBD) is a chronic idiopathic inflammation that can grossly affect the entire gastrointestinal tract (GIT) from the mouth to the anus. Crohn’s disease is the most known type of IBD and has been the focus of attention due to its increase in prevalence worldwide. Although the etiology is yet to be elucidated, recent studies have pointed out Crohn’s disease to arise from a complex interaction between environmental influences, genetic predisposition, and altered gut microbiota, resulting in dysregulated adaptive and innate responses. The presenting hallmarks of Crohn’s disease may include weight loss, nausea, vomiting, abdominal pain, diarrhea, fever, or chills. Treatment is usually done with many approved immunosuppressive drugs and surgery. However, a promising avenue from natural compounds is a safer therapy due to its safe natural active ingredients and the strong activity it shows in the treatment and management of diseases. Diosgenin, “a major biologically active natural steroidal sapogenin found in Chinese yam,” has been widely reported as a therapeutic agent in the treatment of various classes of disorders such as hyperlipidemia, inflammation, diabetes, cancer, infection, and immunoregulation. In this review, an analysis of literature data on diosgenin employed as a therapeutic agent for the treatment of Crohn’s disease is approached, to strengthen the scientific database and curtail the dreadful impact of Crohn’s disease.
Collapse
|
6
|
Fitriana M, Hwang WL, Chan PY, Hsueh TY, Liao TT. Roles of microRNAs in Regulating Cancer Stemness in Head and Neck Cancers. Cancers (Basel) 2021; 13:cancers13071742. [PMID: 33917482 PMCID: PMC8038798 DOI: 10.3390/cancers13071742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are epithelial malignancies with 5-year overall survival rates of approximately 40-50%. Emerging evidence indicates that a small population of cells in HNSCC patients, named cancer stem cells (CSCs), play vital roles in the processes of tumor initiation, progression, metastasis, immune evasion, chemo-/radioresistance, and recurrence. The acquisition of stem-like properties of cancer cells further provides cellular plasticity for stress adaptation and contributes to therapeutic resistance, resulting in a worse clinical outcome. Thus, targeting cancer stemness is fundamental for cancer treatment. MicroRNAs (miRNAs) are known to regulate stem cell features in the development and tissue regeneration through a miRNA-target interactive network. In HNSCCs, miRNAs act as tumor suppressors and/or oncogenes to modulate cancer stemness and therapeutic efficacy by regulating the CSC-specific tumor microenvironment (TME) and signaling pathways, such as epithelial-to-mesenchymal transition (EMT), Wnt/β-catenin signaling, and epidermal growth factor receptor (EGFR) or insulin-like growth factor 1 receptor (IGF1R) signaling pathways. Owing to a deeper understanding of disease-relevant miRNAs and advances in in vivo delivery systems, the administration of miRNA-based therapeutics is feasible and safe in humans, with encouraging efficacy results in early-phase clinical trials. In this review, we summarize the present findings to better understand the mechanical actions of miRNAs in maintaining CSCs and acquiring the stem-like features of cancer cells during HNSCC pathogenesis.
Collapse
Affiliation(s)
- Melysa Fitriana
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Otorhinolaryngology Head and Neck Surgery Department, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Wei-Lun Hwang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Cancer Progression Center of Excellence, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Pak-Yue Chan
- School of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.C.); (T.-Y.H.)
| | - Tai-Yuan Hsueh
- School of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.C.); (T.-Y.H.)
| | - Tsai-Tsen Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Correspondence: ; Tel.: +886-2736-1661 (ext. 3435)
| |
Collapse
|
7
|
Kumar S, Agnihotri N. Piperlongumine targets NF-κB and its downstream signaling pathways to suppress tumor growth and metastatic potential in experimental colon cancer. Mol Cell Biochem 2021; 476:1765-1781. [PMID: 33433833 DOI: 10.1007/s11010-020-04044-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022]
Abstract
NF-κB is the principle transcription factor and plays the central role in orchestrating chronic inflammation by regulating levels of cytokines, chemokines and growth factors. Piperlongumine (PL), a major alkaloid in the fruit of Piper longum Linn. has gained worldwide attention for its anticancer properties, however, its mechanism of action in the chemoprevention of colon cancer has not been investigated yet. Therefore, the present study was designed to elucidate the underlying molecular mechanism of PL in preventing DMH/DSS induced experimental colon cancer in mice. In the current study well established DMH/DSS induced experimental colon cancer mouse model was used to demonstrate the chemopreventive potential of PL. The expression of NF-κB and its downstream target proteins was evaluated mainly through western blotting. In addition, CAM assay, immunohistochemical staining and gelatin zymography was used to show anti-angiogenic and anti-invasive potential of PL. Additionally, important tumor biomarkers such as TSA, LASA, LDH and IL-6 levels were also estimated. The results of current study showed that PL was capable to inhibit NF-κB activation as well as its nuclear translocation. PL administration to DMH/DSS treated mice also inhibited the NF-κB downstream signaling cascades such as including COX-2 pathway, JAK/STAT pathway, β-catenin, Notch signaling pathway, angiogenesis and epithelial to mesenchymal transition pathway. The findings of the present study have claimed PL as promising chemopreventive agent for colon cancer with pleiotropic action. The current study emphasizes that regular consumption of PL can be an effective approach in the prevention of colon cancer in humans.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Biochemistry, Basic Medical Science, Block-II, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
- Pharmacology and Toxicology Lab, Block J, CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India
| | - Navneet Agnihotri
- Department of Biochemistry, Basic Medical Science, Block-II, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
8
|
Dwyer Z, Chaiquin M, Landrigan J, Ayoub K, Shail P, Rocha J, Childers CL, Storey KB, Philpott DJ, Sun H, Hayley S. The impact of dextran sodium sulphate and probiotic pre-treatment in a murine model of Parkinson's disease. J Neuroinflammation 2021; 18:20. [PMID: 33422110 PMCID: PMC7796536 DOI: 10.1186/s12974-020-02062-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recent work has established that Parkinson's disease (PD) patients have an altered gut microbiome, along with signs of intestinal inflammation. This could help explain the high degree of gastric disturbances in PD patients, as well as potentially be linked to the migration of peripheral inflammatory factors into the brain. To our knowledge, this is the first study to examine microbiome alteration prior to the induction of a PD murine model. METHODS We presently assessed whether pre-treatment with the probiotic, VSL #3, or the inflammatory inducer, dextran sodium sulphate (DSS), would influence the PD-like pathology provoked by a dual hit toxin model using lipopolysaccharide (LPS) and paraquat exposure. RESULTS While VSL #3 has been reported to have anti-inflammatory effects, DSS is often used as a model of colitis because of the gut inflammation and the breach of the intestinal barrier that it induces. We found that VSL#3 did not have any significant effects (beyond a blunting of LPS paraquat-induced weight loss). However, the DSS treatment caused marked changes in the gut microbiome and was also associated with augmented behavioral and inflammatory outcomes. In fact, DSS markedly increased taxa belonging to the Bacteroidaceae and Porphyromonadaceae families but reduced those from Rikencellaceae and S24-7, as well as provoking colonic pro-inflammatory cytokine expression, consistent with an inflamed gut. The DSS also increased the impact of LPS plus paraquat upon microglial morphology, along with circulating lipocalin-2 (neutrophil marker) and IL-6. Yet, neither DSS nor VSL#3 influenced the loss of substantia nigra dopamine neurons or the astrocytic and cytoskeleton remodeling protein changes that were provoked by the LPS followed by paraquat treatment. CONCLUSIONS These data suggest that disruption of the intestinal integrity and the associated microbiome can interact with systemic inflammatory events to promote widespread brain-gut changes that could be relevant for PD and at the very least, suggestive of novel neuro-immune communication.
Collapse
Affiliation(s)
- Zach Dwyer
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Melany Chaiquin
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Jeffrey Landrigan
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Kiara Ayoub
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Pragya Shail
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Julianna Rocha
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Christie L Childers
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Hongyu Sun
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada.
| |
Collapse
|
9
|
Tsatsakis A, Calina D, Falzone L, Petrakis D, Mitrut R, Siokas V, Pennisi M, Lanza G, Libra M, Doukas SG, Doukas PG, Kavali L, Bukhari A, Gadiparthi C, Vageli DP, Kofteridis DP, Spandidos DA, Paoliello MMB, Aschner M, Docea AO. SARS-CoV-2 pathophysiology and its clinical implications: An integrative overview of the pharmacotherapeutic management of COVID-19. Food Chem Toxicol 2020; 146:111769. [PMID: 32979398 PMCID: PMC7833750 DOI: 10.1016/j.fct.2020.111769] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
Common manifestations of COVID-19 are respiratory and can extend from mild symptoms to severe acute respiratory distress. The severity of the illness can also extend from mild disease to life-threatening acute respiratory distress syndrome (ARDS). SARS-CoV-2 infection can also affect the gastrointestinal tract, liver and pancreatic functions, leading to gastrointestinal symptoms. Moreover, SARS-CoV-2 can cause central and peripheral neurological manifestations, affect the cardiovascular system and promote renal dysfunction. Epidemiological data have indicated that cancer patients are at a higher risk of contracting the SARS-CoV-2 virus. Considering the multitude of clinical symptoms of COVID-19, the objective of the present review was to summarize their pathophysiology in previously healthy patients, as well as in those with comorbidities. The present review summarizes the current, though admittedly fluid knowledge on the pathophysiology and symptoms of COVID-19 infection. Although unclear issues still remain, the present study contributes to a more complete understanding of the disease, and may drive the direction of new research. The recognition of the severity of the clinical symptoms of COVID-19 is crucial for the specific therapeutic management of affected patients.
Collapse
Affiliation(s)
- Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece; I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146, Moscow, Russia.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", 80131, Naples, Italy.
| | - Dimitrios Petrakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece.
| | - Radu Mitrut
- Department of Cardiology, University and Emergency Hospital, 050098, Bucharest, Romania.
| | - Vasileios Siokas
- Department of Neurology, University of Thessaly, University Hospital of Larissa, 41221, Larissa, Greece.
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123, Catania, Italy; Department of Neurology IC, Oasi Research Institute-IRCCS, 94018, Troina, Italy.
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123, Catania, Italy.
| | - Sotirios G Doukas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece; Department of Internal Medicine, Saint Peter's University Hospital, 254 Easton Ave, New Brunswick, NJ, 08901, USA.
| | - Panagiotis G Doukas
- University of Pavol Josef Safarik University, Faculty of Medicine, Kosice, Slovakia.
| | - Leena Kavali
- Department of Internal Medicine, Saint Peter's University Hospital, 254 Easton Ave, New Brunswick, NJ, 08901, USA.
| | - Amar Bukhari
- Department of Medicine, Division of Pulmonary and Critical Care 240 Easton Ave, Adult Ambulatory at Cares Building 4th Floor, New Brunswick, NJ, 08901, USA.
| | - Chiranjeevi Gadiparthi
- Division of Gastroenterology, Hepatology and Clinical Nutrition, Saint Peter's University Hospital, New Brunswick, NJ, USA.
| | - Dimitra P Vageli
- Department of Surgery, The Yale Larynx Laboratory, New Haven, CT, 06510, USA.
| | - Diamantis P Kofteridis
- Department of Internal Medicine, University Hospital of Heraklion, 71110, Heraklion, Crete, Greece.
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, 71003, Greece.
| | - Monica M B Paoliello
- Department of Molecular Pharmacology, Albert Eisntein College of Medicine, 1300 Morris Park Avenue Bronx, NY, 10461, USA.
| | - Michael Aschner
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146, Moscow, Russia; Department of Molecular Pharmacology, Albert Eisntein College of Medicine, 1300 Morris Park Avenue Bronx, NY, 10461, USA.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
10
|
Singh A, Devkar R, Basu A. Myeloid Differentiation Primary Response 88-Cyclin D1 Signaling in Breast Cancer Cells Regulates Toll-Like Receptor 3-Mediated Cell Proliferation. Front Oncol 2020; 10:1780. [PMID: 33072559 PMCID: PMC7531238 DOI: 10.3389/fonc.2020.01780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/11/2020] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 3 (TLR3)-mediated apoptotic changes in cancer cells are well-documented, and hence, several synthetic ligands of TLR3 are being used for adjuvant therapy, but there are reports showing a contradictory effect of TLR3 signaling, which include our previous report that had shown cell proliferation following surface localization of TLR 3. However, the underlying mechanism of cell surface localization of TLR3 and subsequent cell proliferation lacks clarity. This study addresses the TLR3 ligand-mediated signaling cascade that regulates a proliferative effect in breast cancer cells (MDA-MB-231 and T47D) challenged with TLR3 ligand in the presence of myeloid differentiation primary response 88 (MyD88) inhibitor. Evidences were obtained using immunoblotting, coimmunoprecipitation, confocal microscopy, immunocytochemistry, ELISA, and flow cytometry. Results had revealed that TLR3 ligand treatment significantly enhanced breast cancer cell proliferation marked by an upregulated expression of cyclinD1, but the same was suppressed by the addition of MyD88 inhibitor. Also, expression of interleukin 1 receptor-associated kinase 1 (IRAK1)-TNF receptor-associated factor 6 (TRAF6)-transforming growth factor beta-activated kinase 1 (TAK1) was altered in the given TLR3-signaling pathway. Inhibition of MyD88 disrupted the downstream adaptor complex and mediated signaling through the TLR3-MyD88-NF-κB (p65)-IL-6-cyclin D1 pathway. TLR3-mediated alternative signaling of the TLR3-MyD88-IRAK1-TRAF6-TAK1-TAB1-NF-κB axis leads to upregulation of IL6 and cyclin D1. This response is hypothesized to be via the MyD88 gateway that culminates in the proliferation of breast cancer cells. Overall, this study provides first comprehensive evidence on the involvement of canonical signaling of TLR3 using MyD88-cyclin D1-mediated breast cancer cell proliferation. The findings elucidated herein will provide valuable insights into understanding the TLR3-mediated adjuvant therapy in cancer.
Collapse
Affiliation(s)
- Aradhana Singh
- Molecular Biology and Human Genetics Laboratory, Department of Zoology, The University of Burdwan, Bardhaman, India
| | - Ranjitsinh Devkar
- Department of Zoology, Faculty of Science, The M.S. University of Baroda, Vadodara, India
| | - Anupam Basu
- Molecular Biology and Human Genetics Laboratory, Department of Zoology, The University of Burdwan, Bardhaman, India
| |
Collapse
|
11
|
Dysregulation of IL6/IL6R-STAT3-SOCS3 signaling pathway in IBD-associated colorectal dysplastic lesions as compared to sporadic colorectal adenomas in non-IBD patients. Pathol Res Pract 2020; 216:153211. [PMID: 32979687 DOI: 10.1016/j.prp.2020.153211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND IL6-IL6R-STAT3-SOCS3 signaling pathway is known to play important roles in regulating intestinal epithelial homeostasis, in pathogenesis of inflammatory bowel disease (IBD), and in tumorigenesis of colorectal neoplasia. We studied the expressions of these factors in IBD-associated dysplasia and compared to sporadic colorectal adenomas in non-IBD individuals. MATERIALS AND METHODS The expression of IL6, IL6R, STAT3, and SOCS3 within dysplastic as well as background non-dysplastic epithelial cells was evaluated by immunohistochemistry in 26 sporadic colorectal adenomas in non-IBD patients, 32 adenoma-like and 30 non-adenoma-like dysplastic lesions in IBD (41 ulcerative colitis, 21 Crohn's disease) patients. The level of expression of each factor was arbitrarily scored as 0, 1, 2, and 3. RESULTS In both IBD and non-IBD lesions, neoplastic epithelium showed a higher expression of all factors, except the IL6R, as compared to non-neoplastic epithelium. For non-neoplastic epithelium between IBD and non-IBD settings, the colitic epithelium showed a similar IL6, lower IL6R, higher STAT3, and higher SOCS3 expression. As compared to non-IBD adenomas, IBD-associated dysplasia showed a significantly lower IL6, lower IL6R, higher STAT3, and lower SOCS3 expression. Most notably, a parallel-elevation pattern of STAT3/SOCS3 expressions was seen in non-IBD adenomas but an inverse-expression pattern of STAT3/SOCS3 seen in IBD dysplastic lesions. No significant differences existed between adenoma-like and non-adenoma-like lesions. CONCLUSIONS IL6/IL6R-STAT3-SOCS3 signaling pathway does not seem to be preferentially associated with IBD-associated dysplasia. However, the STAT3-SOCS3 interaction appears dysregulated in IBD, characterized by a loss of STAT3/SOCS3 balance,i.e., loss of the normal negative regulation of SOCS3.
Collapse
|
12
|
Fei X, Zhang P, Pan Y, Liu Y. MicroRNA-98-5p Inhibits Tumorigenesis of Hepatitis B Virus-Related Hepatocellular Carcinoma by Targeting NF-κB-Inducing Kinase. Yonsei Med J 2020; 61:460-470. [PMID: 32469170 PMCID: PMC7256008 DOI: 10.3349/ymj.2020.61.6.460] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/23/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023] Open
Abstract
PURPOSE MicroRNAs play key regulatory roles in the tumorigenesis of hepatitis B virus-related hepatocellular carcinoma (HBV-HCC). This study aimed to explore the regulatory effects of microRNA-98-5p (miR-98-5p) on the proliferation, migration, invasion, and apoptosis of HBV-HCC cells, as well as the underlying mechanisms involving nuclear factor-κB-inducing kinase (NIK). MATERIALS AND METHODS The expressions of miR-98-5p and NIK in HBV-HCC tissues and cells, and the level of HBV DNA in HBV-HCC cells were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation, migration, invasion, and apoptosis of HBV-HCC cells were analyzed by cell counting kit-8, wound healing, transwell, and flow cytometry assay, respectively. The targeting relationship between miR-98-5p and NIK was predicted by StarBase3.0 and verified by dual-luciferase reporter assay. HBV-HCC xenograft tumor model was constructed in mice to observe the tumor growth in vivo. RESULTS The expression of miR-98-5p was declined in HBV-HCC tissues and cells. Overexpression of miR-98-5p markedly reduced the level of HBV DNA; inhibited the proliferation, migration, and invasion; and promoted the apoptosis of HBV-HCC cells. NIK was a target of miR-98-5p. Overexpression of miR-98-5p markedly decreased the protein expression of NIK in MHCC97H-HBV cells. NIK reversed the tumor-suppressing effect of miR-98-5p on HBV-HCC cells. Furthermore, overexpression of miR-98-5p significantly inhibited the xenograft tumor growth and decreased the expression of NIK in mice. CONCLUSION MiR-98-5p inhibits the secretion of HBV, proliferation, migration, and invasion of HBV-HCC cells by targeting NIK.
Collapse
Affiliation(s)
- Xiukun Fei
- Department of Infectious Diseases, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, China
| | - Peipei Zhang
- Department of Liver Disease, Zaozhuang Traditional Chinese Medicine Hospital, Zaozhuang, China
| | - Yu Pan
- Department of Infectious Diseases, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, China
| | - Yuanyuan Liu
- Department of Infectious Diseases, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, China.
| |
Collapse
|
13
|
IL-1β/IL-6 network in the tumor microenvironment of human colorectal cancer. Pathol Res Pract 2018; 214:986-992. [PMID: 29803656 DOI: 10.1016/j.prp.2018.05.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/28/2018] [Accepted: 05/15/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Recent studies suggest that the interaction between interleukin (IL)-1β and IL-6 in the microenvironment might be involved in the development and progression of human colorectal cancer (CRC). However, the expression of IL-1β/IL-6 network within the CRC microenvironment is not fully understood. MATERIALS AND METHODS The level of IL-1β/IL-6 network expression in 40 biopsies of sporadic CRC and 15 biopsies of controls was assessed using quantitative real-time polymerase chain reaction (PCR) assay, immunohistochemistry (IHC) and double immunofluorescence staining. RESULTS Quantitative results obtained by real-time PCR revealed that both IL-1β and IL-6 mRNA expressions were increased in CRC tissues compared with expressions in controls. In which, IL-6 mRNA expression in primary CRC tissues showed a statistically significant relationship with tumor invasion depth. IHC observations confirmed that increased expression of IL-1β and IL-6 immunoreactivities was located in both the CRC epithelium and stroma. Furthermore, IHC results also revealed that increased expression of IL-1β receptor type 1 (IL-1R1) and IL-6 receptor (IL-6R) were observed in both CRC epithelial and stromal cells. IHCs in serial CRC sections and double immunofluorescence staining revealed a highly co-expression of IL-1R1 immunoreactivity with IL-6 immunoreactivity in the same cells, which confirmed a histological fundament of IL-1β/IL-6 network. CONCLUSION The IL-1β/IL-6 network is highly expressed in the CRC microenvironment, indicating that this network is important in the progression of CRC.
Collapse
|
14
|
Zhang MJ, Su H, Yan JY, Li N, Song ZY, Wang HJ, Huo LG, Wang F, Ji WS, Qu XJ, Qu MH. Chemopreventive effect of Myricetin, a natural occurring compound, on colonic chronic inflammation and inflammation-driven tumorigenesis in mice. Biomed Pharmacother 2017; 97:1131-1137. [PMID: 29136951 DOI: 10.1016/j.biopha.2017.11.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023] Open
Abstract
Myricetin is a flavonoids compound extracted from edible myrica rubra. We aimed to evaluate the efficacy of Myricetin on colonic chronic inflammation and inflammation-driven tumorigenesis in mice. Myricetin was administrated by gavage for 4 consecutive weeks. Mice were sacrificed and the number of colonic polyps was counted. Myricetin significantly inhibited AOM/DSS-induced colitis and colorectal tumorigenesis. Myricetin prevented the incidence of colorectal tumorigenesis and reduced the size of colorectal polyps. Histopathologic analysis showed that Myricetin could attenuate the degree of colonic inflammation and colorectal tumorigenesis. Further analysis showed that Myricetin strongly reduced the levels of inflammatory factors TNF-α, IL-1β, IL-6, NF-κB, p-NF-κB, cyclooxygenase-2 (COX-2), PCNA and Cyclin D1 in the colonic tissues as analyzed by the assays of immunohistochemical staining, Western blotting and Q-RT-PCR. Our results demonstrated that Myricetin possesses the biological activities of chemoprevention colonic chronic inflammation and inflammation-driven tumorigenesis. We suggest that Myricetin could be developed as a promising chemopreventive drug for reducing the risk of colorectal cancer.
Collapse
Affiliation(s)
- Mei-Jia Zhang
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Han Su
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Jing-Yue Yan
- Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Na Li
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Zhi-Yu Song
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Huai-Jie Wang
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Lian-Guang Huo
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Feng Wang
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Wan-Sheng Ji
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China.
| | - Xian-Jun Qu
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China; Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Mei-Hua Qu
- Department of Pharmacology, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
15
|
Liu D, Miao H, Zhao Y, Kang X, Shang S, Xiang W, Shi R, Hou A, Wang R, Zhao K, Liu Y, Ma Y, Luo H, Miao H, He F. NF-κB potentiates tumor growth by suppressing a novel target LPTS. Cell Commun Signal 2017; 15:39. [PMID: 29017500 PMCID: PMC5634951 DOI: 10.1186/s12964-017-0196-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023] Open
Abstract
Background Chronic inflammation is causally linked to the carcinogenesis and progression of most solid tumors. LPTS is a well-identified tumor suppressor by inhibiting telomerase activity and cancer cell growth. However, whether and how LPTS is regulated by inflammation signaling is still incompletely elucidated. Methods Real-time PCR and western blotting were used to determine the expression of p65 and LPTS. Reporter gene assay, electrophoretic mobility shift assay and chromatin immunoprecipitation were performed to decipher the regulatory mechanism between p65 and LPTS. Cell counting kit-8 assays and xenograt models were used to detect p65-LPTS-regulated cancer cell growth in vitro and in vivo, respectively. Results Here we for the first time demonstrated that NF-κB could inhibit LPTS expression in the mRNA and protein levels in multiple cancer cells (e.g. cervical cancer and colon cancer cells). Mechanistically, NF-κB p65 could bind to two consensus response elements locating at −1143/−1136 and −888/−881 in the promoter region of human LPTS gene according to EMSA and ChIP assays. Mutation of those two binding sites rescued p65-suppressed LPTS promoter activity. Functionally, NF-κB regulated LPTS-dependent cell growth of cervical and colon cancers in vitro and in xenograft models. In translation studies, we verified that increased p65 expression was associated with decreased LPTS level in multiple solid cancers. Conclusions Taken together, we revealed that NF-κB p65 potentiated tumor growth via suppressing a novel target LPTS. Modulation of NF-κB-LPTS axis represented a potential strategy for treatment of those inflammation-associated malignancies.
Collapse
Affiliation(s)
- Dongbo Liu
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Hongping Miao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuanyin Zhao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Shenglan Shang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Wei Xiang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Rongchen Shi
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Along Hou
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yingzhe Liu
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yue Ma
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Huan Luo
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
16
|
Huang FT, Peng JF, Cheng WJ, Zhuang YY, Wang LY, Li CQ, Tang J, Chen WY, Li YH, Zhang SN. MiR-143 Targeting TAK1 Attenuates Pancreatic Ductal Adenocarcinoma Progression via MAPK and NF-κB Pathway In Vitro. Dig Dis Sci 2017; 62:944-957. [PMID: 28194669 DOI: 10.1007/s10620-017-4472-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/20/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Transforming growth factor (TGF)-β-activated kinase 1 (TAK1) is one of the major regulators of inflammation-induced cancer cell growth and progression. MiR-143 dysregulation is a common event in a variety of human diseases including pancreatic ductal adenocarcinoma (PDA). AIMS To identify the interaction between TAK1 and miR-143 in PDA. METHODS Data mining of TAK1 expression in PDA patient gene profiling was conducted. QRT-PCR and western blot were performed to detect the expression of TAK1 in PDA tissues and cell lines. Ectopic miR-143 and TAK1 were introduced to PDA cells. Cell growth, apoptosis and migration were examined. Xenograft models were used to examine the function of TAK1 in vivo. Western blot and luciferase assay were carried out to investigate the direct target of miR-143. RESULTS PDA patient gene profiling data (GSE15471 and GSE16515) showed that TAK1 mRNA was aberrantly up-regulated in PDA tissues. TAK1 protein levels were overexpressed in PDA tissues and cell lines. Overexpression of TAK1 was strongly associated with positive lymph node metastasis. Inhibition of TAK1 suppressed cell growth, migration, and induced cell apoptosis in vitro and in vivo. Further studies demonstrated that TAK1 was a direct target gene of miR-143. MiR-143 also inhibited PDA cells proliferation and migration, induced apoptosis and G1/S arrest. Moreover, TAK1 depletion inactivated MAPK and NF-κB pathway, mimicking the function of miR-143. CONCLUSIONS The study highlights that miR-143 acts as a tumor suppressor in PDA through directly targeting TAK1, and their functional regulation may provide potential therapeutic strategies in clinics.
Collapse
Affiliation(s)
- Feng-Ting Huang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Juan-Fei Peng
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Wen-Jie Cheng
- Department of Ultrasound, the Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong Province, People's Republic of China
| | - Yan-Yan Zhuang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Ling-Yun Wang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Chu-Qiang Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Jian Tang
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong Province, People's Republic of China
| | - Wen-Ying Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Yuan-Hua Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China
| | - Shi-Neng Zhang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, Guangdong Province, People's Republic of China.
| |
Collapse
|
17
|
Zeng J, Tang ZH, Liu S, Guo SS. Clinicopathological significance of overexpression of interleukin-6 in colorectal cancer. World J Gastroenterol 2017; 23:1780-1786. [PMID: 28348483 PMCID: PMC5352918 DOI: 10.3748/wjg.v23.i10.1780] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/06/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To compare the expression levels of interleukin (IL)-6 in colorectal cancer (CRC) tissues and adjacent non-cancerous tissues, and analyse the correlation of IL-6 expression with the clinicopathological parameters of CRC.
METHODS Fifty CRC tissue specimens and 50 matched adjacent mucosa specimens were collected. The expression of IL-6 in these clinical samples was examined by immunohistochemical staining. The correlation between IL-6 expression and clinicopathological parameters was assessed by statistical analysis.
RESULTS IL-6 expression was significantly elevated in CRC tissues compared with noncancerous tissues (P < 0.001). IL-6 expression was positively correlated with tumour TNM stage (P < 0.001), but a negative correlation was detected between IL-6 expression and tumor histological differentiation in CRC (P < 0.05). Furthermore, IL-6 expression was associated with invasion depth and lymph node metastasis in CRC.
CONCLUSION IL-6 might be a useful marker for predicting a poor prognosis in patients with CRC and might be used as a potential therapeutic target in CRC.
Collapse
|
18
|
Zhang K, Zhao P, Guo G, Guo Y, Tian L, Sun X, Li S, He Y, Sun Y, Chai H, Zhang W, Xing M. Arsenic Trioxide Attenuates NF-κB and Cytokine mRNA Levels in the Livers of Cocks. Biol Trace Elem Res 2016; 170:432-7. [PMID: 26276563 DOI: 10.1007/s12011-015-0455-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/22/2015] [Indexed: 01/03/2023]
Abstract
Arsenic (As) is a trace element widely found in nature. It exists in several forms, including organic arsenic, inorganic arsenic, and trivalent arsenic, the most toxic. Arsenic trioxide (As2O3) is widespread in nature. This form tends to accumulate in animals and humans and therefore has a potential harm for them. Cytokines play essential roles in the immune response and inflammation. Although the importance of cytokines in the responses to arsenic exposure has been demonstrated in many types of mammals, the function of these in poultry, especially in chickens, remains unclear. The purpose of the present study was to examine the effect of As2O3 exposure on cytokines in cock livers. In this study, 72 1-day-old male Hy-line cocks were randomly divided into four groups including the control group, low-As group, middle-As group, and high-As group. The livers were collected on days 30, 60, and 90 of the experiment. The levels of nuclear factor-kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-12 beta (IL-12β), and interleukin-1 beta (IL-1β) mRNA in the livers of the cocks were measured using real-time PCR. The results showed that the expression levels of IL-6, IL-8, TNF-α, and NF-κB which seemed to be a critical mediator in the inflammatory response tended to increase in the birds chronically treated with As2O3. However, the mRNA expression levels of IL-4, IL-12β, and IL-1β were decreased in the experiment. The information regarding the effects of As2O3 on cytokine mRNA expression generated in this study will be important information for arsenic toxicology evaluation.
Collapse
Affiliation(s)
- Kexin Zhang
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Panpan Zhao
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Guangyang Guo
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Ying Guo
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Li Tian
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Xiao Sun
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Siwen Li
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Ying He
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Ying Sun
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Hongliang Chai
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China
| | - Wen Zhang
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China.
| | - Mingwei Xing
- College of Wildlife Resources, Northeast Forestry University, 26 Hexing Road, Harbin, 150040, Heilongjiang Province, China.
| |
Collapse
|
19
|
Castillo EF, Ray AL, Beswick EJ. MK2: an unrecognized regulator of tumor promoting macrophages in colorectal cancer? ACTA ACUST UNITED AC 2016; 3. [PMID: 26998523 PMCID: PMC4798244 DOI: 10.14800/macrophage.1166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and is associated closely with inflammation before and after development. Macrophages promote colitis and colitis-associated CRC. M1 macrophages contribute to colitis directly through the production of proinflammatory cytokines and through activation of proinflammatory immune cell phenotypes. In cancer, both M1 and M2 macrophages participate in tumor development and progression through cytokine production, changes in cell signaling and activation of T cells. We have identified the mitogen-activated protein kinase-activated protein kinase 2 (MK2) as a regulator of macrophages during colitis-associated CRC (CAC). MK2 is a proinflammatory kinase that promotes production of IL-1α, IL-1β, IL-6 and TNF-α. MK2−/− mice have decreases in macrophages, macrophage-associated chemokines, and proinflammatory cytokines. Most significantly, MK2−/− mice do not develop neoplasms in an inflammatory model of CRC. However, addition of MK2+/+ macrophages to MK2−/− mice increases production of proinflammatory cytokines. In wild type mice, both cytokines and tumor burdens increase upon addition of additional macrophages. These data support the importance of MK2 in macrophage regulation during inflammation-associated CRC.
Collapse
Affiliation(s)
- Eliseo F Castillo
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Anita L Ray
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
20
|
QU L, HE L, ZHAO X, XU W. Downregulation of miR-518a-3p activates the NIK-dependent NF-κB pathway in colorectal cancer. Int J Mol Med 2015; 35:1266-72. [PMID: 25812680 PMCID: PMC4380201 DOI: 10.3892/ijmm.2015.2145] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/11/2015] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the biological role and underlying mechanisms of action of miR-518a-3p in the progression and invasion of colorectal cancer (CRC). Reverse transcription-quantitative PCR (RT-qPCR) was used to examine the mRNA expression levels of miR-518a-3p in 5 CRC cell lines (SW480, SW620, HCT116, HT29 and LoVo) in a normal colonic cell line, NCM460, as well as in tumor tissues with or without metastases. The biological effects of miR-518a-3p were assessed in the CRC cell lines by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometric analysis, and RT‑qPCR and western blot analyses were employed to evaluate the expression of miR-518a-3p targets. The regulation of NF-κB-inducing kinase (NIK) by miR-518a-3p was confirmed using luciferase activity assays. Our results revealed that miR-518a-3p was significantly downregulated in the CRC cell lines compared with the normal colonic cell line (P<0.05), as well as in the CRC tissues with distant metastases compared with the tissues without metastases. The downregulation of miR-518a-3p was associated with tumor size, distant metastasis and TNM stage in the patients with CRC. Moreover, the ectopic expression of miR-518a-3p and the inhibition of NIK by RNA interference markedly reduced cell proliferation and enhanced the apoptosis of CRC cells. Further experiments revealed that NIK, a regulator of NF-κB, was a downstream target of miR-518a-3p. The presents findings indicate that miR-518a-3p plays an important role in the progression of CRC by targeting NIK.
Collapse
Affiliation(s)
- L.L. QU
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - L. HE
- Department of Gastroenterology Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - X. ZHAO
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - W. XU
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|