1
|
Cruceriu D, Balacescu L, Baldasici O, Gaal OI, Balacescu O, Russom A, Irimia D, Tudoran O. Gene expression-phenotype association study reveals the dual role of TNF-α/TNFR1 signaling axis in confined breast cancer cell migration. Life Sci 2024; 354:122982. [PMID: 39151886 DOI: 10.1016/j.lfs.2024.122982] [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: 04/29/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
AIMS While enhanced tumor cell migration is a key process in the tumor dissemination, mechanistic insights into causal relationships between tumor cells and mechanical confinement are still limited. Here we combine the use of microfluidic platforms to characterize confined cell migration with genomic tools to systematically unravel the global signaling landscape associated with the migratory phenotype of breast cancer (BC) cells. METERIALS AND METHODS The spontaneous migration capacity of seven BC cell lines was evaluated in 3D microfluidic devices and their migration capacity was correlated with publicly available molecular signatures. The role of identified signaling pathways on regulating BC migration capacity was determined by receptor stimulation through ligand binding or inhibition through siRNA silencing. Downstream effects on cell migration were evaluated in microfluidic devices, while the molecular changes were monitored by RT-qPCR. KEY FINDINGS Expression of 715 genes was correlated with BC cells migratory phenotype, revealing TNF-α as one of the top upstream regulators. Signal transduction experiments revealed that TNF-α stimulates the confined migration of triple negative, mesenchymal-like BC cells that are also characterized by high TNFR1 expression, but inhibits the migration of epithelial-like cells with low TNFR1 expression. TNFR1 was strongly associated with the migration capacity and triple-negative, mesenchymal phenotype. Downstream of TNF/TNFR1 signaling, transcriptional regulation of NFKB seems to be important in driving cell migration in confined spaces. SIGNIFICANCE TNF-α/TNFR1 signaling axis reveals as a key player in driving BC cells confined migration, emerging as a promising therapeutic strategy in targeting dissemination and metastasis of triple negative, mesenchymal BC cells.
Collapse
Affiliation(s)
- Daniel Cruceriu
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania; "Babes-Bolyai" University, Department of Molecular Biology and Biotechnology, 1 Mihail Kogalniceanu Street, Cluj-Napoca, Romania.
| | - Loredana Balacescu
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania.
| | - Oana Baldasici
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania.
| | - Orsolya Ildiko Gaal
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania; Iuliu Hațieganu University of Medicine and Pharmacy, Department of Medical Genetics, 8 Victor Babes Street, Cluj-Napoca, Romania.
| | - Ovidiu Balacescu
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania.
| | - Aman Russom
- KTH Royal Institute of Technology, Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, Tomtebodavägen 23a 171 65, Solna, Sweden.
| | - Daniel Irimia
- Harvard Medical School, Center for Engineering in Medicine and Surgery, Department of Surgery, 51 Blossom Street, Boston, MA, United States of America.
| | - Oana Tudoran
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Department of Genetics, Genomics and Experimental Pathology, 34-36 Republicii Street, Cluj-Napoca, Romania; KTH Royal Institute of Technology, Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, Tomtebodavägen 23a 171 65, Solna, Sweden.
| |
Collapse
|
2
|
Sharma B, Dhiman C, Hasan GM, Shamsi A, Hassan MI. Pharmacological Features and Therapeutic Implications of Plumbagin in Cancer and Metabolic Disorders: A Narrative Review. Nutrients 2024; 16:3033. [PMID: 39275349 PMCID: PMC11397539 DOI: 10.3390/nu16173033] [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: 08/16/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/16/2024] Open
Abstract
Plumbagin (PLB) is a naphthoquinone extracted from Plumbago indica. In recent times, there has been a growing body of evidence suggesting the potential importance of naphthoquinones, both natural and artificial, in the pharmacological world. Numerous studies have indicated that PLB plays a vital role in combating cancers and other disorders. There is substantial evidence indicating that PLB may have a significant role in the treatment of breast cancer, brain tumours, lung cancer, hepatocellular carcinoma, and other conditions. Moreover, its potent anti-oxidant and anti-inflammatory properties offer promising avenues for the treatment of neurodegenerative and cardiovascular diseases. A number of studies have identified various pathways that may be responsible for the therapeutic efficacy of PLB. These include cell cycle regulation, apoptotic pathways, ROS induction pathways, inflammatory pathways, and signal transduction pathways such as PI3K/AKT/mTOR, STAT3/PLK1/AKT, and others. This review aims to provide a comprehensive analysis of the diverse pharmacological roles of PLB, examining the mechanisms through which it operates and exploring its potential applications in various medical conditions. In addition, we have conducted a review of the various formulations that have been reported in the literature with the objective of enhancing the efficacy of the compound. However, the majority of the reviewed data are based on in vitro and in vivo studies. To gain a comprehensive understanding of the safety and efficacy of PLB in humans and to ascertain its potential integration into therapeutic regimens for cancer and chronic diseases, rigorous clinical trials are essential. Finally, by synthesizing current research and identifying gaps in knowledge, this review seeks to enhance our understanding of PLB and its therapeutic prospects, paving the way for future studies and clinical applications.
Collapse
Affiliation(s)
- Bhoomika Sharma
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Chitra Dhiman
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Md Imtiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
3
|
Mehrtabar E, Khalaji A, Pandeh M, Farhoudian A, Shafiee N, Shafiee A, Ojaghlou F, Mahdavi P, Soleymani-Goloujeh M. Impact of microRNA variants on PI3K/AKT signaling in triple-negative breast cancer: comprehensive review. Med Oncol 2024; 41:222. [PMID: 39120634 DOI: 10.1007/s12032-024-02469-4] [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: 06/10/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Breast cancer (BC) is a significant cause of cancer-related mortality, and triple-negative breast cancer (TNBC) is a particularly aggressive subtype associated with high mortality rates, especially among younger females. TNBC poses a considerable clinical challenge due to its aggressive tumor behavior and limited therapeutic options. Aberrations within the PI3K/AKT pathway are prevalent in TNBC and correlate with increased therapeutic intervention resistance and poor outcomes. MicroRNAs (miRs) have emerged as crucial PI3K/AKT pathway regulators influencing various cellular processes involved in TNBC pathogenesis. The levels of miRs, including miR-193, miR-4649-5p, and miR-449a, undergo notable changes in TNBC tumor tissues, emphasizing their significance in cancer biology. This review explored the intricate interplay between miR variants and PI3K/AKT signaling in TNBC. The review focused on the molecular mechanisms underlying miR-mediated dysregulation of this pathway and highlighted specific miRs and their targets. In addition, we explore the clinical implications of miR dysregulation in TNBC, particularly its correlation with TNBC prognosis and therapeutic resistance. Elucidating the roles of miRs in modulating the PI3K/AKT signaling pathway will enhance our understanding of TNBC biology and unveil potential therapeutic targets. This comprehensive review aims to discuss current knowledge and open promising avenues for future research, ultimately facilitating the development of precise and effective treatments for patients with TNBC.
Collapse
Affiliation(s)
- Ehsan Mehrtabar
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Pandeh
- School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Aram Farhoudian
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nadia Shafiee
- Children's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefe Shafiee
- Board-Certified Cardiologist, Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ojaghlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parinaz Mahdavi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Soleymani-Goloujeh
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
4
|
Zhuang Z, Chen Y, Zheng J, Chen S. The role of TRIF protein in regulating the proliferation and antigen presentation ability of myeloid dendritic cells through the ERK1/2 signaling pathway in chronic low-grade inflammation of intestinal mucosa mediated by flagellin-TLR5 complex signal. PeerJ 2024; 12:e16716. [PMID: 38188180 PMCID: PMC10768658 DOI: 10.7717/peerj.16716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Objective The objective is to explore whether the flagellin-TLR5 complex signal can enhance the antigen presentation ability of myeloid DCs through the TRIF-ERK1/2 pathway, and the correlation between this pathway and intestinal mucosal inflammation response. Methods Mouse bone marrow-derived DC line DC2.4 was divided into four groups: control group (BC) was DC2.4 cells cultured normally; flagellin single signal stimulation group (DC2.4+CBLB502) was DC2.4 cells stimulated with flagellin derivative CBLB502 during culture; TLR5-flagellin complex signal stimulation group (ov-TLR5-DC2.4+CBLB502) was flagellin derivative CBLB502 stimulated ov-TLR5-DC2.4 cells with TLR5 gene overexpression; TRIF signal interference group (ov-TLR5-DC2.4+CBLB502+Pepinh-TRIFTFA) was ov-TLR5-DC2.4 cells with TLR5 gene overexpression stimulated with flagellin derivative CBLB502 and intervened with TRIF-specific inhibitor Pepinh-TRIFTFA. WB was used to detect the expression of TRIF and p-ERK1/2 proteins in each group of cells; CCK8 was used to detect cell proliferation in each group; flow cytometry was used to detect the expression of surface molecules MHCI, MHCII, CD80, 86 in each group of cells; ELISA was used to detect the levels of IL-12 and IL-4 cytokines in each group. Results Compared with the BC group, DC2.4+CBLB502 group, and ov-TLR5-DC2.4+CBLB502+Pepinh-TRIFTFA group, the expression of TRIF protein and p-ERK1/2 protein in ov-TLR5-DC2.4+CBLB502 group was significantly upregulated (TRIF: p = 0.02, = 0.007, = 0.048) (ERK1: p < 0.001, =0.0003, = 0.0004; ERK2:p = 0.0003, = 0.0012, = 0.0022). The cell proliferation activity in ov-TLR5-DC2.4+CBLB502 group was enhanced compared with the other groups (p = 0.0001, p < 0.0001, p = 0.0015); at the same time, the expression of surface molecules MHCI, MHCII, CD80, 86 on DCs was upregulated (p < 0.05); and the secretion of IL-12 and IL-4 cytokines was increased, with significant differences (IL-12: p < 0.0001, p < 0.0001, p = 0.0005; IL-4: p = < 0.0001, p = < 0.0001, p = 0.0001). However, the ov-TLR5-DC2.4+CBLB502+Pepinh-TRIFTFA group, which was treated with TRIF signal interference, showed a decrease in intracellular TRIF protein and p-ERK1/2 protein, as well as a decrease in cell proliferation ability and surface stimulation molecules, and a decrease in the secretion of IL-12 and IL-4 cytokines (p < 0.05). Conclusion After stimulation of flagellin protein-TLR5 complex signal, TRIF protein and p-ERK1/2 protein expression in myeloid dendritic cells were significantly up-regulated, accompanied by increased proliferation activity and maturity of DCs, enhanced antigen presentation function, increased secretion of pro-inflammatory cytokines IL-12 and IL-4. This process can be inhibited by the specific inhibitor of TRIF signal, suggesting that the TLR5-TRIF-ERK1/2 pathway may play an important role in abnormal immune response and mucosal chronic inflammation infiltration mediated by flagellin protein in DCs, which can provide a basis for our subsequent animal experiments.
Collapse
Affiliation(s)
- Zhaomeng Zhuang
- Gastroenterology, Zhejiang Chinese Medical University Affifiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yi Chen
- Gastroenterology, Zhejiang Chinese Medical University Affifiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| | - Juanhong Zheng
- Gastroenterology, Zhejiang Chinese Medical University Affifiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| | - Shuo Chen
- Gastroenterology, Zhejiang Chinese Medical University Affifiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| |
Collapse
|
5
|
Yang Y, Liu P, Zhou M, Yin L, Wang M, Liu T, Jiang X, Gao H. Small-molecule drugs of colorectal cancer: Current status and future directions. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166880. [PMID: 37696461 DOI: 10.1016/j.bbadis.2023.166880] [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: 07/26/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023]
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the world's fourth most deadly cancer. CRC, as a genetic susceptible disease, faces significant challenges in optimizing prognosis through optimal drug treatment modalities. In recent decades, the development of innovative small-molecule drugs is expected to provide targeted interventions that accurately address the different molecular characteristics of CRC. Although the clinical application of single-target drugs is limited by the heterogeneity and high metastasis of CRC, novel small-molecule drug treatment strategies such as dual/multiple-target drugs, drug repurposing, and combination therapies can help overcome these challenges and provide new insights for improving CRC treatment. In this review, we focus on the current status of a range of small molecule drugs that are being considered for CRC therapy, including single-target drugs, dual/multiple-target drugs, drug repurposing and combination strategies, which will pave the way for targeting CRC vulnerabilities with small-molecule drugs in future personalized treatment.
Collapse
Affiliation(s)
- Yiren Yang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Pengyu Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Mingyang Zhou
- University of Pennsylvania, Philadelphia, PA 19104-6323, United States
| | - Linzhou Yin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Miao Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ting Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiaowen Jiang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Huiyuan Gao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
6
|
Gorji AE, Ostaszewski P, Urbańska K, Sadkowski T. Does β-Hydroxy-β-Methylbutyrate Have Any Potential to Support the Treatment of Duchenne Muscular Dystrophy in Humans and Animals? Biomedicines 2023; 11:2329. [PMID: 37626825 PMCID: PMC10452677 DOI: 10.3390/biomedicines11082329] [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: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Skeletal muscle is the protein reservoir of our body and an important regulator of glucose and lipid homeostasis. The dystrophin gene is the largest gene and has a key role in skeletal muscle construction and function. Mutations in the dystrophin gene cause Duchenne and Becker muscular dystrophy in humans, mice, dogs, and cats. Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular condition causing progressive muscle weakness and premature death. β-hydroxy β-methylbutyrate (HMB) prevents deleterious muscle responses under pathological conditions, including tumor and chronic steroid therapy-related muscle losses. The use of HMB as a dietary supplement allows for increasing lean weight gain; has a positive immunostimulatory effect; is associated with decreased mortality; and attenuates sarcopenia in elderly animals and individuals. This study aimed to identify some genes, metabolic pathways, and biological processes which are common for DMD and HMB based on existing literature and then discuss the consequences of that interaction.
Collapse
Affiliation(s)
- Abdolvahab Ebrahimpour Gorji
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| | - Piotr Ostaszewski
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| | - Kaja Urbańska
- Department of Morphological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| |
Collapse
|
7
|
Di Paola R, De A, Izhar R, Abate M, Zappavigna S, Capasso A, Perna AF, La Russa A, Capasso G, Caraglia M, Simeoni M. Possible Effects of Uremic Toxins p-Cresol, Indoxyl Sulfate, p-Cresyl Sulfate on the Development and Progression of Colon Cancer in Patients with Chronic Renal Failure. Genes (Basel) 2023; 14:1257. [PMID: 37372437 DOI: 10.3390/genes14061257] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic kidney disease (CKD) induces several systemic effects, including the accumulation and production of uremic toxins responsible for the activation of various harmful processes. Gut dysbiosis has been widely described in CKD patients, even in the early stages of the disease. The abundant discharge of urea and other waste substances into the gut favors the selection of an altered intestinal microbiota in CKD patients. The prevalence of bacteria with fermentative activity leads to the release and accumulation in the gut and in the blood of several substances, such as p-Cresol (p-C), Indoxyl Sulfate (IS) and p-Cresyl Sulfate (p-CS). Since these metabolites are normally eliminated in the urine, they tend to accumulate in the blood of CKD patients proportionally to renal impairment. P-CS, IS and p-C play a fundamental role in the activation of various pro-tumorigenic processes, such as chronic systemic inflammation, the increase in the production of free radicals and immune dysfunction. An up to two-fold increase in the incidence of colon cancer development in CKD has been reported in several studies, although the pathogenic mechanisms explaining this compelling association have not yet been described. Based on our literature review, it appears likely the hypothesis of a role of p-C, IS and p-CS in colon cancer development and progression in CKD patients.
Collapse
Affiliation(s)
- Rossella Di Paola
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Ananya De
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Raafiah Izhar
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Marianna Abate
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anna Capasso
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, TX 75063, USA
| | - Alessandra F Perna
- Nephrology and Dialysis Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Antonella La Russa
- Department of Sperimental Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | | | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem S.c.a.r.l. Research Institute, 83031 Ariano Irpino, Italy
| | - Mariadelina Simeoni
- Nephrology and Dialysis Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| |
Collapse
|
8
|
Shelash Al-Hawary SI, Abdalkareem Jasim S, M Kadhim M, Jaafar Saadoon S, Ahmad I, Romero Parra RM, Hasan Hammoodi S, Abulkassim R, M Hameed N, K Alkhafaje W, Mustafa YF, Javed Ansari M. Curcumin in the treatment of liver cancer: From mechanisms of action to nanoformulations. Phytother Res 2023; 37:1624-1639. [PMID: 36883769 DOI: 10.1002/ptr.7757] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/10/2023] [Accepted: 01/22/2023] [Indexed: 03/09/2023]
Abstract
Liver cancer is the sixth most prevalent cancer and ranks third in cancer-related death, after lung and colorectal cancer. Various natural products have been discovered as alternatives to conventional cancer therapy strategies, including radiotherapy, chemotherapy, and surgery. Curcumin (CUR) with antiinflammatory, antioxidant, and antitumor activities has been associated with therapeutic benefits against various cancers. It can regulate multiple signaling pathways, such as PI3K/Akt, Wnt/β-catenin, JAK/STAT, p53, MAPKs, and NF-ĸB, which are involved in cancer cell proliferation, metastasis, apoptosis, angiogenesis, and autophagy. Due to its rapid metabolism, poor oral bioavailability, and low solubility in water, CUR application in clinical practices is restricted. To overcome these limitations, nanotechnology-based delivery systems have been applied to use CUR nanoformulations with added benefits, such as reducing toxicity, improving cellular uptake, and targeting tumor sites. Besides the anticancer activities of CUR in combating various cancers, especially liver cancer, here we focused on the CUR nanoformulations, such as micelles, liposomes, polymeric, metal, and solid lipid nanoparticles, and others, in the treatment of liver cancer.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-Anbar-Ramadi, Iraq
| | - Mustafa M Kadhim
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, Iraq.,Medical Laboratory Techniques Department, Al-Turath University College, Baghdad, Iraq
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | | | - Noora M Hameed
- Anesthesia Techniques, Al-Nisour University College, Baghdad, Iraq
| | - Waleed K Alkhafaje
- Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| |
Collapse
|
9
|
Euxanthone Suppresses the Proliferation, Migration and Invasion of Human Medulloblastoma Cells by Inhibiting the RANK/RANKL Pathway. Pharmacogn Mag 2023. [DOI: 10.1177/09731296221138649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Background: Euxanthone is a plant-based flavonoid that is mostly isolated from a Chinese medicinal plant, Polygala caudate. This study was designed to evaluate the anticancer effects of euxanthone against human medulloblastoma cells. Materials and Methods: Cell viability was evaluated by CCK-8 and EdU assays. Apoptotic cell percentage was determined by annexin V/PI assay. The mRNA expression was determined by qRT-PCR. Wound healing and transwell assays were used to assess cell migration and invasion. Results: The results revealed aberrant activation of RANK/RANKL pathway in human medulloblastoma tissues and cell lines. Euxanthone suppressed the proliferation of the D425 medulloblastoma cells with comparatively lower cytotoxic effects against the normal human cerebellar granule cells. The IC50 of euxanthone against the D425 cells was found to be 10 µM. Interestingly, silencing of receptor activator of nuclear factor kβ (RANK) could also suppress the proliferation of the D425 cells via induction of apoptosis. Nonetheless, overexpression of RANK could abolish the cytotoxic effects of euxanthone on the D425 cells. Finally, wound-heal and transwell assay showed that euxanthone suppressed the migration and invasion of the D425 medulloblastoma cells. Conclusion: Collectively, the results revealed the anticancer effects of euxanthone against human medulloblastoma cells via RANK/RANKL pathway. These results suggest the potential of euxanthone as a lead molecule in the development of chemotherapy for medulloblastoma.
Collapse
|
10
|
Global Trends in Research of NF-κB in Melanoma from 2000 to 2021: A Study of Bibliometric Analysis. JOURNAL OF ONCOLOGY 2022; 2022:3684228. [PMID: 36124033 PMCID: PMC9482551 DOI: 10.1155/2022/3684228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/03/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022]
Abstract
In the pathogenesis of melanoma, NF-κB is a key signaling pathway. Appling bibliometric analysis, we identify the frontiers and hotspots about NF-κB in melanoma, as well as distinguishing features of scientific research and output all over the world during the past 22 years. 2226 publications published from 2000 to 2021 and related information were retrieved based on Science Citation Index Expanded (SCI-expanded) of Web of Science Core Collection (WoSCC). VOSviewer and Citespace were used to analyze bibliometric indicators and visualize the hotspots and research trend of studies on NF-κB in melanoma. The results indicated that despite fluctuations, the number of publications (Np) related to the research of NF-κB in melanoma per year increased over the past 22 years. The USA had the most publications. H-index and the number of citations (Nc) of the USA were also in the first place. PloS One was the most productive journal, and League of European Research Universities (LERU) was the most productive affiliation. Recently, the keywords “NF-kappa-b,” “melanoma,” “apoptosis,” “expression,” “activation,” “cancer,” and “metastasis” appeared most frequently. Our study suggested that articles associated with NF-κB in melanoma tend to increase. In this field, the USA was an influential country and a big producer. Most publications focused on clinical and basic research in the past 22 years, and keywords “tumor necrosis factor” and “trail induced apoptosis” had the highest burst strength.
Collapse
|
11
|
Yang X, Niu Z, Wang X, Lu X, Sun J, Carpena M, Prieto M, Simal-Gandara J, Xiao J, Liu C, Li N. The Nutritional and Bioactive Components, Potential Health Function and Comprehensive Utilization of Pomegranate: A Review. FOOD REVIEWS INTERNATIONAL 2022. [DOI: 10.1080/87559129.2022.2110260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Xuhan Yang
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Tai’an, China
| | - Zhonglu Niu
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Tai’an, China
| | - Xiaorui Wang
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Tai’an, China
| | - Xiaoming Lu
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Tai’an, China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - M. Carpena
- Faculty of Science, Department of Analytical Chemistry and Food Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| | - M.A. Prieto
- Faculty of Science, Department of Analytical Chemistry and Food Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| | - Jesus Simal-Gandara
- Faculty of Science, Department of Analytical Chemistry and Food Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| | - Jianbo Xiao
- Faculty of Science, Department of Analytical Chemistry and Food Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Ningyang Li
- Key Laboratory of Food Processing Technology and Quality Control in Shandong Province, College of Food Science and Engineering, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
12
|
Therapeutic effect of the sulforaphane derivative JY4 on ulcerative colitis through the NF-κB-p65 pathway. Inflammopharmacology 2022; 30:1717-1728. [PMID: 35943671 DOI: 10.1007/s10787-022-01044-5] [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: 03/23/2022] [Accepted: 06/22/2022] [Indexed: 11/05/2022]
Abstract
The efficacy of the sulforaphane derivative JY4 was evaluated in acute and chronic mouse models of ulcerative colitis induced by dextran sodium sulfate. Oral administration of JY4 led to significant improvements in symptoms, with recovery of body weight and colorectal length, together with reduced diarrhoea, bloody stools, ulceration of colonic tissue and infiltration of inflammatory cells. The oral bioavailability of JY4, determined by comparing oral dosing with injection into the tail vein, was 5.67%, which was comply with the idea in the intestinal function. Using a dual-luciferase reporter assay, immunofluorescence studies, western blot analysis and immunohistochemical staining, JY4 was shown to significant interfere with the NF-κB-p65 signaling pathway. By preventing the activation of NF-κB-p65, JY4 inhibited the overexpression of downstream inflammatory factors, thereby exerting an anti-inflammatory effect on the intestinal tract. This study thus provides a promising candidate drug, and a new concept for the treatment of ulcerative colitis.
Collapse
|
13
|
Lau TY, Kwan HY. Fucoxanthin Is a Potential Therapeutic Agent for the Treatment of Breast Cancer. Mar Drugs 2022; 20:md20060370. [PMID: 35736173 PMCID: PMC9229252 DOI: 10.3390/md20060370] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers diagnosed and the leading cause of cancer-related death in women. Although there are first-line treatments for BC, drug resistances and adverse events have been reported. Given the incidence of BC keeps increasing, seeking novel therapeutics is urgently needed. Fucoxanthin (Fx) is a dietary carotenoid commonly found in seaweeds and diatoms. Both in vitro and in vivo studies show that Fx and its deacetylated metabolite fucoxanthinol (Fxol) inhibit and prevent BC growth. The NF-κB signaling pathway is considered the major pathway contributing to the anti-proliferation, anti-angiogenesis and pro-apoptotic effects of Fx and Fxol. Other signaling molecules such as MAPK, MMP2/9, CYP and ROS are also involved in the anti-cancer effects by regulating the tumor microenvironment, cancer metastasis, carcinogen metabolism and oxidation. Besides, Fx also possesses anti-obesity effects by regulating UCP1 levels and lipid metabolism, which may help to reduce BC risk. More importantly, mounting evidence demonstrates that Fx overcomes drug resistance. This review aims to give an updated summary of the anti-cancer effects of Fx and summarize the underlying mechanisms of action, which will provide novel strategies for the development of Fx as an anti-cancer therapeutic agent.
Collapse
|
14
|
Xu Q, Yu J, Jia G, Li Z, Xiong H. Crocin attenuates NF-κB-mediated inflammation and proliferation in breast cancer cells by down-regulating PRKCQ. Cytokine 2022; 154:155888. [PMID: 35447530 DOI: 10.1016/j.cyto.2022.155888] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/06/2022] [Accepted: 04/06/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer confronting women worldwide. Crocin, a glycosylated carotenoid extracted from Crocus sativus L., possesses anti-cancer and anti-inflammatory activities. This study tried to explore the influences of crocin on proliferation and inflammation of BC cells, and to investigate the possible mechanism. The protein levels of protein kinase C theta (PRKCQ) and nuclear factor kappa B (NF-κB) p-p65 and p65 were examined using western blot analysis. The potential targets of crocin were predicted using the PharmMapper database. Cell viability and proliferation were determined utilizing CCK-8 and EdU incorporation assays, respectively. Inflammation was assessed by detecting the levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) using RT-qPCR and ELISA. Results showed that crocin inhibited NF-κB activation and suppressed cell viability and proliferation in BC cells. Crocin caused a significant reduction of levels of TNF-α and IL-1β, suggesting that crocin suppressed inflammation in BC cells. NF-κB inhibition decreased proliferation and inflammation in BC cells. Additionally, PRKCQ was identified as a potential target of crocin according to PharmMapper database. Crocin treatment inhibited the activation of NF-κB in BC cells by reducing PRKCQ expression. Mechanistically, PRKCQ-dependent activation of NF-κB pathway reversed the effects of crocin on the proliferation and inflammation in BC cells. In conclusion, crocin inhibited NF-κB-mediated inflammation and proliferation in BC cells through reducing PRKCQ expression.
Collapse
Affiliation(s)
- Quanxiao Xu
- Department of Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Jinsong Yu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Guangwei Jia
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China.
| | - Zhong Li
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| | - Hui Xiong
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China; Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang 473004, China
| |
Collapse
|
15
|
Wang J, Wen B, Zeng Y, Wang H, Zhao W, Zhou Y, Liu L, Wang P, Pan K, Jing B, Ni X, Zeng D. Assessment the role of some Bacillus strains in improvement rex rabbits resistance against ETEC challenge. Microb Pathog 2022; 165:105477. [DOI: 10.1016/j.micpath.2022.105477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/05/2021] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
|
16
|
Aoyama T, Yoshikawa T, Ida S, Cho H, Sakamaki K, Ito Y, Fujitani K, Takiguchi N, Kawashima Y, Nishikawa K, Nunobe S, Hiki N. Effects of perioperative eicosapentaenoic acid‑enriched oral nutritional supplement on the long‑term oncological outcomes after total gastrectomy for gastric cancer. Oncol Lett 2022; 23:151. [PMID: 35836480 PMCID: PMC9258592 DOI: 10.3892/ol.2022.13272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Basic and clinical reports have suggested that eicosapentaenoic acid (EPA) exhibits anti-tumor activity. The present study evaluated whether perioperative EPA could improve the survival of patients with localized gastric cancer as a key secondary endpoint of a randomized clinical study. The present study was designed as multicenter, open-label, superiority, randomized trial to confirm the preventive effect of EPA on body weight loss after total gastrectomy for gastric cancer. Eligible patients were randomized to either the standard-diet group (EPA-off group) or EPA-on group by a centralized dynamic method. An EPA-enriched supplement (ProSure®) was given to the EPA-on group in addition to their standard diet. This supplement included 600 kcal with 2.2 g/day of EPA. Among the 126 patients who were randomized, 123 patients (EPA-off group, n=60; EPA-on group, n=63) were examined in the survival analyses. All background factors were well balanced between the two groups. The 3-year and 5-year overall survival rates were 74.6 and 67.8%, respectively, in the EPA-off group, and 77.8 and 76.2% in the EPA-on group. There was no significant difference between the EPA-off and EPA-on groups (hazard ratio, 0.77; P=0.424). In the subgroup analysis, the hazard ratio was 0.39 in patients who received neoadjuvant chemotherapy and 0.57 in patients with nodal metastasis. In conclusion, a clear survival benefit of perioperative EPA was not observed in localized gastric cancer. The value of EPA should be further tested in a future study in patients with unfavorable advanced gastric cancer. Clinical trial number: UMIN000006380; date of registration, September 21, 2011.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Satoshi Ida
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Haruhiko Cho
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Kentaro Sakamaki
- Department of Biostatistics and Epidemiology, Yokohama City University Medical Center, Yokohama, Kanagawa 232‑0024, Japan
| | - Yuichi Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Aichi 464‑8681, Japan
| | - Kazumasa Fujitani
- Department of Surgery, Osaka General Medical Center, Osaka 558‑8558, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260‑8781, Japan
| | - Yoshiyuki Kawashima
- Department of Gastroenterological Surgery, Saitama Cancer Center, Kitaadachi, Saitama 362‑0806, Japan
| | | | - Soya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Naoki Hiki
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| |
Collapse
|
17
|
Vaseghi G, Pourhadi M, Ghasemi A, Abediny R, Haghjooy Javanmard S. The inhibitory effects of vanillin on the growth of melanoma by reducing nuclear factor-κB activation. Adv Biomed Res 2022; 11:68. [DOI: 10.4103/abr.abr_280_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 10/12/2021] [Indexed: 11/04/2022] Open
|
18
|
Atabilen B, Akbulut G, Bacanli M, Uncu D. Is the nuclear factor kappa-b (NF-κB) pathway and inflammatory status associated with colorectal cancer? Saudi J Gastroenterol 2021; 28:60-66. [PMID: 34380870 PMCID: PMC8919927 DOI: 10.4103/sjg.sjg_44_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Although genetic predisposition has a role in the etiology of colorectal cancer, there are many other factors that affect its development. In this study, it was aimed to evaluate the NF-κB pathway, inflammatory status and dietary antioxidant capacity in individuals with colorectal cancer. METHODS The study was carried out with 40 male subjects diagnosed with colorectal cancer aged between 39-65, years and a control group of the same number of healthy men. Subjects in the case and control groups were subdivided according to body mass index (BMI), as normal (BMI 20-24.9 kg/m2) or overweight/obese (BMI ≥25 kg/m2). RESULTS At the end of the study, NF-κB and interleukin-22 levels were higher in the case group, but no significant difference was found between the groups. Interleukin-23 and 8-Hydroxy-2-deoxyguanosine levels in the case group classified as overweight/obese according to BMI were significantly higher than in the control group (P = 0.001 and P < 0.001, respectively). Considering diet antioxidant capacity, it was higher in individuals in the control group than in the case group. However, there was no significant difference between the groups. CONCLUSION Inflammatory status and reduced dietary antioxidant capacity are risk factors in the development of colorectal cancer.
Collapse
Affiliation(s)
- Büşra Atabilen
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Turkey,Address for correspondence: Prof. Büşra Atabilen, Emek Neighborhood Bişkek Street, 6th Street (Former 81st Street), Gazi University Faculty of Health Sciences, Department of Nutrition and Dietetics, Ankara, Turkey. E-mail:
| | - Gamze Akbulut
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Turkey
| | - Merve Bacanli
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, University of Health Sciences Gülhane, Ankara, Turkey
| | - Doğan Uncu
- Department of Medical Oncology, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
19
|
Purawarga Matada GS, Dhiwar PS, Abbas N, Singh E, Ghara A, Das A, Bhargava SV. Molecular docking and molecular dynamic studies: screening of phytochemicals against EGFR, HER2, estrogen and NF-KB receptors for their potential use in breast cancer. J Biomol Struct Dyn 2021; 40:6183-6192. [PMID: 33525984 DOI: 10.1080/07391102.2021.1877823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Breast cancer (BC) is a second common malignancy in female globally. Hence, identification of novel therapeutic agents is extremely important. Molecular docking and MD simulation are the important tools in the process of drug discovery for searching the potential hits. The structure-based drug designing technique also reveals the information about ligands behavior in computational environment. Docking tools help in visualization and analysis of protein-ligand complex at atomic level. Molecular dynamics shows the stability of the molecules in the receptor cavity in the simulated environment. In this research work, we have screened potent phytochemicals against the BC. We docked the phytochemicals and examined the binding affinities of ligands towards the EGFR, HER2, estrogen and NF-κB receptors. Pristimerin, ixocarpalactone A, viscosalactone B and zhankuic acid A have shown higher binding affinities and energies towards targeted receptors among the screened phytochemicals. MD simulation study shows stability of docked complex for pristimerin and HER2 receptor. These phytochemicals can be repurposed for their anticancer activity. This in-silico work provides a strong ground for further investigation of their anticancer activity.
Collapse
Affiliation(s)
- Gurubasavaraj Swamy Purawarga Matada
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Prasad Sanjay Dhiwar
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Nahid Abbas
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Ekta Singh
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Abhishek Ghara
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Arka Das
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Rajiv Gandhi University of Health & Science Bengaluru, Karnataka, India
| | - Sapna Vyas Bhargava
- Department of Zoology, Maa Bharti PG Science College, University of Kota, Rajasthan, India
| |
Collapse
|
20
|
Yang Z, Zhang Q, Yu L, Zhu J, Cao Y, Gao X. The signaling pathways and targets of traditional Chinese medicine and natural medicine in triple-negative breast cancer. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113249. [PMID: 32810619 DOI: 10.1016/j.jep.2020.113249] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Triple-negative breast cancer (TNBC) has a poorer prognosis than other subtypes due to its strong invasion and higher risk of distant metastasis. Traditional Chinese medicine (TCM) and natural medicine have the unique advantages of multitargets and small side-effects and may be used as long-term complementary and alternative therapies. AIM OF THE REVIEW The present article summarizes the classical signaling pathways and potential targets by the action of TCM and natural medicine (including extracts, active constituents and formulas) on TNBC and provides evidence for its clinical efficacy. METHODS The literature information was acquired from the scientific databases PubMed, Web of Science and CNKI from January 2010 to June 2020, and it was designed to elucidate the internal mechanism and role of TCM and natural medicine in the treatment of TNBC. The search key words included "Triple negative breast cancer" or "triple negative breast carcinoma", "TNBC" and "traditional Chinese medicine" or "Chinese herbal medicine", "medicinal plant", "natural plant", and "herb". RESULTS We described the antitumor activity of TCM and natural medicine in TNBC based on different signaling pathways. Plant medicine and herbal formulas regulated the related gene and protein expression via pathways such as PI3K/AKT/mTOR, MAPK and Wnt/β-catenin, which inhibit the growth, proliferation, migration, invasion and metastasis of TNBC cells. CONCLUSION The inhibitory effect of TCM and natural medicine on tumors was reflected in multiple levels and multiple pathways, providing reasonable evidence for new drug development. To make TCM and natural medicine widely and flexibly used in clinical practice, the efficacy, safety and mechanism of action need more in-depth experimental research.
Collapse
Affiliation(s)
- Zimei Yang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310053, China.
| | - Qiuhua Zhang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310053, China.
| | - Linghong Yu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310053, China.
| | - Jiayan Zhu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310053, China.
| | - Yi Cao
- The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, NO. 54 Youdian Road, Hangzhou, Zhejiang, 310006, China.
| | - Xiufei Gao
- The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, NO. 54 Youdian Road, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
21
|
Du X, Wang S, Liu X, He T, Lin X, Wu S, Wang D, Li J, Huang W, Yang H. MiR-1307-5p targeting TRAF3 upregulates the MAPK/NF-κB pathway and promotes lung adenocarcinoma proliferation. Cancer Cell Int 2020; 20:502. [PMID: 33061854 PMCID: PMC7552495 DOI: 10.1186/s12935-020-01595-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) includes lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). MicroRNA (miRNA) plays an important role in the regulation of post-transcriptional gene expression in animals and plants, especially in lung adenocarcinoma. METHODS MiR-1307-5p is an miRNA with significant differences screened by the second generation of high-throughput sequencing in the early stage of our research group. In the current study, a series of in vitro and in vivo experiments were carried out. MiR-1307-5p mimic, miR-1307-5p inhibitor, and NC were transfected into A549 and H1299 lung adenocarcinoma cells. The correlation between miR-1307-5p and clinicopathological features in pathological samples was analyzed using a lung adenocarcinoma tissue microarray, and miR-1307-5p expression was detected by qPCR. CCK-8, EdU, colony formation, scratch test, and Transwell assays were used to observe cell proliferation and migration. Double luciferase assay, western blot, qPCR, and immunohistochemistry were employed in confirming the target relationship between miR-1307-5p and TRAF3. Western blotting was used to analyze the relationship between miR-1307-5p and the NF-κB/MAPK pathway. Finally, the effect of miR-1307-5p on tumor growth was studied using a subcutaneous tumorigenesis model in nude mice. RESULTS Increased miR-1307-5p expression was significantly related to decreased overall survival rate of lung adenocarcinoma patients, revealing miR-1307-5p as a potential oncogene in lung adenocarcinoma. MiR-1307-5p mimic significantly promoted while miR-1307-5p inhibitor reduced the growth and proliferation of A549 and H1299 cells. MiR-1307-5p overexpression significantly enhanced the migration ability while miR-1307-5p inhibition reduced the migration ability of A549 and H1299 cells. Target binding of miR-1307-5p to TRAF3 was confirmed by double luciferase assay, western blot, qPCR, and immunohistochemistry. miR-1307-5p caused degradation of TRAF3 mRNA and protein. MiR-1307-5p targeted TRAF3 and activated the NF-κB/MAPK pathway. TRAF3 colocalized with p65 and the localization of TRAF3 and p65 changed in each treatment group. Tumor volume of the lv-miR-1307-5p group was significantly larger than that of the lv-NC group, and that of the lv-miR-1307-5p-inhibitor group was significantly smaller than that of the lv-NC group. CONCLUSION In conclusion, miR-1307-5p targets TRAF3 and activates the NF-κB/MAPK pathway to promote proliferation in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xinyue Du
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Shuangmiao Wang
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xingyan Liu
- Dongguan Scientific Research Center, Guangdong Medical University, Zhanjiang, China
| | - Tao He
- Department of Biology, School of Basic Medical Sciences of Guangdong Medical University, Guangzhou, China
| | - Xiangui Lin
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Simin Wu
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Dan Wang
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jiao Li
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Wenhua Huang
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Huiling Yang
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and Mucin1 in breast cancer vaccination: On the verge of a breakthrough. Life Sci 2020; 258:118128. [PMID: 32710947 DOI: 10.1016/j.lfs.2020.118128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/22/2023]
Abstract
Although breast cancer is one of the leading troublesome cancers, the available therapeutic options have not fulfilled the desired outcomes. Immune-based therapy has gained special attention for breast cancer treatment. Although this approach is highly tolerable, its low response rate has rendered it as an undesirable approach. This review aims to describe the essential oncogenic pathways involved in breast cancer, elucidate the immunosuppression and oncogenic effect of Mucin1, and introduce myeloid-derived suppressor cells, which are the main culprits of anti-tumoral immune response attenuation. The various auto-inductive loops between Mucin1 and myeloid-derived suppressor cells are focal in the suppression of anti-tumoral immune responses in patients with breast cancer. These cross-talks between the Mucin1 and myeloid-derived suppressor cells can be the underlying causes of immunotherapy's impotence for patients with breast cancer. This approach can pave the road for the development of a potent vaccine for patients with breast cancer and is translated into clinical settings.
Collapse
Affiliation(s)
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Molecular mechanism of gossypol mediating CCL2 and IL‑8 attenuation in triple‑negative breast cancer cells. Mol Med Rep 2020; 22:1213-1226. [PMID: 32627003 PMCID: PMC7339712 DOI: 10.3892/mmr.2020.11240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/28/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation associated with cancer is characterized by the production of different types of chemokines and cytokines. In cancer, numerous signaling pathways upregulate the expression levels of several cytokines and evolve cells to the neoplastic state. Therefore, targeting these signaling pathways through the inhibition of distinctive gene expression is a primary target for cancer therapy. The present study investigated the anticancer effects of the natural polyphenol gossypol (GOSS) in triple-negative breast cancer (TNBC) cells, the most aggressive breast cancer type with poor prognosis. GOSS effects were examined in two TNBC cell lines: MDA-MB-231 (MM-231) and MDA-MB-468 (MM-468), representing Caucasian Americans (CA) and African Americans (AA), respectively. The obtained IC50s revealed no significant difference between the two cell lines' response to the compound. However, the use of microarray assays for cytokine determination indicated the ability of GOSS to attenuate the expression levels of cancer-related cytokines in the two cell lines. Although GOSS did not alter CCL2 expression in MM-468 cells, it was able to cause 30% inhibition in TNF-α-stimulated MM-231 cells. Additionally, IL-8 was not altered by GOSS treatment in MM-231 cells, while its expression was inhibited by 60% in TNF-α-activated MM-468 cells. ELISA assays supported the microarray data and indicated that CCL2 expression was inhibited by 40% in MM-231 cells, and IL-8 expression was inhibited by 50% in MM-468 cells. Furthermore, in MM-231 cells, GOSS inhibited CCL2 release via the repression of IKBKE, CCL2 and MAPK1 gene expression. Additionally, in MM-468 cells, the compound downregulated the release of IL-8 through repressing IL-8, MAPK1, MAPK3, CCDC88A, STAT3 and PIK3CD gene expression. In conclusion, the data obtained in the present study indicate that the polyphenol compound GOSS may provide a valuable tool in TNBC therapy.
Collapse
|
24
|
Li J, Xun K, Pei K, Liu X, Peng X, Du Y, Qiu L, Tan W. Cell-Membrane-Anchored DNA Nanoplatform for Programming Cellular Interactions. J Am Chem Soc 2019; 141:18013-18020. [PMID: 31626550 DOI: 10.1021/jacs.9b04725] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell-cell interactions are mediated through compositions expressed on the membrane. Engineering the cell surface to display functional modules with high biocompatibility, high controllability, and high stability would offer great opportunities for studying and manipulating these intercellular reactions. However, it remains a technical challenge because of the complex and dynamic nature of the cell membrane. Herein, by using three-dimensional (3D) amphiphilic pyramidal DNA as the scaffold, we develop a biocompatible, effective, and versatile strategy for engineering the cell surface with DNA probes. Compared with linear DNA constructs, these pyramidal probes show higher (nearly 100-fold) membrane-anchoring stability and higher (about 2.5-fold) target accessibility. They enable specific, effective, and tunable connections between cells. Meanwhile, our results indicate that connecting cells in close proximity are critical to initiate intercellular communication. By combining high programmability and high diversity of DNA probes, this strategy is expected to provide a powerful and designable membrane-anchored nanoplatform for studying multicellular communication networks.
Collapse
Affiliation(s)
- Jin Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Kanyu Xun
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Ke Pei
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Xiaojing Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Xueyu Peng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Yulin Du
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Life Sciences, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha , Hunan 410082 , China.,Center for Research at Bio/Nano Interface, Department of Chemistry and Department of Physiology and Functional Genomics, University Health Cancer Center, UF Genetics Institute and McKnight Brain Institute , University of Florida , Gainesville , Florida 32611-7200 , United States
| |
Collapse
|
25
|
Xia Y, Jia C, Xue Q, Jiang J, Xie Y, Wang R, Ran Z, Xu F, Zhang Y, Ye T. Antipsychotic Drug Trifluoperazine Suppresses Colorectal Cancer by Inducing G0/G1 Arrest and Apoptosis. Front Pharmacol 2019; 10:1029. [PMID: 31572198 PMCID: PMC6753363 DOI: 10.3389/fphar.2019.01029] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/12/2019] [Indexed: 02/05/2023] Open
Abstract
Repurposing existing drugs for cancer treatment is an effective strategy. An approved antipsychotic drug, trifluoperazine (TFP), has been reported to have potential anticancer effects against several cancer types. Here, we investigated the effect and molecular mechanism of TFP in colorectal cancer (CRC). In vitro studies showed that TFP induced G0/G1 cell cycle arrest to dramatically inhibit CRC cell proliferation through downregulating cyclin-dependent kinase (CDK) 2, CDK4, cyclin D1, and cyclin E and upregulating p27. TFP also induced apoptosis, decreased mitochondrial membrane potential, and increased reactive oxygen species levels in CRC cells, indicating that TFP induced mitochondria-mediated intrinsic apoptosis. Importantly, TFP significantly suppressed tumor growth in two CRC subcutaneous tumor models without side effects. Interestingly, TFP treatment increased the expression levels of programmed death-1 ligand 1 (PD-L1) in CRC cells and programmed death-1 (PD-1) in tumor-infiltrating CD4+ and CD8+ T cells, implying that the combination of TFP with an immune checkpoint inhibitor, such as an anti-PD-L1 or anti-PD-1 antibody, might have synergistic anticancer effects. Taken together, our study signifies that TFP is a novel treatment strategy for CRC and indicates the potential for using the combination treatment of TFP and immune checkpoint blockade to increase antitumor efficiency.
Collapse
Affiliation(s)
- Yong Xia
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengsen Jia
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Xue
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jinrui Jiang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yao Xie
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Ranran Wang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhiqiang Ran
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tinghong Ye
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
26
|
Miyake S, Ogo A, Kubota H, Teramoto F, Hirai T. β-Hydroxy-β-methylbutyrate Suppresses NF-ĸB Activation and IL-6 Production in TE-1 Cancer Cells. In Vivo 2019; 33:353-358. [PMID: 30804112 DOI: 10.21873/invivo.11481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM Stress reactions, especially those related to surgery, cause poor convalescence of cancer patients. β-Hydroxyβ-methylbutyrate (HMB) is known to regulate excessive inflammation in the body. The objective of this work was to investigate the capacity of HMB to suppress activation of nuclear factor-kappa B (NF-ĸB) and production of interleukin-6 (IL-6) in a human esophageal squamous cell carcinoma cell line (TE-1). MATERIALS AND METHODS Cell proliferation was measured using the water-soluble tetrazolium-1 method, while tumor necrosis factor alpha (TNFα)-induced IL-6 production was measured using an enzyme-linked immunosorbent assay (ELISA) assay. Nuclear translocation of NF-ĸB was detected by immunofluorescence staining. RESULTS HMB did not affect cell proliferation. However, HMB suppressed the TNFα-induced increase in IL-6 production in TE-1 cells by inhibiting NF-ĸB activation. CONCLUSION HMB did not influence TE-1 cell proliferation, but inhibited activation of NF-ĸB and IL-6 production. This result may be useful for improving excessive stress reactions during and after surgery.
Collapse
Affiliation(s)
- Sachi Miyake
- Department of Doctoral Program in Health Science, Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Ayako Ogo
- Department of Doctoral Program in Health Science, Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Hisako Kubota
- Department of Digestive Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Fusako Teramoto
- Department of Doctoral Program in Health Science, Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Toshihiro Hirai
- Department of Doctoral Program in Health Science, Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| |
Collapse
|
27
|
Tang Z, Ma Q, Wang L, Liu C, Gao H, Yang Z, Liu Z, Zhang H, Ji L, Jiang G. A brief review: some compounds targeting YAP against malignancies. Future Oncol 2019; 15:1535-1543. [PMID: 31066301 DOI: 10.2217/fon-2019-0035] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
YAP, acting as a crucial transcription factor in nucleus, regulates the organ size, tissue homeostasis and tumorigenesis. Dysregulation of Hippo-YAP pathway brings a significant impact on the occurrence and development of various tumor types. Moreover, regulation of YAP/TAZ far exceeds the core kinase of the Hippo pathway, and gradually opens up new therapeutic targets. For the moment, chemotherapy together with radiotherapy act as routine methods to prolong the lives of cancer patients. Seeking more effective anti-neoplastic agents seems to be the urgent problem. This brief review focuses on the research progress of YAP inhibitors as the antineoplastic targets. Small molecule inhibitors or drugs have been discovered including verteporfin, dasatinib, statins, A35, JQ1, norcantharidin, agave, MLN8237, dobutamine and peptide-based YAP inhibitors. We are trying to seek novel therapies from the relationship between known drugs and potential mechanisms.
Collapse
Affiliation(s)
- Zhenxue Tang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Qingxia Ma
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Luyao Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Chaolong Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Hui Gao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Zhihong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Zhantao Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Huimin Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Lixia Ji
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| | - Guohui Jiang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao 266021, PR China
| |
Collapse
|
28
|
Hosseini N, Khoshnazar A, Saidijam M, Azizi Jalilian F, Najafi R, Mahdavinezhad A, Ezati R, Sotanian A, Amini R. Zerumbone Suppresses Human Colorectal Cancer Invasion and Metastasis via Modulation of FAk/PI3k/NFκB-uPA Pathway. Nutr Cancer 2019; 71:159-171. [PMID: 30650987 DOI: 10.1080/01635581.2018.1540719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The current study explored the basic molecular mechanisms of zerumbone (ZER), an herbal compound, in inhibiting the migration and invasion of colorectal cancer (CRC) cells in vitro. Two types of CRC cells, namely HCT-116 and SW48, were treated with various concentrations of ZER (8, 16, and 24 µM) for 24, 48, and 72 h, respectively. In vitro assays were performed to determine alterations in proliferation ability, mRNA expression and protein levels, and migration and invasion potential of CRC cells. An SYBR Green-based quantitative polymerase chain reaction (PCR) was utilized to detect the gene expression of focal adhesion kinase (FAK), nuclear factor (NF)-κB, and urokinase-type plasminogen activator (uPA) followed by the evaluation of the level of proteins by western blotting. Migration and invasion potentials of HCT-116 and SW48 cells treated by ZER were examined using migration and invasion assay kits, respectively. We compared the results of all experiments with control groups, including FAK inhibitor, ZER + FAK inhibitor-treated cells, NF-β inhibitor, ZER + NF-β inhibitor, and untreated cells. The data in the present study suggest that ZER may exert its antimetastatic effects through inhibition of FAk/PI3k/NF-κB-uPA signaling pathway, thereby possibly representing a novel class of FAK inhibitors.
Collapse
Affiliation(s)
- Narges Hosseini
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Amineh Khoshnazar
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Massoud Saidijam
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Farid Azizi Jalilian
- b Microbiology Department, Faculty of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Rezvan Najafi
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Ali Mahdavinezhad
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Razie Ezati
- c Institute of Medical Biotechnology , National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Alireza Sotanian
- d Modeling of Noncommunicable Disease Research Center , School of Public Health, Hamadan University of Medical Sciences , Hamadan , Iran
| | - Razieh Amini
- a Research Center for Molecular Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| |
Collapse
|
29
|
Ferraiuolo M, Pulito C, Finch-Edmondson M, Korita E, Maidecchi A, Donzelli S, Muti P, Serra M, Sudol M, Strano S, Blandino G. Agave negatively regulates YAP and TAZ transcriptionally and post-translationally in osteosarcoma cell lines. Cancer Lett 2018; 433:18-32. [DOI: 10.1016/j.canlet.2018.06.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 06/01/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
|
30
|
Messeha SS, Zarmouh NO, Mendonca P, Alwagdani H, Kolta MG, Soliman KFA. The inhibitory effects of plumbagin on the NF-қB pathway and CCL2 release in racially different triple-negative breast cancer cells. PLoS One 2018; 13:e0201116. [PMID: 30059519 PMCID: PMC6066199 DOI: 10.1371/journal.pone.0201116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022] Open
Abstract
Breast cancer (BC) is the second leading cause of death among women in the US, and its subtype triple-negative BC (TNBC) is the most aggressive BC with poor prognosis. In the current study, we investigated the anticancer effects of the natural product plumbagin (PL) on racially different TNBC cells. The PL effects were examined in two TNBC cell lines: MDA-MB-231 (MM-231) and MDA-MB-468 (MM-468), representing Caucasian Americans and African Americans, respectively. The results obtained indicate that PL inhibited cell viability and cell proliferation and induced apoptosis in both cell lines. Notably, MM-468 cells were 5-fold more sensitive to PL than MM-231 cells were. Testing PL and Taxol® showed the superiority of PL over Taxol® as an antiproliferative agent in MM-468 cells. PL treatment resulted in an approximately 20-fold increase in caspase-3 activity with 3 μM PL in MM-468 cells compared with an approximately 3-fold activity increase in MM-231 cells with 8 μM PL. Moreover, the results indicate a higher sensitivity to PL in MM-468 cells than in MM-231 cells. The results also show that PL downregulated CCL2 cytokine expression in MM-468 cells by 30% compared to a 90% downregulation in MM-231 cells. The ELISA results confirmed the array data (35% vs. 75% downregulation in MM-468 and MM-231 cells, respectively). Moreover, PL significantly downregulated IL-6 and GM-CSF in the MM-231 cells. Indeed, PL repressed many NF-қB-regulated genes involved in the regulation of apoptosis, proliferation, invasion, and metastasis. The compound significantly downregulated the same genes (BIRC3, CCL2, TLR2, and TNF) in both types of cells. However, PL impacted five more genes in MM-231 cells, including BCL2A1, ICAM1, IKBKE, IL1β, and LTA. In conclusion, the data obtained in this study indicate that the quinone compound PL could be a novel cancer treatment for TNBC in African American women.
Collapse
Affiliation(s)
- Samia S. Messeha
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
| | - Najla O. Zarmouh
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
| | - Patricia Mendonca
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
| | - Hayfaa Alwagdani
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
| | - Malak G. Kolta
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
| | - Karam F. A. Soliman
- College of Pharmacy and Pharmaceutical Science, Florida A & M University, Tallahassee, Florida, United States of America
- * E-mail:
| |
Collapse
|
31
|
Li JM, Lee YC, Li CC, Lo HY, Chen FY, Chen YS, Hsiang CY, Ho TY. Vanillin-Ameliorated Development of Azoxymethane/Dextran Sodium Sulfate-Induced Murine Colorectal Cancer: The Involvement of Proteasome/Nuclear Factor-κB/Mitogen-Activated Protein Kinase Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:5563-5573. [PMID: 29790745 DOI: 10.1021/acs.jafc.8b01582] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Vanillin is a natural dietary flavoring widely used in the food industry. Colorectal cancer (CRC) is one of the common malignancies in the world. Chronic intestinal inflammation is a risk factor for the development of CRC. We have previously found that vanillin improves and prevents colitis in mice. Here we evaluated the inhibitory activities of vanillin on a mouse model of colitis-induced CRC. Mice were challenged intraperitoneally with azoxymethane (AOM) and orally with dextran sodium sulfate (DSS). Various dosages of vanillin were orally administered for 13 consecutive weeks. Vanillin alleviated the development of tumors in AOM/DSS-induced mice. The total number of tumors in 100 mg/kg vanillin group was significantly reduced by 57.14 ± 7.67%, compared with sham group. Gene expression analysis showed that vanillin downregulated the expression levels of proteasome genes in colon tissues. Moreover, vanillin at 10 mM significantly suppressed proteasome activities in HCT-116 cells by 41.27 ± 0.41%. Furthermore, vanillin diminished the phosphorylation of mitogen-activated protein kinases (MAPKs) and reduced the number of p65-positive cells, proliferating cells, and granulocytes in colon tissues with statistical significance. In conclusion, our data suggested that vanillin was a bioactive compound that ameliorated the development of AOM/DSS-induced colon cancer in mice. Moreover, the amelioration of vanillin might be associated with the downregulation of proteasome, nuclear factor-κB, and MAPK pathways.
Collapse
Affiliation(s)
- Jung-Miao Li
- Graduate Institute of Chinese Medicine , China Medical University , Taichung 40402 , Taiwan
- Department of Chinese Medicine , Show Chwan Memorial Hospital , Changhua 50008 , Taiwan
| | - Yu-Chen Lee
- Graduate Institute of Acupuncture Science , China Medical University , Taichung 40402 , Taiwan
| | - Chia-Cheng Li
- Graduate Institute of Chinese Medicine , China Medical University , Taichung 40402 , Taiwan
| | - Hsin-Yi Lo
- Graduate Institute of Chinese Medicine , China Medical University , Taichung 40402 , Taiwan
| | - Feng-Yuan Chen
- Graduate Institute of Chinese Medicine , China Medical University , Taichung 40402 , Taiwan
| | - Yi-Siou Chen
- Department of Microbiology , China Medical University , Taichung 40402 , Taiwan
| | - Chien-Yun Hsiang
- Department of Microbiology , China Medical University , Taichung 40402 , Taiwan
| | - Tin-Yun Ho
- Graduate Institute of Chinese Medicine , China Medical University , Taichung 40402 , Taiwan
- Department of Health and Nutrition Biotechnology , Asia University , Taichung 41354 , Taiwan
| |
Collapse
|
32
|
Schlapbach A, Revesz L, Pissot Soldermann C, Zoller T, Régnier CH, Bornancin F, Radimerski T, Blank J, Schuffenhauer A, Renatus M, Erbel P, Melkko S, Heng R, Simic O, Endres R, Wartmann M, Quancard J. N-aryl-piperidine-4-carboxamides as a novel class of potent inhibitors of MALT1 proteolytic activity. Bioorg Med Chem Lett 2018; 28:2153-2158. [PMID: 29759726 DOI: 10.1016/j.bmcl.2018.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/16/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
Abstract
Starting from a weak screening hit, potent and selective inhibitors of the MALT1 protease function were elaborated. Advanced compounds displayed high potency in biochemical and cellular assays. Compounds showed activity in a mechanistic Jurkat T cell activation assay as well as in the B-cell lymphoma line OCI-Ly3, which suggests potential use of MALT1 inhibitors in the treatment of autoimmune diseases as well as B-cell lymphomas with a dysregulated NF-κB pathway. Initially, rat pharmacokinetic properties of this compound series were dominated by very high clearance which could be linked to amide cleavage. Using a rat hepatocyte assay a good in vitro-in vivo correlation could be established which led to the identification of compounds with improved PK properties.
Collapse
Affiliation(s)
- Achim Schlapbach
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland.
| | - Laszlo Revesz
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Thomas Zoller
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | | | - Thomas Radimerski
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jutta Blank
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Martin Renatus
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Paulus Erbel
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Samu Melkko
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Richard Heng
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Oliver Simic
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Ralf Endres
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Markus Wartmann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jean Quancard
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
33
|
The ERβ4 variant induces transformation of the normal breast mammary epithelial cell line MCF-10A; the ERβ variants ERβ2 and ERβ5 increase aggressiveness of TNBC by regulation of hypoxic signaling. Oncotarget 2018; 9:12201-12211. [PMID: 29552303 PMCID: PMC5844739 DOI: 10.18632/oncotarget.24134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/05/2017] [Indexed: 01/03/2023] Open
Abstract
Triple negative breast cancer (TNBC) still remains a challenge to treat in the clinic due to a lack of good targets for treatment. Although TNBC lacks expression of ERα, the expression of ERβ and its variants are detected quite frequently in this cancer type and can represent an avenue for treatment. We show that two of the variants of ERβ, namely ERβ2 and ERβ5, control aggressiveness of TNBC by regulating hypoxic signaling through stabilization of HIF-1α. RNA-seq of patient derived xenografts (PDX) from TNBC shows expression of ERβ2, ERβ4 and ERβ5 variants in more than half of the samples. Furthermore, expression of ERβ4 in the immortalized, normal mammary epithelial cell line MCF-10A that is resistant to tumorsphere formation caused transformation and development of tumorspheres. By contrast, ERβ1, ERβ2 or ERβ5 were unable to support tumorsphere formation. We have previously shown that all variants except ERβ1 stabilize HIF-1α but only ERβ4 appears to have the ability to transform normal mammary epithelial cells, pointing towards a unique property of ERβ4. We propose that ERβ variants may be good diagnostic tools and also serve as novel targets for treatment of breast cancer.
Collapse
|
34
|
Wang X, Hassan W, Jabeen Q, Khan GJ, Iqbal F. Interdependent and independent multidimensional role of tumor microenvironment on hepatocellular carcinoma. Cytokine 2017; 103:150-159. [PMID: 29029799 DOI: 10.1016/j.cyto.2017.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/03/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
Abstract
The novelty of an effective therapeutic targeting for hepatocellular carcinoma (HCC) is based on improved understanding of each component of tumor microenvironment (TME) and its correspondent interactions at biological and molecular levels. In this context, new expansions for the treatment against TME and its communication with HCC are under exploration. Despite of the fact that blockage of growth factor receptors has become a treatment of choice in late phases of HCC in clinical practice, still a precise targeted treatment should address all the components of TME. Targeting one specific element out of cellular (cancer associated fibroblasts, endothelial cells, hepatic stellate cells, Kupffer cells and lymphocytes) or non-cellular (extracellular matrix, growth factors, inflammatory cytokines, proteolytic enzymes) parts of TME may not be a successful remedy for the disease because of well-designed hindrances of each component and their functional alternativeness. Meanwhile there are some elements of TME like epithelial-mesenchymal transition and CAF, which are considerably important and need thorough investigations. Ascertaining the potential role of these elements, and a single or combinational drug therapy targeting these elements of TME simultaneously, may provide the appreciable considerations to eventually improve in clinical practices and may also minimize the chances of reoccurrence of HCC.
Collapse
Affiliation(s)
- Xue Wang
- Jiangnan University, Wuxi Medical School, Wuxi 214122, China; China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Waseem Hassan
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan; Department of Pharmacy, The University of Lahore, Pakistan.
| | - Qaiser Jabeen
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Ghulam Jilany Khan
- China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Furqan Iqbal
- Department of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| |
Collapse
|
35
|
Handayani S, Susidarti RA, Jenie RI, Meiyanto E. Two Active Compounds from Caesalpinia sappan L. in Combination with Cisplatin Synergistically Induce Apoptosis and Cell Cycle Arrest on WiDr Cells. Adv Pharm Bull 2017; 7:375-380. [PMID: 29071219 PMCID: PMC5651058 DOI: 10.15171/apb.2017.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/25/2022] Open
Abstract
Purpose: The aim of this study is to observe the synergistic effect of two active compounds of secang, brazilin and brazilein, combined with cisplatin on WiDr colon cancer cells. Methods: Cytotoxic activities of brazilin (Bi) and brazilein (Be) in single and in combination with cisplatin (Cisp) were examined by MTT assay. Synergistic effect was analyzed by combination index (CI) parameter. Apoptosis and cell cycle profiles were observed by using flow cytometry. Results: The result of MTT assay showed that IC50 value of brazilin and brazilein on WiDr cancer cells were 41 µM and 52 µM respectively. The combination of ½ IC50 of Bi-Cisp reduced cells viability up to 64% and showed synergistic effect with CI value less than 1 (CI = 0.8). The combinations of ½ IC50 of Be-Cisp also reduced cells viability up to 78% and showed synergistic effect (CI=0.65). Combination of Bi-Cisp and Be-Cisp induced apoptosis higher than the single treatments. Further analysis on the cell cycle progression showed that single treatment of ½ IC50 of Be and Bi induced S-phase and G2/M-phase accumulation, while combination of Be-Cisp and Bi-Cisp enhanced S-phase accumulation. Conclusion: Both combination of Bi-Cisp and Be-Cisp showed synergistic effect on WiDr cells through induction of apoptosis and halted the cell cycle progression, thus, WiDr cells growth were significantly reduced.
Collapse
Affiliation(s)
- Sri Handayani
- Research Center for Chemistry, Indonesian Institute of Sciences (LIPI), Serpong, Indonesia.,Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ratna Asmah Susidarti
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Riris Istighfari Jenie
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Edy Meiyanto
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
36
|
Aguiar MAN, Wanderley CWS, Nobre LMS, Alencar MRM, Saldanha MDPS, Souza AM, Wong DVT, Barros PG, Almeida PRC, Lima-Júnior RCP, Ribeiro RA. Interleukin-18 (IL-18) is equally expressed in inflammatory breast cancer and noninflammatory locally advanced breast cancer: A possible association with chemotherapy response. Asia Pac J Clin Oncol 2017; 14:e138-e144. [DOI: 10.1111/ajco.12722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/28/2017] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Lívia Maria Soares Nobre
- Department of Physiology and Pharmacology; Faculty of Medicine; Federal University of Ceará; Brazil
| | | | | | - Alceu Machado Souza
- Department of Pathology and Forensic Medicine; Faculty of Medicine; Federal University of Ceará; Brazil
| | | | | | | | | | - Ronaldo Albuquerque Ribeiro
- Department of Physiology and Pharmacology; Faculty of Medicine; Federal University of Ceará; Brazil
- Cancer Institute of Ceará; Brazil
| |
Collapse
|
37
|
Li C, Zhao Z, Zhou J, Liu Y, Wang H, Zhao X. Relationship between the TERT, TNIP1 and OBFC1 genetic polymorphisms and susceptibility to colorectal cancer in Chinese Han population. Oncotarget 2017; 8:56932-56941. [PMID: 28915643 PMCID: PMC5593614 DOI: 10.18632/oncotarget.18378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common diseases worldwide, and telomere length has been reported correlate with CRC. This study aimed to investigate whether polymorphisms of telomere length related genes are associated with susceptibility to CRC in Chinese Han population. 11 SNPs from TERT, TNIP1 and OBFC1 genes were selected and genotyped, in addition odds ratio (OR) and 95% confidence intervals (CI) were used to evaluate association between the SNPs and CRC risk in 247 patients clinically and 300 controls in a Chinese Han population. Our results showed that minor allele "G" of rs7708392 and minor allele "C" of rs10036748 in TNIP1 gene were significantly associated with an increased the CRC risk in genotype model, dominant model and additive model after Bonferroni's multiple adjusted (P<0.0011). Moreover, the two SNPs rs7708392 and rs10036748 were in strong linkage disequilibrium. We observed that the haplotype "G-C" was more frequent among CRC patients and associated with a 1.58-fold increased CRC risk (95%CI=1.17-2.13, P=0.003). Contrarily, haplotype "C-T" was associated with a 0.63-fold reduced CRC risk (95%CI=0.47-0.86, P=0.003). Additionally, SNPs in this study except rs7708392 and rs10036748 were found a modest connection with CRC risk. In conclusion, our study firstly provides evidence for a novel association between polymorphisms of telomere length related TNIP1 gene and CRC susceptibility in Chinese Han population, and the results need a further identification in a large sample size and other populations.
Collapse
Affiliation(s)
- Chuang Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.,Department of Interventional Therapy, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Zixuan Zhao
- Elite Property Management Ltd, Saskatoon, SK S7H 0S6, Canada
| | - Jun Zhou
- Department of Interventional Therapy, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Ying Liu
- Department of Interventional Therapy, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Hao Wang
- Department of Interventional Therapy, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Xinhan Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
38
|
Anti-Inflammatory Mechanism Involved in Pomegranate-Mediated Prevention of Breast Cancer: the Role of NF-κB and Nrf2 Signaling Pathways. Nutrients 2017; 9:nu9050436. [PMID: 28452959 PMCID: PMC5452166 DOI: 10.3390/nu9050436] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Pomegranate (Punica granatum L.), a nutrient-rich unique fruit, has been used for centuries for the prevention and treatment of various inflammation-driven diseases. Based on our previous study, a characterized pomegranate emulsion (PE) exhibited a striking inhibition of dimethylbenz(a)anthracene (DMBA)-initiated rat mammary tumorigenesis via antiproliferative and apoptosis-inducing mechanisms. The objective of the present work is to investigate the anti-inflammatory mechanism of action of PE during DMBA rat mammary carcinogenesis by evaluating the expression of cyclooxygenase-2 (COX-2), heat shock protein 90 (HSP90), nuclear factor-κB (NF-κB) and nuclear factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2). Mammary tumor samples were harvested from our previous chemopreventive study in which PE (0.2–5.0 g/kg) was found to reduce mammary tumorigenesis in a dose-dependent manner. The expressions of COX-2, HSP90, NF-κB, inhibitory κBα (IκBα) and Nrf2 were detected by immunohistochemical techniques. PE decreased the expression of COX-2 and HSP90, prevented the degradation of IκBα, hindered the translocation of NF-κB from cytosol to nucleus and increased the expression and nuclear translocation of Nrf2 during DMBA-induced mammary tumorigenesis. These findings, together with our previous results, indicate that PE-mediated prevention of DMBA-evoked mammary carcinogenesis may involve anti-inflammatory mechanisms through concurrent but differential regulation of two interrelated molecular pathways, namely NF-κB and Nrf2 signaling.
Collapse
|
39
|
Youssef MR, Attia ZI, El-Baz RA, Roshdy S, Settin A. Genetic polymorphisms of NFκB1-94ins/delATTG and NFκBIA-881A/G genes in Egyptian patients with colorectal cancer. Fam Cancer 2017; 16:517-524. [DOI: 10.1007/s10689-017-9992-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
40
|
Yu H, Aravindan N, Xu J, Natarajan M. Inter- and intra-cellular mechanism of NF-kB-dependent survival advantage and clonal expansion of radio-resistant cancer cells. Cell Signal 2017; 31:105-111. [DOI: 10.1016/j.cellsig.2017.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
|
41
|
Forcados GE, James DB, Sallau AB, Muhammad A, Mabeta P. Oxidative Stress and Carcinogenesis: Potential of Phytochemicals in Breast Cancer Therapy. Nutr Cancer 2017; 69:365-374. [PMID: 28103111 DOI: 10.1080/01635581.2017.1267777] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Breast cancer remains a burden in both developed and developing countries, with higher mortality in developing countries. Attempts to eradicate cancer have not been successful despite the progress made in the development of more novel chemotherapeutic drugs. Reactive-oxygen-species-mediated oxidative stress is known to play a role in breast cancer pathogenesis via genetic and epigenetic modifications, resulting in uncontrolled cell proliferation. Phytochemicals could provide leads for the development of alternative therapeutic agents due to their antioxidant activity, as well as their ability to induce apoptosis in cancer cells. However, most of the studies carried out using in vitro models do not continue with further studies in estrogen-receptor-positive in vivo breast cancer models, or fail to examine the possible biochemical mechanisms of phytochemical-based amelioration. This review examines oxidative-stress-mediated carcinogenesis and the potential of phytochemicals as anticancer agents.
Collapse
Affiliation(s)
- Gilead Ebiegberi Forcados
- a Division of Biochemistry , National Veterinary Research Institute , Vom , Nigeria.,b Department of Biochemistry , Faculty of Science, Ahmadu Bello University , Zaria , Nigeria.,c Department of Anatomy and Physiology , Faculty of Veterinary Sciences, University of Pretoria , Pretoria , South Africa
| | - Dorcas Bolanle James
- b Department of Biochemistry , Faculty of Science, Ahmadu Bello University , Zaria , Nigeria
| | | | - Aliyu Muhammad
- b Department of Biochemistry , Faculty of Science, Ahmadu Bello University , Zaria , Nigeria
| | - Peace Mabeta
- c Department of Anatomy and Physiology , Faculty of Veterinary Sciences, University of Pretoria , Pretoria , South Africa
| |
Collapse
|
42
|
Fu K, Sun X, Wier EM, Hodgson A, Liu Y, Sears CL, Wan F. Sam68/KHDRBS1 is critical for colon tumorigenesis by regulating genotoxic stress-induced NF-κB activation. eLife 2016; 5. [PMID: 27458801 PMCID: PMC4959885 DOI: 10.7554/elife.15018] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/29/2016] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor kappa B (NF-κB)-mediated transcription is an important mediator for cellular responses to DNA damage. Genotoxic agents trigger a 'nuclear-to-cytoplasmic' NF-κB activation signaling pathway; however, the early nuclear signaling cascade linking DNA damage and NF-κB activation is poorly understood. Here we report that Src-associated-substrate-during-mitosis-of-68kDa/KH domain containing, RNA binding, signal transduction associated 1 (Sam68/KHDRBS1) is a key NF-κB regulator in genotoxic stress-initiated signaling pathway. Sam68 deficiency abolishes DNA damage-stimulated polymers of ADP-ribose (PAR) production and the PAR-dependent NF-κB transactivation of anti-apoptotic genes. Sam68 deleted cells are hypersensitive to genotoxicity caused by DNA damaging agents. Upregulated Sam68 coincides with elevated PAR production and NF-κB-mediated anti-apoptotic transcription in human and mouse colon cancer. Knockdown of Sam68 sensitizes human colon cancer cells to genotoxic stress-induced apoptosis and genetic deletion of Sam68 dampens colon tumor burden in mice. Together our data reveal a novel function of Sam68 in the genotoxic stress-initiated nuclear signaling, which is crucial for colon tumorigenesis. DOI:http://dx.doi.org/10.7554/eLife.15018.001 Cells use signaling pathways to detect and respond to harmful conditions by switching on genes that keep the cell healthy. One important pathway is the nuclear factor kappa B (NF-κB) signaling pathway, which is activated by many stimuli. These stimuli may come from infections from outside the cell or may originate inside the cell, as seen for DNA damage caused by irradiation, chemicals or rapid DNA replication in cancer cells. Most of a cell’s DNA is located in the cell nucleus. However, NF-κB proteins are normally located outside the nucleus, in the cell’s cytoplasm. Damage to DNA triggers a signal from the nucleus to the cytoplasm. This signal activates the NF-κB proteins, which move into the nucleus and turn on genes that help the cell to recover from the damage. These genes include those that prevent the cell from self-destructing. In one step of the NF-κB activation process, chain-like molecules called polymers are made from a compound called poly(ADP-ribose), or PAR for short. However, few other details are known about how the damaged DNA in the nucleus signals to the cytoplasm. A protein called Sam68, which is found in the cell nucleus, has been linked to DNA damage signaling. Fu, Sun et al. now present evidence that suggests that if mouse cells lack Sam68, they do not produce PAR polymers in response to DNA damage. In addition, these cells could not trigger the PAR-dependent signaling cascade that is essential for activating NF-κB and for turning on the protective genes. Consequently, cells that lacked Sam68 were extremely sensitive to agents that cause DNA damage, such as chemicals and irradiation. The NF-κB pathway is regulated incorrectly in some cancers, but is also activated by DNA damage caused by cancer treatments. Therefore, Fu, Sun et al. also explored the role of Sam68 in cancer. Reducing the levels of Sam68 made human colon cancer cells more likely to self-destruct when they were exposed to DNA-damaging agents. Furthermore, removing Sam68 from mice that spontaneously grow colon cancer caused their tumors to develop more slowly than mice that retained Sam68 in their cells. Overall, the findings presented by Fu, Sun et al. suggest that Sam68 regulates the signal from the nucleus to the cytoplasm that activates NF-κB proteins in response to DNA damage. Sam68 also appears to be important for helping colon cancer cells grow and survive. Future challenges will be to understand how Sam68 regulates the production of the PAR polymer in this response and to explore whether Sam68 can be targeted for treating cancer. DOI:http://dx.doi.org/10.7554/eLife.15018.002
Collapse
Affiliation(s)
- Kai Fu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States
| | - Xin Sun
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States
| | - Eric M Wier
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States
| | - Andrea Hodgson
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, John Hopkins University, Baltimore, United States
| | - Yue Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States
| | - Cynthia L Sears
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, John Hopkins University, Baltimore, United States.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, United States
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, United States
| |
Collapse
|
43
|
Liu X, Liu Y, Hao J, Zhao X, Lang Y, Fan F, Cai C, Li G, Zhang L, Yu G. In Vivo Anti-Cancer Mechanism of Low-Molecular-Weight Fucosylated Chondroitin Sulfate (LFCS) from Sea Cucumber Cucumaria frondosa. Molecules 2016; 21:molecules21050625. [PMID: 27187337 PMCID: PMC6273849 DOI: 10.3390/molecules21050625] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 02/02/2023] Open
Abstract
The low-molecular-weight fucosylated chondroitin sulfate (LFCS) was prepared from native fucosylated chondroitin sulfate (FCS), which was extracted and isolated from sea cucumber Cucumaria frondosa, and the anti-cancer mechanism of LFCS on mouse Lewis lung carcinoma (LLC) was investigated. The results showed that LFCS remarkably inhibited LLC growth and metastasis in a dose-dependent manner. LFCS induced cell cycle arrest by increasing p53/p21 expression and apoptosis through activation of caspase-3 activity in LLC cells. Meanwhile, LFCS suppressed the expression of vascular endothelial growth factor (VEGF), increased the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and downregulated the matrix metalloproteinases (MMPs) level. Furthermore, LFCS significantly suppressed the activation of ERK1/2/p38 MAPK/NF-κB pathway, which played a prime role in expression of MMPs. All of these data indicate LFCS may be used as anti-cancer drug candidates and deserve further study.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Yong Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Jiejie Hao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Xiaoliang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Yinzhi Lang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Fei Fan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Chao Cai
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Guoyun Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Lijuan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
44
|
Promoter Methylation and mRNA Expression of Response Gene to Complement 32 in Breast Carcinoma. J Cancer Epidemiol 2016; 2016:7680523. [PMID: 27118972 PMCID: PMC4828546 DOI: 10.1155/2016/7680523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/23/2015] [Accepted: 03/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background. Response gene to complement 32 (RGC32), induced by activation of complements, has been characterized as a cell cycle regulator; however, its role in carcinogenesis is still controversial. In the present study we compared RGC32 promoter methylation patterns and mRNA expression in breast cancerous tissues and adjacent normal tissues. Materials and Methods. Sixty-three breast cancer tissues and 63 adjacent nonneoplastic tissues were included in our study. Design. Nested methylation-specific polymerase chain reaction (Nested-MSP) and quantitative PCR (qPCR) were used to determine RGC32 promoter methylation status and its mRNA expression levels, respectively. Results. RGC32 methylation pattern was not different between breast cancerous tissue and adjacent nonneoplastic tissue (OR = 2.30, 95% CI = 0.95–5.54). However, qPCR analysis displayed higher levels of RGC32 mRNA in breast cancerous tissues than in noncancerous tissues (1.073 versus 0.959; P = 0.001), irrespective of the promoter methylation status. The expression levels and promoter methylation of RGC32 were not correlated with any of patients' clinical characteristics (P > 0.05). Conclusion. Our findings confirmed upregulation of RGC32 in breast cancerous tumors, but it was not associated with promoter methylation patterns.
Collapse
|
45
|
Ge Y, Cao X, Wang D, Sun W, Sun H, Han B, Cui J, Liu B. Overexpression of Livin promotes migration and invasion of colorectal cancer cells by induction of epithelial-mesenchymal transition via NF-κB activation. Onco Targets Ther 2016; 9:1011-21. [PMID: 27013894 PMCID: PMC4778785 DOI: 10.2147/ott.s93738] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Livin is a novel member of the inhibitors of apoptosis protein family and has been implicated in the development and progression of colorectal cancer (CRC). However, the underlying mechanisms of Livin in CRC remain not fully understood. In this study, we investigated the effects of Livin expression on the proliferation and metastasis of CRC cells and also addressed its related molecular mechanism to metastasis. The expression of Livin in CRC cells (HCT116, SW480, and HT-29 cell lines) was determined by Western blot analysis. Our results show that the overexpression of Livin significantly promotes the proliferation, migration, and invasion of SW480 cells. Concurrently, the inhibition of Livin reduces the proliferation, migration, and invasion of HCT116 cells. In addition, Livin overexpression promotes the epithelial–mesenchymal transition, as evidenced by a decrease in epithelial E-cadherin expression and an increase in mesenchymal markers, including vimentin, Slug, and Snail. Furthermore, adding the NF-κB inhibitor, BAY 11-7028, or transfecting with small interfering RNA against p65 notably restores the expression level of E-cadherin and attenuates the invasive ability of Livin-overexpressing cells. Taken together, these results indicate that Livin potentiates migration and invasion of CRC cells partially through the induction of epithelial–mesenchymal transition via NF-κB activation. Livin may be a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Yang Ge
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of General Surgery, General Hospital Under the Fushun Mining Affairs Bureau, Fushun, People's Republic of China
| | - Xiankui Cao
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dalu Wang
- The Eighth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wei Sun
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hongli Sun
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Bing Han
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Junpeng Cui
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Baolin Liu
- The Sixth Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
46
|
Negi AK, Renuka, Bhatnagar A, Agnihotri N. Celecoxib and fish oil: a combination strategy for decreased inflammatory mediators in early stages of experimental mammary cancer. Inflammopharmacology 2016; 24:11-22. [PMID: 26749133 DOI: 10.1007/s10787-015-0259-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/23/2015] [Indexed: 12/27/2022]
Abstract
Chronic inflammation has been directly linked to cancer progression. Therefore, current study was designed to understand the mechanism of action of chemo-preventive effect of celecoxib and fish oil on inflammatory mediators in experimental mammary carcinoma. Female Wistar rats were distributed into control and DMBA treated groups and further subdivided based on pretreatment with celecoxib and/or fish oil. Inflammation was measured by assessing expression of NF-κB, COX-2 and cytokines. The results indicated an elevation in expression of NF-κB, COX-2 and cytokines' levels (IFN-γ, IL-4 and IL-10) in DMBA group as compared to controls. On pretreatment with celecoxib and/or fish oil in DMBA treated animals, a significant reduction in expression of NF-κB, COX-2 and cytokines' levels was observed. The decrease was more pronounced with combinatorial regimen than either celecoxib or fish oil alone. To conclude, a combinatorial strategy of celecoxib and fish oil may generate an immune response against the tumor cell by altering cytokine repertoire and decrease the tendency of tumor cells to escape immune surveillance.
Collapse
Affiliation(s)
- Anjana Kumari Negi
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Renuka
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Archana Bhatnagar
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Navneet Agnihotri
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
47
|
Uifălean A, Schneider S, Ionescu C, Lalk M, Iuga CA. Soy Isoflavones and Breast Cancer Cell Lines: Molecular Mechanisms and Future Perspectives. Molecules 2015; 21:E13. [PMID: 26703550 PMCID: PMC6273223 DOI: 10.3390/molecules21010013] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/13/2015] [Accepted: 12/14/2015] [Indexed: 01/29/2023] Open
Abstract
The potential benefit of soy isoflavones in breast cancer chemoprevention, as suggested by epidemiological studies, has aroused the interest of numerous scientists for over twenty years. Although intensive work has been done in this field, the preclinical results continue to be controversial and the molecular mechanisms are far from being fully understood. The antiproliferative effect of soy isoflavones has been commonly linked to the estrogen receptor interaction, but there is growing evidence that other pathways are influenced as well. Among these, the regulation of apoptosis, cell proliferation and survival, inhibition of angiogenesis and metastasis or antioxidant properties have been recently explored using various isoflavone doses and various breast cancer cells. In this review, we offer a comprehensive perspective on the molecular mechanisms of isoflavones observed in in vitro studies, emphasizing each time the dose-effect relationship and estrogen receptor status of the cells. Furthermore, we present future research directions in this field which could provide a better understanding of the inner molecular mechanisms of soy isoflavones in breast cancer.
Collapse
Affiliation(s)
- Alina Uifălean
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, Louis Pasteur Street 6, Cluj-Napoca 400349, Romania.
- Institute of Biochemistry, Ernst-Moritz-Arndt-University, Felix-Hausdorff Street 4, Greifswald 17487, Germany.
| | - Stefanie Schneider
- Institute of Biochemistry, Ernst-Moritz-Arndt-University, Felix-Hausdorff Street 4, Greifswald 17487, Germany.
| | - Corina Ionescu
- Department of Pharmaceutical Biochemistry and Clinical Laboratory, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, Louis Pasteur Street 6, Cluj-Napoca 400349, Romania.
| | - Michael Lalk
- Institute of Biochemistry, Ernst-Moritz-Arndt-University, Felix-Hausdorff Street 4, Greifswald 17487, Germany.
| | - Cristina Adela Iuga
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, Louis Pasteur Street 6, Cluj-Napoca 400349, Romania.
| |
Collapse
|
48
|
Lv Z, Fan J, Zhang X, Huang Q, Han J, Wu F, Hu G, Guo M, Jin Y. Integrative genomic analysis of interleukin-36RN and its prognostic value in cancer. Mol Med Rep 2015; 13:1404-12. [PMID: 26676204 DOI: 10.3892/mmr.2015.4667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/28/2015] [Indexed: 11/05/2022] Open
Abstract
Interleukin (IL)-36RN, previously known as IL1-F5 and IL-1δ, shares a 360-kb region of chromosome 2q13 with members of IL-1 systems. IL-36RN encodes an anti-inflammatory cytokine, IL-36 receptor antagonist (IL-36Ra). In spite of IL-36Ra showing the highest homology to IL-1 receptor (IL-1R) antagonist, it differs from the latter in aspects including its binding to IL-lRrp2 but not to IL-1R1. IL-36RN is mainly expressed in epithelial cells and has important roles in inflammatory diseases. In the present study, IL-36RN was identified in the genomes of 27 species, including human, chimpanzee, mouse, horse and dolphin. Human IL-36RN was mainly expressed in the eye, head and neck, fetal heart, lung, testis, cervix and placenta; furthermore, it was highly expressed in bladder and parathyroid tumors. Furthermore, a total of 30 single nucleotide polymorphisms causing missense mutations were determined, which are considered to be the causes of various diseases, such as generalized pustular psoriasis. In addition, the link between IL-36RN and the prognosis of certain cancer types was revealed through meta-analysis. Tumor-associated transcriptional factors c-Fos, activator protein-1, c-Jun and nuclear factor κB were found to bind to the upstream region in the IL-36RN gene. This may indicate that IL-36RN is involved in tumorigenesis and tumor progression through the regulation of tumor-associated transcriptional factors. The present study identified IL-36RN in various species and investigated the associations between IL-36RN and cancer prognosis, which would determine whether IL-36RN drove the evolution of the various species with regard to tumorigenesis.
Collapse
Affiliation(s)
- Zhilei Lv
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jinshuo Fan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiuxiu Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qi Huang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jieli Han
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guorong Hu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of the Ministry of Health, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
49
|
Jamshidi M, Fagerholm R, Khan S, Aittomäki K, Czene K, Darabi H, Li J, Andrulis IL, Chang-Claude J, Devilee P, Fasching PA, Michailidou K, Bolla MK, Dennis J, Wang Q, Guo Q, Rhenius V, Cornelissen S, Rudolph A, Knight JA, Loehberg CR, Burwinkel B, Marme F, Hopper JL, Southey MC, Bojesen SE, Flyger H, Brenner H, Holleczek B, Margolin S, Mannermaa A, Kosma VM, Dyck LV, Nevelsteen I, Couch FJ, Olson JE, Giles GG, McLean C, Haiman CA, Henderson BE, Winqvist R, Pylkäs K, Tollenaar RA, García-Closas M, Figueroa J, Hooning MJ, Martens JW, Cox A, Cross SS, Simard J, Dunning AM, Easton DF, Pharoah PD, Hall P, Blomqvist C, Schmidt MK, Nevanlinna H. SNP-SNP interaction analysis of NF-κB signaling pathway on breast cancer survival. Oncotarget 2015; 6:37979-94. [PMID: 26317411 PMCID: PMC4741978 DOI: 10.18632/oncotarget.4991] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/16/2015] [Indexed: 12/03/2022] Open
Abstract
In breast cancer, constitutive activation of NF-κB has been reported, however, the impact of genetic variation of the pathway on patient prognosis has been little studied. Furthermore, a combination of genetic variants, rather than single polymorphisms, may affect disease prognosis. Here, in an extensive dataset (n = 30,431) from the Breast Cancer Association Consortium, we investigated the association of 917 SNPs in 75 genes in the NF-κB pathway with breast cancer prognosis. We explored SNP-SNP interactions on survival using the likelihood-ratio test comparing multivariate Cox' regression models of SNP pairs without and with an interaction term. We found two interacting pairs associating with prognosis: patients simultaneously homozygous for the rare alleles of rs5996080 and rs7973914 had worse survival (HRinteraction 6.98, 95% CI=3.3-14.4, P=1.42E-07), and patients carrying at least one rare allele for rs17243893 and rs57890595 had better survival (HRinteraction 0.51, 95% CI=0.3-0.6, P = 2.19E-05). Based on in silico functional analyses and literature, we speculate that the rs5996080 and rs7973914 loci may affect the BAFFR and TNFR1/TNFR3 receptors and breast cancer survival, possibly by disturbing both the canonical and non-canonical NF-κB pathways or their dynamics, whereas, rs17243893-rs57890595 interaction on survival may be mediated through TRAF2-TRAIL-R4 interplay. These results warrant further validation and functional analyses.
Collapse
Affiliation(s)
- Maral Jamshidi
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00029 HUS, Finland
| | - Rainer Fagerholm
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00029 HUS, Finland
| | - Sofia Khan
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00029 HUS, Finland
| | - Kristiina Aittomäki
- Department of Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00029 HUS, Finland
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Hatef Darabi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Jingmei Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Irene L. Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jenny Chang-Claude
- Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Devilee
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Kyriaki Michailidou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Manjeet K. Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Qi Guo
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Valerie Rhenius
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Sten Cornelissen
- Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia A. Knight
- Prosserman Centre for Health Research, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Christian R. Loehberg
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Barbara Burwinkel
- Molecular Epidemiology Group, German Cancer Research Center, Heidelberg, Germany
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Frederik Marme
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - John L. Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Melissa C. Southey
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stig E. Bojesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Henrik Flyger
- Department of Breast Surgery, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sara Margolin
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arto Mannermaa
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Veli-Matti Kosma
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | | | - Laurien Van Dyck
- Vesalius Research Center (VRC), VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Ines Nevelsteen
- Multidisciplinary Breast Center, Medical Oncology, University Hospital Leuven, Leuven, Belgium
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Janet E. Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global health, The University of Melbourne, Melbourne, Australia
| | - Catriona McLean
- Anatomical Pathology, The Alfred Hospital, Melbourne, Australia
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Cancer Research and Translational Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre NordLab, Oulu, Finland
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Cancer Research and Translational Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre NordLab, Oulu, Finland
| | - Rob A.E.M. Tollenaar
- Department of Surgical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, SM2 5NG, UK
- Breakthrough Breast Cancer Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jonine Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, AE Rotterdam, The Netherlands
| | - John W.M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, AE Rotterdam, The Netherlands
| | - Angela Cox
- Sheffield Cancer Research, Department of Oncology, University of Sheffield, Sheffield, UK
| | - Simon S. Cross
- Academic Unit of Pathology, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Jacques Simard
- Centre Hospitalier Universitaire de Québec Research Center, Laval University, Québec City, Canada
| | - Alison M. Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Paul D.P. Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Carl Blomqvist
- Department of Oncology, University of Helsinki and Helsinki University Central Hospital, Helsinki, HUS, Finland
| | - Marjanka K. Schmidt
- Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00029 HUS, Finland
| |
Collapse
|
50
|
Nagaraju GP, Madanraj AS, Aliya S, Rajitha B, Alese OB, Kariali E, Alam A, El-Rayes BF. MicroRNAs as biomarkers and prospective therapeutic targets in colon and pancreatic cancers. Tumour Biol 2015; 37:97-104. [PMID: 26537581 DOI: 10.1007/s13277-015-4346-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/28/2015] [Indexed: 12/15/2022] Open
Abstract
Colon and pancreatic cancers have high mortality rates due to early metastasis prior to the onset of symptoms. Screening tests for colorectal cancer are invasive and expensive. No effective screening is available for pancreatic cancer. Identification of biomarkers for early detection in both of these cancers is being extensively researched. MicroRNAs (miRNA) are small non-coding molecule biomarkers that regulate cancers. Measurement of miRNAs in pancreatic fluid or blood could be a preferred non-invasive screening method. The regulation of colon and pancreatic cancers by miRNA is complex. miRNA play a central role in inflammation, invasiveness, and tumor progression in these two cancers, as well as regulation of the NF-κB pathway. miRNA's evolving role in screening is also reviewed.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 1365 Clifton RD NE, Office 3025, Atlanta, GA, 30322, USA.
| | - Appiya Santharam Madanraj
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK, 12 LE1 9HN
| | - Sheik Aliya
- Department of Biotechnology, Jawaharlal Nehru Technological University, Hyderabad, Andhra Pradesh, 500085, India
| | - Balney Rajitha
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 1365 Clifton RD NE, Office 3025, Atlanta, GA, 30322, USA.,Department of Microbiology, Banasthali University, Banasthali, Rajasthan, 304022, India
| | - Olatunji Boladale Alese
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 1365 Clifton RD NE, Office 3025, Atlanta, GA, 30322, USA
| | - Ekamber Kariali
- School of Life Sciences, Department of Biotechnology, Sambalpur University, Jyoti Vihar, Sambalpur, Odisha, 768019, India
| | - Afroz Alam
- Department of Microbiology, Banasthali University, Banasthali, Rajasthan, 304022, India
| | - Bassel F El-Rayes
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 1365 Clifton RD NE, Office 3025, Atlanta, GA, 30322, USA
| |
Collapse
|