1
|
Saleh N, Mahmoud HE, Eltaher H, Helmy M, El-Khordagui L, Hussein AA. Prodigiosin-Functionalized Probiotic Ghosts as a Bioinspired Combination Against Colorectal Cancer Cells. Probiotics Antimicrob Proteins 2023; 15:1271-1286. [PMID: 36030493 PMCID: PMC10491537 DOI: 10.1007/s12602-022-09980-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 12/02/2022]
Abstract
Lactobacillus acidophilus ghosts (LAGs) with the unique safety of a probiotic, inherent tropism for colon cells, and multiple bioactivities offer promise as drug carriers for colon targeting. Our objective was to evaluate LAGs functionalized with prodigiosin (PG), apoptotic secondary bacterial metabolite, as a bioinspired formulation against colorectal cancer (CRC). LAGs were prepared by a chemical method and highly purified by density gradient centrifugation. LAGs were characterized by microscopic and staining techniques as relatively small-sized uniform vesicles (≈1.6 µm), nearly devoid of cytoplasmic and genetic materials and having a negatively charged intact envelope. PG was highly bound to LAGs envelope, generating a physiologically stable bioactive entity (PG-LAGs), as verified by multiple microscopic techniques and lack of PG release under physiological conditions. PG-LAGs were active against HCT116 CRC cells at both the cellular and molecular levels. Cell viability data highlighted the cytotoxicity of PG and LAGs and LAGs-induced enhancement of PG selectivity for HCT116 cells, anticipating dose reduction for PG and LAGs. Molecularly, expression of the apoptotic caspase 3 and P53 biomarkers in HCT116 intracellular proteins was significantly upregulated while that of the anti-apoptotic Bcl-2 (B-cell lymphoma 2) was downregulated by PG-LAGs relative to PG and 5-fluorouracil. PG-LAGs provide a novel bacteria-based combination for anticancer biomedicine.
Collapse
Affiliation(s)
- Nessrin Saleh
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Hoda E Mahmoud
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Hoda Eltaher
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
- Regenerative Medicine and Cellular Therapies Division, Faculty of Science, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Maged Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Labiba El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Ahmed A Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Abstract
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
Collapse
|
3
|
Emerging applications of bacteria as antitumor agents. Semin Cancer Biol 2021; 86:1014-1025. [PMID: 33989734 DOI: 10.1016/j.semcancer.2021.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Bacteria are associated with the human body and colonize the gut, skin, and mucous membranes. These associations can be either symbiotic or pathogenic. In either case, bacteria derive more benefit from their host. The ability of bacteria to enter and survive within the human body can be exploited for human benefit. They can be used as a vehicle for delivering or producing bioactive molecules, such as toxins and lytic enzymes, and eventually for killing tumor cells. Clostridium and Salmonella have been shown to infect and survive within the human body, including in tumors. There is a need to develop genetic circuits, which enable bacterial cells to carry out the following activities: (i) escape the human immune system, (ii) invade tumors, (iii) multiply within the tumorous cells, (iv) produce toxins via quorum sensing at low cell densities, and (v) express suicide genes to undergo cell death or cell lysis after the tumor has been lysed. Thus, bacteria have the potential to be exploited as anticancer agents.
Collapse
|
4
|
Angius A, Scanu AM, Arru C, Muroni MR, Rallo V, Deiana G, Ninniri MC, Carru C, Porcu A, Pira G, Uva P, Cossu-Rocca P, De Miglio MR. Portrait of Cancer Stem Cells on Colorectal Cancer: Molecular Biomarkers, Signaling Pathways and miRNAome. Int J Mol Sci 2021; 22:1603. [PMID: 33562604 PMCID: PMC7915330 DOI: 10.3390/ijms22041603] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death worldwide, and about 20% is metastatic at diagnosis and untreatable. Increasing evidence suggests that the heterogeneous nature of CRC is related to colorectal cancer stem cells (CCSCs), a small cells population with stemness behaviors and responsible for tumor progression, recurrence, and therapy resistance. Growing knowledge of stem cells (SCs) biology has rapidly improved uncovering the molecular mechanisms and possible crosstalk/feedback loops between signaling pathways that directly influence intestinal homeostasis and tumorigenesis. The generation of CCSCs is probably connected to genetic changes in members of signaling pathways, which control self-renewal and pluripotency in SCs and then establish function and phenotype of CCSCs. Particularly, various deregulated CCSC-related miRNAs have been reported to modulate stemness features, controlling CCSCs functions such as regulation of cell cycle genes expression, epithelial-mesenchymal transition, metastasization, and drug-resistance mechanisms. Primarily, CCSC-related miRNAs work by regulating mainly signal pathways known to be involved in CCSCs biology. This review intends to summarize the epigenetic findings linked to miRNAome in the maintenance and regulation of CCSCs, including their relationships with different signaling pathways, which should help to identify specific diagnostic, prognostic, and predictive biomarkers for CRC, but also develop innovative CCSCs-targeted therapies.
Collapse
Affiliation(s)
- Andrea Angius
- Institute of Genetic and Biomedical Research (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| | - Antonio Mario Scanu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| | - Caterina Arru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| | - Vincenzo Rallo
- Institute of Genetic and Biomedical Research (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| | - Giulia Deiana
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| | - Maria Chiara Ninniri
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Alberto Porcu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Paolo Uva
- IRCCS G. Gaslini, 16147 Genoa, Italy;
| | - Paolo Cossu-Rocca
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
- Department of Diagnostic Services, “Giovanni Paolo II” Hospital, ASSL Olbia-ATS Sardegna, 07026 Olbia, Italy
| | - Maria Rosaria De Miglio
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (A.M.S.); (M.R.M.); (G.D.); (M.C.N.); (A.P.); (P.C.-R.)
| |
Collapse
|
5
|
Ambrose AJ, Pham NT, Sivinski J, Guimarães L, Mollasalehi N, Jimenez P, Abad MA, Jeyaprakash AA, Shave S, Costa-Lotufo LV, La Clair JJ, Auer M, Chapman E. A two-step resin based approach to reveal survivin-selective fluorescent probes. RSC Chem Biol 2021; 2:181-186. [PMID: 34458780 PMCID: PMC8342005 DOI: 10.1039/d0cb00122h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/07/2020] [Indexed: 01/24/2023] Open
Abstract
The identification of modulators for proteins without assayable biochemical activity remains a challenge in chemical biology. The presented approach adapts a high-throughput fluorescence binding assay and functional chromatography, two protein-resin technologies, enabling the discovery and isolation of fluorescent natural product probes that target proteins independently of biochemical function. The resulting probes also suggest targetable pockets for lead discovery. Using human survivin as a model, we demonstrate this method with the discovery of members of the prodiginine family as fluorescent probes to the cancer target survivin.
Collapse
Affiliation(s)
- Andrew J Ambrose
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Nhan T Pham
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | - Jared Sivinski
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Larissa Guimarães
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
- Departamento de Farmacologia, Universidade de São Paulo São Paulo SP 05508-900 Brazil
| | - Niloufar Mollasalehi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Paula Jimenez
- Instituto do Mar, Universidade Federal de São Paulo Santos SP 11.070-100 Brazil
| | - Maria A Abad
- Wellcome Centre for Cell Biology, University of Edinburgh Edinburgh EH9 3BF UK
| | | | - Steven Shave
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | | | - James J La Clair
- Xenobe Research Institute P. O. Box 3052 San Diego CA 92163-1052 USA
| | - Manfred Auer
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| |
Collapse
|
6
|
Branco PC, Pontes CA, Rezende-Teixeira P, Amengual-Rigo P, Alves-Fernandes DK, Maria-Engler SS, da Silva AB, Pessoa ODL, Jimenez PC, Mollasalehi N, Chapman E, Guallar V, Machado-Neto JA, Costa-Lotufo LV. Survivin modulation in the antimelanoma activity of prodiginines. Eur J Pharmacol 2020; 888:173465. [PMID: 32814079 DOI: 10.1016/j.ejphar.2020.173465] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 10/23/2022]
Abstract
Melanoma is a type of skin cancer with an elevated incidence of metastasis and chemoresistance. Such features hamper treatment success of these neoplasms, demanding the search for new therapeutic options. Using a two-step resin-based approach, we recently demonstrated that cytotoxic prodiginines bind to the inhibitor of apoptosis protein, survivin. Herein, we explore the role of survivin in melanoma and whether its modulation is related to the antimelanoma properties of three cytotoxic prodiginines (prodigiosin, cyclononylprodigiosin, and nonylprodigiosin) isolated from marine bacteria. In melanoma patients and cell lines, survivin is overexpressed, and higher levels negatively impact survival. All three prodiginines caused a decrease in cell growth with reduced cytotoxicity after 24 h compared to 72 h treatment, suggesting that low concentrations promote cytostatic effects in SK-Mel-19 (BRAF mutant) and SK-Mel-28 (BRAF mutant), but not in SK-Mel-147 (NRAS mutant). An increase in G1 population was observed after 24 h treatment with prodigiosin and cyclononylprodigiosin in SK-Mel-19. Further studies indicate that prodigiosin induced apoptosis and DNA damage, as detected by increased caspase-3 cleavage and histone H2AX phosphorylation, further arguing for the downregulation of survivin. Computer simulations suggest that prodigiosin and cyclononylprodigiosin bind to the BIR domain of survivin. Moreover, knockdown of survivin increased long-term toxicity of prodigiosin, as observed by reduced clonogenic capacity, but did not alter short-term cytotoxicity. In summary, prodiginine treatment provoked cytostatic rather than cytotoxic effects, cell cycle arrest at G0/G1 phase, induction of apoptosis and DNA damage, downregulation of survivin, and decreased clonogenic capacity in survivin knockdown cells.
Collapse
Affiliation(s)
- Paola C Branco
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, Sao Paulo, SP, Brazil
| | - Cristine A Pontes
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, Sao Paulo, SP, Brazil
| | - Paula Rezende-Teixeira
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, Sao Paulo, SP, Brazil
| | - Pep Amengual-Rigo
- Department of Life Sciences, Barcelona Supercomputing Center, 08034, Barcelona, Spain
| | - Débora K Alves-Fernandes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | - Silvya Stuchi Maria-Engler
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | - Alison B da Silva
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, 60021, Fortaleza, CE, Brazil
| | - Otília Deusdênia L Pessoa
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, 60021, Fortaleza, CE, Brazil
| | - Paula C Jimenez
- Institute of Marine Sciences, Institute of Marine Sciences, Federal University of São Paulo, 11.070-100, Santos, SP, Brazil
| | - Niloufar Mollasalehi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 85721-0207, Tucson, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 85721-0207, Tucson, USA
| | - Victor Guallar
- Department of Life Sciences, Barcelona Supercomputing Center, 08034, Barcelona, Spain
| | - João A Machado-Neto
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, Sao Paulo, SP, Brazil
| | - Leticia V Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-900, Sao Paulo, SP, Brazil.
| |
Collapse
|
7
|
Zhou L, Zhang Y, Jin J, Gu X. Correlation between lncRNA SNHG16 gene polymorphism and its interaction with environmental factors and susceptibility to colorectal cancer. Medicine (Baltimore) 2020; 99:e23372. [PMID: 33235108 PMCID: PMC7710222 DOI: 10.1097/md.0000000000023372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To study the relationship between long-chain non-coding RNA small nucleolar RNA host gene 16 (lncRNA SNHG16) polymorphisms and its interaction with environmental factors and susceptibility to colorectal cancer (CRC). METHODS Sanger sequencing was used to analyze genotypes of lncRNA SNHG16 gene rs7353, rs8038, and rs15278 sites. Multifactor dimensionality reduction was used to analyze interactions between lncRNA SNHG16 gene rs7353, rs8038, rs15278 sites, and environmental factors. Haploview 4.1 software was used to analyze linkage disequilibrium of lncRNA SNHG16 gene rs7353, rs8038, and rs15278 sites. Quantitative real-time polymerase chain reaction was used to analyze plasma lncRNA SNHG16 levels of CRC patients and control subjects. RESULTS Variation of the lncRNA SNHG16 gene rs7353 site A>G variation was associated with decreased CRC susceptibility (Odds ratio [OR] = 0.50, 95% confidence interval [CI]: 0.40-0.62, P < .01). The rs8038 site G>A and rs15278 site A>G variation were associated with increased CRC susceptibility (OR = 1.87, 95% CI: 1.47-2.36, P < .01). The rs15278 site G>A variation was associated with increased CRC susceptibility (OR = 2.24, 95% CI: 1.61-3.11, P < .01). Interaction combinations featuring age, rs7353, rs8038, and rs15278 single nucleotide polymorphism are 13.53 times more susceptible to CRC than other interactions (95% CI: 9.43-19.41, P < .01). The rs15278, rs8038, and rs7353 site AGA haplotypes were significantly associated with a decreased CRC risk (OR = 0.65, 95% CI: 0.48-0.88, P = .01), AAG haplotypes were significantly associated with an increased CRC risk (OR = 2.00, 95% CI: 1.27-3.17, P < .01). High lncRNA SNHG16 expression was associated with tumor progression in CRC patients (χ = 8.85, P = .03). The rs7353 site A>G variation caused a significant decrease in plasma lncRNA SNHG16 level (P < .01), while the rs8038 site G>A variation and rs15278 site A>G variation resulted in increased plasma lncRNA SNHG16 levels. CONCLUSION Polymorphisms of lncRNA SNHG16 gene rs7353, rs8038, rs15278 loci and their interaction with age are significantly associated with CRC susceptibility.
Collapse
Affiliation(s)
- Li Zhou
- Department of Medical Oncology, First People's Hospital of Yuhang District, Hangzhou
| | - Yuefeng Zhang
- Department of Hematology, First People's Hospital of Yuhang District, Hangzhou
| | - Jianjiang Jin
- Department of Medical Oncology, First People's Hospital of Yuhang District, Hangzhou
| | - Xuewei Gu
- Department of Gastroenterology, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Shaoxing, Zhejiang Province, China
| |
Collapse
|
8
|
Das PK, Zahan T, Abdur Rakib M, Khanam JA, Pillai S, Islam F. Natural Compounds Targeting Cancer Stem Cells: A Promising Resource for Chemotherapy. Anticancer Agents Med Chem 2020; 19:1796-1808. [PMID: 31272363 DOI: 10.2174/1871520619666190704111714] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/04/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cancer Stem Cells (CSCs) are the subpopulation of cancer cells which are directly involved in drug resistance, metastases to distant organ and cancer recurrence. METHODS A systematic literature search was conducted through various electronic databases including, Pubmed, Scopus, Google scholar using the keywords "cancer stem cells" and "natural compounds" in the present study. Articles published between 1999 and 2019 were reviewed. All the expositions concerning CSCs associated cancer pathogenesis and therapy resistance, as well as targeting these properties of CSCs by natural compounds were selected for the current study. RESULTS Natural compounds have always been thought as a rich source of biologically active principles, which target aberrantly activated signaling pathways and other modalities of CSCs, while tethering painful side effects commonly involved in the first-line and second-line chemo-radiotherapies. In this review, we have described the key signaling pathways activated in CSCs to maintain their survival and highlighted how natural compounds interrupt these signaling pathways to minimize therapy resistance, pathogenesis and cancer recurrence properties of CSCs, thereby providing useful strategies to treat cancer or aid in cancer therapy improvement. Like normal stem cells, CSCs rely on different signaling pathways and other properties for their maintenance. Therefore, the success of cancer treatment depends on the development of proper anti-neoplastic drugs capable of intercepting those signaling pathways as well as other properties of CSCs in order to eradicate this evasive subpopulation of cancer cells. CONCLUSION Compounds of natural origin might act as an outstanding source to design novel therapies against cancer stem cells.
Collapse
Affiliation(s)
- Plabon K Das
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Tasnim Zahan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md Abdur Rakib
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Jahan A Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Suja Pillai
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh.,School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
9
|
Farace C, Pisano A, Griñan-Lison C, Solinas G, Jiménez G, Serra M, Carrillo E, Scognamillo F, Attene F, Montella A, Marchal JA, Madeddu R. Deregulation of cancer-stem-cell-associated miRNAs in tissues and sera of colorectal cancer patients. Oncotarget 2020; 11:116-130. [PMID: 32010426 PMCID: PMC6968784 DOI: 10.18632/oncotarget.27411] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a deadly tumour in Western countries characterized by high cellular/molecular heterogeneity. Cancer stem cells (CSC) act in cancer recurrence, drug-resistance and in metastatic epithelial-to-mesenchymal transition. microRNAs (miRNAs) contribute to cancer is increasing, and miRNA roles in CSC phenotype and fate and their utility as CRC biomarkers have also been reported. Here, we investigated miR-21, miR-221, miR-18a, miR-210, miR-31, miR-34a, miR-10b and miR-16 expression in experimental ALDH+ and CD44+/CD326+ colorectal CSCs obtained from the human CRC cell lines HCT-116, HT-29 and T-84. Then, we moved our analysis in cancer tissue (CT), healthy tissue (HT) and serum (S) of adult CRC patients (n=12), determining relationships with clinical parameters (age, sex, metastasis, biochemical serum markers). Specific miRNA patterns were evident in vitro (normal, monolayers and CSCs) and in patients’ samples stratified by TNM stage (LOW vs HIGH) or metastasis (Met vs no-Met). miR-21, miR-210, miR-34a upregulation ad miR-16 dowregulation associated with the CSCs phenotype. miR-31b robustly overexpressed in monolayers and CSCs, and in CT ad S of HIGH grade and Met patients, suggesting a role as marker of CRC progression and metastasis. miR-18a upregulated in all cancer models and associated to CSC phenotype, and to metastasis and age in patients. miR-10b downregulated in CT and S of LOW/HIGH grade and no-Met patients. Our results identify miRNAs useful as colorectal CSC biomarker and that miR-21, miR-210, miR-10b and miR-31b are promising markers of CRC. A specific role of miR-18a as metastatic CRC serum biomarker in adult patients was also highlighted.
Collapse
Affiliation(s)
- Cristiano Farace
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Institute of Biostructures and Biosystems, Rome, Italy
| | - Andrea Pisano
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Carmen Griñan-Lison
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.Granada), Granada, Spain
| | - Giuliana Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.Granada), Granada, Spain.,Bio-Health Research Foundation of Eastern Andalusia - Alejandro Otero (FIBAO), Granada, Spain
| | - Marina Serra
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Esmeralda Carrillo
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.Granada), Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | | | - Federico Attene
- O.U. of Surgery I (Surgical Pathology), A.O.U. Sassari, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.Granada), Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Roberto Madeddu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Institute of Biostructures and Biosystems, Rome, Italy
| |
Collapse
|
10
|
Small molecule modulators targeting protein kinase CK1 and CK2. Eur J Med Chem 2019; 181:111581. [DOI: 10.1016/j.ejmech.2019.111581] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022]
|
11
|
He Z, Yu L, Luo S, Li Q, Huang S, An Y. RGS4 Regulates Proliferation And Apoptosis Of NSCLC Cells Via microRNA-16 And Brain-Derived Neurotrophic Factor. Onco Targets Ther 2019; 12:8701-8714. [PMID: 31695428 PMCID: PMC6821062 DOI: 10.2147/ott.s221657] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/11/2019] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Regulator of G-protein signaling (RGS) proteins are GTPase-activating proteins that target the α-subunit of heterotrimeric G proteins. Many studies have shown that RGS proteins contribute to tumorigenesis and metastasis. However, the mechanism in which RGS proteins, especially RGS4, affect the development of non-small cell lung cancer (NSCLC) remains unclear. The aim of this study was to characterize the role of RGS4 in NSCLC. METHODS RGS4 expression in NSCLC tissues was assessed using an immunohistochemistry tissue microarray. Additionally, RGS4 was knocked down using short-hairpin RNA to assess the regulatory function of RGS4 in the biological behaviors of human NSCLC cell lines. A xenograft lung cancer model in nude BALB/c mice was established to study whether RGS4 knockdown inhibits cancer cell proliferation in vivo. RESULTS We observed an increase in RGS4 protein levels in NSCLC samples. RGS4 knockdown inhibited cell proliferation and induced apoptosis in H1299 and PC9 cell lines, but did not affect cell migration. Moreover, we found that RGS4 negatively regulated the expression of microRNA-16 (miR-16), a tumor suppressor. The inhibition of miR-16 resulted in upregulated RGS4 expression. We also found that RGS4 regulated the expression of brain-derived neurotrophic factor (BDNF) and activated the BDNF-tropomyosin receptor kinase B signaling pathway. CONCLUSION This study revealed that RGS4 overexpression positively correlated with the development of NSCLC. TDownstream RGS4 targets (eg, miR-16 and BDNF) might be involved in the development of NSCLC and may serve as potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Zheng He
- Biotechnology Department, Beijing Center for Physical and Chemical Analysis, Beijing100094, People’s Republic of China
- Department of Clinical Laboratory, Chinese People’s Liberation Army General Hospital, Beijing100853, People’s Republic of China
| | - Lianhua Yu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou318000, People’s Republic of China
| | - Shiyi Luo
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Department of Chemistry College and Chemical Engineering, Xiamen University, Xiamen361005, People’s Republic of China
| | - Qi Li
- Department of Clinical Laboratory, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing100091, People’s Republic of China
| | - Shuhong Huang
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong250062, People’s Republic of China
| | - Yunhe An
- Biotechnology Department, Beijing Center for Physical and Chemical Analysis, Beijing100094, People’s Republic of China
| |
Collapse
|
12
|
Chen M, Wang JM, Wang D, Wu R, Hou HW. Triptolide inhibits migration and proliferation of fibroblasts from ileocolonic anastomosis of patients with Crohn's disease via regulating the miR‑16‑1/HSP70 pathway. Mol Med Rep 2019; 19:4841-4851. [PMID: 30942423 PMCID: PMC6522880 DOI: 10.3892/mmr.2019.10117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/27/2019] [Indexed: 01/26/2023] Open
Abstract
Anastomotic fibrosis is highly likely to lead to reoperation in Crohn's disease (CD) patients. Triptolide (TPL) is considered to have anti-inflammatory and antifibrotic effects in a variety of autoimmune diseases, including CD. The present study aimed to investigate the effects of TPL on fibroblasts from strictured ileocolonic anastomosis of patients with CD and its underlying mechanism. Primary fibroblasts were obtained from strictured anastomosis tissue (SAT) samples and matched anastomosis-adjacent normal tissue (NT) samples which were collected from 10 CD patients who underwent reoperation because of anastomotic stricture. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to measure miR-16-1 and heat shock protein 70 (HSP70) levels. Western blotting was conducted to determine expression of HSP70, collagen I (Col-I), collagen III (Col-III) and α-smooth muscle actin (α-SMA) proteins. Agomir-16-1 and antagomir-16-1 were used to up and downregulate the expression of miR-16-1, respectively. Small interfering RNA (siRNA) was employed to inhibit the expression of HSP70. A wound healing assay was performed to measure the migration of fibroblasts. Cell proliferation was evaluated by MTT and 5-bromo-2-deoxyrudidine assays. Cell apoptosis was determined by caspase-3 activity and TUNEL assays. The results demonstrated that the levels of Col-I, Col-III and α-SMA were all significantly upregulated in SAT compared with NT. miR-16-1 levels in the SAT group were significantly compared with the NT group; conversely, the expression levels of HSP70 mRNA and protein in the SAT group were significantly lower compared with the NT group. Next, fibroblasts were treated with TPL to examine its effect on the miR-16-1/HSP70 pathway. The results demonstrated that the elevated expression of miR-16-1 in the SAT group was effectively inhibited by TPL treatment. Compared with the NT group, both the mRNA and protein levels of HSP70 were significantly downregulated in the SAT group cells, while TPL exhibited a strong promoting effect on HSP70 synthesis. Furthermore, upregulation of miR-16-1 reversed the effect of TPL on the miR-16-1/HSP70 pathway in fibroblasts from SAT. Overexpression of miR-16-1 significantly reversed the inhibitory effects of TPL treatment on migration, proliferation and extracellular matrix (ECM)-associated protein expression of fibroblasts from SAT. Finally, downregulation of miR-16-1 caused similar effects to the fibroblasts as the TPL treatment; however, the inhibitory effects on cell biological functions induced by antagomir-16-1 were all significantly reversed by HSP70 silencing. The present findings indicated that TPL may be a potential therapeutic option for postoperative anastomosis fibrosis of patients with CD. The miR-16-1/HSP70 pathway had a substantial role in the inhibitory effects of TPL on migration, proliferation and ECM synthesis rate of fibroblasts from strictured anastomosis tissues.
Collapse
Affiliation(s)
- Min Chen
- Department of Radiology, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210009, P.R. China
| | - Jin-Min Wang
- Department of General Surgery, Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Dong Wang
- Department of General Surgery, Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Rong Wu
- Department of General Surgery, Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Hong-Wei Hou
- Department of General Surgery, Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
13
|
Targeting cancer stem cells as therapeutic approach in the treatment of colorectal cancer. Int J Biochem Cell Biol 2019; 110:75-83. [PMID: 30818083 DOI: 10.1016/j.biocel.2019.02.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is one of the most common cancers globally. A large portion of colorectal cancer patients who are treated with conventional chemotherapy eventually develop local recurrence or metastases. The failure of a complete cure in colorectal cancer patients may be related to the lack of complete eradication of cancer stem cells when using conventional therapy. Colorectal cancer stem cells comprise a small population of tumor cells that possess the properties of rapid proliferation and differentiation. The colorectal cancer stem cells are also phenotypically and molecularly distinct, and resistant to conventional chemo-radiotherapy. Therefore, it is important to identify approaches in combination with conventional therapy for targeting and eradicating cancer cells. The aim of this review was to summarize the main findings of recent studies on targeting colorectal cancer stem cells as a novel therapeutic approach in colorectal cancer treatment.
Collapse
|
14
|
Lin SR, Weng CF. PG-Priming Enhances Doxorubicin Influx to Trigger Necrotic and Autophagic Cell Death in Oral Squamous Cell Carcinoma. J Clin Med 2018; 7:jcm7100375. [PMID: 30347872 PMCID: PMC6210351 DOI: 10.3390/jcm7100375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
Synergistic effects between natural compounds and chemotherapy drugs are believed to have fewer side effects with equivalent efficacy. However, the synergistic potential of prodigiosin (PG) with doxorubicin (Dox) chemotherapy is still unknown. This study explores the synergistic mechanism of PG and Dox against oral squamous cell carcinoma (OSCC) cells. Three OSCC cell lines were treated with different PG/Dox combinatory schemes for cytotoxicity tests and were further investigated for cell death characteristics by cell cycle flow cytometry and autophagy/apoptosis marker labelling. When OSCC cells were pretreated with PG, the cytotoxicity of the subsequent Dox-treatment was 30% higher than Dox alone. The cytotoxic efficacy of PG-pretreated was found better than those of PG plus Dox co-treatment and Dox-pretreatment. Increase of Sub-G1 phase and caspase-3/LC-3 levels without poly (ADP-ribose) polymeras (PARP) elevation indicated both autophagy and necrosis occurred in OSCC cells. Dox flux after PG-priming was further evaluated by rhodamine-123 accumulation and Dox transporters analysis to elucidate the PG-priming effect. PG-priming autophagy enhanced Dox accumulation according to the increase of rhodamine-123 accumulation without the alterations of Dox transporters. Additionally, the cause of PG-triggered autophagy was determined by co-treatment with endoplasmic reticulum (ER) stress or AMP-activated protein kinase (AMPK) inhibitor. PG-induced autophagy was not related to nutrient deprivation and ER stress was proved by co-treatment with specific inhibitor. Taken together, PG-priming autophagy could sensitize OSCC cells by promoting Dox influx without regulation of Dox transporter. The PG-priming might be a promising adjuvant approach for the chemotherapy of OSCC.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| |
Collapse
|
15
|
Sam MR, Ghoreishi S. Prodigiosin produced by Serratia marcescens inhibits expression of MMP-9 and survivin and promotes caspase-3 activation with induction of apoptosis in acute lymphoblastic leukaemia cells. J Appl Microbiol 2018; 125:1017-1029. [PMID: 29896797 DOI: 10.1111/jam.13949] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
AIMS Matrix metalloproteinase-9 (MMP-9) and survivin are involved in several steps of carcinogenesis in acute lymphoblastic leukaemia (ALL). Yet, no MMP-9 and survivin-modulating drugs with low toxicity on normal cells but high efficacy against high MMP-9- and survivin-expressing leukaemia cells have been approved for clinical application in ALL. Prodigiosin, a secondary metabolite of Serratia marcescens, induces apoptosis in different kinds of cancer cells with low toxicity on normal cells. However, little is known about the effects of this compound on the high MMP-9- and survivin-expressing leukaemia cells. METHODS AND RESULTS CCRF-CEM cells as a model for high MMP-9- and survivin-expressing ALL cells were treated with 100, 200 and 400 nmol l-1 prodigiosin after which cell number, proliferation rate, MMP-9 and survivin expression, caspase-3 activation and apoptosis were evaluated. After 24-, 48-, and 72-h treatments with 100, 200 and 400 nmol l-1 prodigiosin, proliferation rates were measured to be 92·3-76·7%, 82-63% and 63·7-46·6% respectively. Treatment with prodigiosin for 48 h decreased MMP-9 mRNA levels followed by decreases in secreted (S) and intracellular (I) MMP-9 protein levels by 20-22% and 69-72% for 100-400 nmol l-1 prodigiosin respectively. Prodigiosin decreased survivin protein levels from 40 to 26% followed by 3·7-5·6-fold increases in caspase-3 activation for the aforementioned prodigiosin concentration ranges. Treatment with 100-400 nmol l-1 prodigiosin increased the caspase-3/survivin, caspase-3/I-MMP-9 and caspase-3/S-MMP-9 ratios by 6-7·3-, 11·5-19·1- and 4·9-6·8-fold increases respectively. A dramatic increase in the number of apoptotic cells was also observed with increasing prodigiosin concentrations. CONCLUSION The inhibitory effects of prodigiosin on MMP-9 and survivin expression, as well as its pro-apoptotic capacity, represent a novel therapeutic avenue against ALL cells. SIGNIFICANCE AND IMPACT OF THE STUDY These findings provide an important and interesting basis to develop a new therapeutic compound with high potential against ALL cells.
Collapse
Affiliation(s)
- M R Sam
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
| | - S Ghoreishi
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
| |
Collapse
|
16
|
Rahiman N, Akaberi M, Sahebkar A, Emami SA, Tayarani-Najaran Z. Protective effects of saffron and its active components against oxidative stress and apoptosis in endothelial cells. Microvasc Res 2018. [PMID: 29524452 DOI: 10.1016/j.mvr.2018.03.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this study, we investigated the role of mitogen-activated protein kinase (MAPK) signaling pathways in mediation of the protective effects of saffron extract, saffron essential oil, safranal and crocin on bovine aortic endothelial cells against oxidative injury. The viability of cells in response to H2O2-induced toxicity (0.4, 2 and 10 mM) was measured using resazurin assay in the presence or absence of saffron extract (2-40 μg/ml), saffron oil (2-40 μg/ml), safranal (2-40 μM) and crocin (2-40 μM). Dichlorodihydrofluorescin diacetate was used as an indicator for the amount of reactive oxygen species (ROS) in cells at the same concentrations of samples as the former test. In addition, propidium iodide staining of the fragmented DNA was performed to measure the level of apoptotic cells by the application of 2-10 μM of crocin and safranal. Finally, the proteins involved in apoptosis were detected using western blotting at the concentration of 0, 2, 10 μM for crocin and safranal. The results indicated that all tested moieties improved viability and reduced ROS production in H2O2-treated cells (p < 0.001 compared to H2O2). In addition, a significant decrease in apoptosis (3-35%) was observed in the cells that were treated with crocin and safranal. The observed protective effects of crocin and safranal were associated with the activation of SAPK/JNK and inhibition of ERK ½ that are related to MAPK pathways. The antioxidant and anti-apoptotic activities of saffron and its ingredients in endothelial cells are mediated via MAPK signaling pathways and might be of therapeutic potential for endothelial dysfunctionalities.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Akaberi
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ahmad Emami
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Alotaibi AAA, Najafzadeh M, Davies JD, Baumgartner A, Anderson D. Inhibition of survivin expression after using oxaliplatin and vinflunine to induce cytogenetic damage in vitro in lymphocytes from colon cancer patients and healthy individuals. Mutagenesis 2017; 32:517-524. [PMID: 29040706 DOI: 10.1093/mutage/gex022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy drugs usually inflict a lethal dose to tumour cells with the consequence that these cells are being killed by cell death. However, each round of chemotherapy also causes damage to normal somatic cells. The DNA cross-linking agent oxaliplatin (OXP), which causes DNA double-strand breaks, and vinflunine (VFN), which disrupts the mitotic spindle, are two of these chemotherapy drugs which were evaluated in vitro using peripheral lymphocytes from colorectal cancer patients and healthy individuals to determine any differential response. Endpoints examined included micronucleus (MN) induction using the cytokinesis-blocked micronucleus (CBMN) assay and pancentromeric fluorescence in situ hybridisation. Also, survivin expression was monitored since it regulates the mitotic spindle checkpoint and inhibits apoptosis. OXP produced cytogenetic damage (micronuclei in binucleated cells) via its clastogenic but also previously unknown aneugenic action, possibly through interfering with topoisomerase II, whilst VFN produced micronuclei in mononucleated cells because of incomplete karyokinesis. Survivin expression was found to be significantly reduced in a concentration-dependent manner by not only OXP but surprisingly also VFN. This resulted in large numbers of multinucleated cells found with the CBMN assay. As survivin is upregulated in cancers, eliminating apoptosis inhibition might provide a more targeted chemotherapy approach; particularly, when considering VFN, which only affects cycling cells by inhibiting their mitotic spindle, and alongside possibly other pro-apoptotic compounds. Hence, these newly found properties of VFN -the inhibition of survivin expression-might demonstrate a promising chemotherapeutic approach as VFN induces less DNA damage in normal somatic cells compared to other chemotherapeutic compounds.
Collapse
Affiliation(s)
- Amal A A Alotaibi
- University of Bradford, Biomedical Sciences, Richmond Road, Bradford, West Yorkshire BD7 1DP, UK
| | - Mojgan Najafzadeh
- University of Bradford, Biomedical Sciences, Richmond Road, Bradford, West Yorkshire BD7 1DP, UK
| | - Justin D Davies
- Bradford Royal Infirmary, Bradford Hospitals NHS Trust, Duckworth Lane BD9 6RJ,UK
| | - Adolf Baumgartner
- University of Bradford, Biomedical Sciences, Richmond Road, Bradford, West Yorkshire BD7 1DP, UK.,York St John University, Biomedical Science, Lord Mayor's Walk, York, North Yorkshire YO31 7EX, UK
| | - Diana Anderson
- University of Bradford, Biomedical Sciences, Richmond Road, Bradford, West Yorkshire BD7 1DP, UK
| |
Collapse
|
18
|
Roudi R, Ebrahimi M, Shariftabrizi A, Madjd Z. Cancer stem cell research in Iran: potentials and challenges. Future Oncol 2017; 13:1809-1826. [PMID: 28776391 DOI: 10.2217/fon-2017-0091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Treatment modalities can reduce cancer-related mortality; however, a majority of patients develop drug resistance, metastasis and relapse. It has been proposed that tumorigenic characteristics of tumors are related to a proportion of cancer cells, termed cancer stem cells (CSCs). Following the first evidence regarding the existence of CSC population in acute myeloid leukemia in 1997, publications in CSCs field showed an explosive trend in all cancer types around the world. First research paper in the field of CSCs in Iran was published in 2004 on prostate cancer. Subsequently, an annual number of publications in the field of CSCs displayed a rapidly growing trend. Therefore, in the current review, we have presented a comprehensive evaluation of the CSCs research in Iran.
Collapse
Affiliation(s)
- Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells & Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran
| | - Ahmad Shariftabrizi
- Department of Nuclear Medicine & Molecular Imaging, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Cheng MF, Lin CS, Chen YH, Sung PJ, Lin SR, Tong YW, Weng CF. Inhibitory Growth of Oral Squamous Cell Carcinoma Cancer via Bacterial Prodigiosin. Mar Drugs 2017; 15:md15070224. [PMID: 28714874 PMCID: PMC5532666 DOI: 10.3390/md15070224] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/02/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy drugs for oral cancers always cause side effects and adverse effects. Currently natural sources and herbs are being searched for treated human oral squamous carcinoma cells (OSCC) in an effort to alleviate the causations of agents in oral cancers chemotherapy. This study investigates the effect of prodigiosin (PG), an alkaloid and natural red pigment as a secondary metabolite of Serratia marcescens, to inhibit human oral squamous carcinoma cell growth; thereby, developing a new drug for the treatment of oral cancer. In vitro cultured human OSCC models (OECM1 and SAS cell lines) were used to test the inhibitory growth of PG via cell cytotoxic effects (MTT assay), cell cycle analysis, and Western blotting. PG under various concentrations and time courses were shown to effectively cause cell death and cell-cycle arrest in OECM1 and SAS cells. Additionally, PG induced autophagic cell death in OECM1 and SAS cells by LC3-mediated P62/LC3-I/LC3-II pathway at the in vitro level. These findings elucidate the role of PG, which may target the autophagic cell death pathways as a potential agent in cancer therapeutics.
Collapse
Affiliation(s)
- Ming-Fang Cheng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan.
- Division of Histology and Clinical Pathology, Hualian Army Forces General Hospital, Hualien 97144, Taiwan.
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan.
| | - Yu-Hsin Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
| | - Ping-Jyun Sung
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan.
| | - Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Yi-Wen Tong
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
| |
Collapse
|
20
|
Sam MR, Pourpak RS. Regulation of p53 and survivin by prodigiosin compound derived from Serratia marcescens contribute to caspase-3-dependent apoptosis in acute lymphoblastic leukemia cells. Hum Exp Toxicol 2017; 37:608-617. [PMID: 28681618 DOI: 10.1177/0960327117718052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Tumor suppressor p53 and proto-oncogene survivin are challenging targets for anticancer drugs in acute lymphoblastic leukemia (ALL) which are associated with chemoresistance. Yet, no p53 and survivin-modulating drug with low toxicity and high efficacy has been approved for clinical application in ALL. Consequently, the search for novel compounds which target p53 or survivin is needed to further advance ALL treatment. Prodigiosin, a secondary metabolite of Serratia marcescens induces apoptosis in cancer cells with no toxicity on normal cells. However, the possible potential of prodigiosin as p53- and survivin-modulating agent in ALL cells has not been investigated. Wt-p53 Molt-4 cells were treated with 100 to 600 nM prodigiosin, after which, viability, cell proliferation rates, survivin and p53 protein levels, caspase-3 activation, and apoptosis were evaluated. After 24-, 48-, and 72-h treatments with 100 to 600 nM prodigiosin, cell proliferation rates were measured to be 93.7-77.3%, 75.5-58.3%, and 55-23.3%, respectively. Treatment for 48 hours with 100 to 600 nM prodigiosin resulted in 41-19% decrease in survivin protein levels followed by 450-950% increases in caspase-3 activation levels. Prodigiosin induced remarkably p53 accumulation and increased p53/survivin and caspase-3/survivin ratios by 6.1 to 11.3 and 10.3 to 47.5-fold at 100 to 600 nM, respectively. Survivin protein levels were inversely proportional to p53 accumulation levels. Low survivin protein levels combined with high levels of p53 accumulation were correlated to higher apoptotic rates. P53 and survivin as molecular targets of prodigiosin contribute to caspase-3-dependent apoptosis in ALL cells and this compound represents an attractive p53- and survivin-modulating agent in ALL.
Collapse
Affiliation(s)
- M R Sam
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
| | - R S Pourpak
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
| |
Collapse
|
21
|
Lin SR, Fu YS, Tsai MJ, Cheng H, Weng CF. Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy. Int J Mol Sci 2017; 18:ijms18071412. [PMID: 28671583 PMCID: PMC5535904 DOI: 10.3390/ijms18071412] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 01/07/2023] Open
Abstract
Accumulated evidence indicates that autophagy is a response of cancer cells to various anti-cancer therapies. Autophagy is designated as programmed cell death type II, and is characterized by the formation of autophagic vacuoles in the cytoplasm. Numerous herbs, including Chinese herbs, have been applied to cancer treatments as complementary and alternative medicines, supplements, or nutraceuticals to dampen the side or adverse effects of chemotherapy drugs. Moreover, the tumor suppressive actions of herbs and natural products induced autophagy that may lead to cell senescence, increase apoptosis-independent cell death or complement apoptotic processes. Hereby, the underlying mechanisms of natural autophagy inducers are cautiously reviewed in this article. Additionally, three natural compounds—curcumin, 16-hydroxycleroda-3,13-dien-15,16-olide, and prodigiosin—are presented as candidates for autophagy inducers that can trigger cell death in a supplement or alternative medicine for cancer therapy. Despite recent advancements in therapeutic drugs or agents of natural products in several cancers, it warrants further investigation in preclinical and clinical studies.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, 97401 Hualien, Taiwan.
| | - Yaw-Syan Fu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 807 Kaohsiung city, Taiwan.
| | - May-Jywan Tsai
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan.
- Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan.
| | - Henrich Cheng
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan.
- Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, 97401 Hualien, Taiwan.
| |
Collapse
|
22
|
Zhao D, Zhang Y, Song L. MiR-16-1 Targeted Silences Far Upstream Element Binding Protein 1 to Advance the Chemosensitivity to Adriamycin in Gastric Cancer. Pathol Oncol Res 2017; 24:483-488. [PMID: 28667493 DOI: 10.1007/s12253-017-0263-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/21/2017] [Indexed: 12/15/2022]
Abstract
Chemotherapy can prevent metastasis and recurrence of gastric cancer (GC), and is a well supplement for operation. But, chemotherapy resistance has severely restricted the application of chemotherapy. This study aimed to investigate the regulatory roles and molecular mechanism of miR-16-1 to the chemosensitivity to adriamycin in GC. In this study, the expression of miR-16-1 and FUBP1 was down-regulated and up-regulated respectively in adriamycin-resistant GC tissues and cell lines, and represented a negative relationship between them. MiR-16-1 could silence FUBP1 directly and specifically, FUBP1 was a target gene of miR-16-1. Silence of FUBP1 inhibited the half maximal inhibitory concentration (IC50) of SGC7901/AR cell line to adriamycin, chemosensitivity enhanced significantly. Moreover, FUBP1 silence in SGC7901/AR cell line also inhibited proliferation and invasion, and advanced cell apoptosis. To sum up, the expression of miR-16-1 was positively related with the chemosensitivity of GC to adriamycin, and miR-16-1 could targeted silence FUBP1 to advance the chemosensitivity to adriamycin in GC, which might be a novel potential therapeutic target for GC.
Collapse
Affiliation(s)
- Danyi Zhao
- Department of Oncology, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116027, China
| | - Yang Zhang
- Department of Oncology, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116027, China
| | - Lei Song
- Department of Oncology, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116027, China. .,Department of Interventional Therapy, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116027, China.
| |
Collapse
|
23
|
Li KL, Wang YF, Qin JR, Wang F, Yang YT, Zheng LW, Li MH, Kong J, Zhang W, Yang HY. Rapamycin enhances the anti-angiogenesis and anti-proliferation ability of YM155 in oral squamous cell carcinoma. Tumour Biol 2017; 39:1010428317706213. [PMID: 28618939 DOI: 10.1177/1010428317706213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
YM155, a small molecule inhibitor of survivin, has been studied in many tumors. It has been shown that YM155 inhibited oral squamous cell carcinoma through promoting apoptosis and autophagy and inhibiting proliferation. It was found that YM155 also inhibited the oral squamous cell carcinoma-mediated angiogenesis through the inactivation of the mammalian target of rapamycin pathway. Rapamycin, a mammalian target of rapamycin inhibitor, played an important role in the proliferation and angiogenesis of oral squamous cell carcinoma cell lines. In our study, cell proliferation assay, transwell assay, tube formation assay, and western blot assay were used to investigate the synergistic effect of rapamycin on YM155 in oral squamous cell carcinoma. Either in vitro or in vivo, rapamycin and YM155 exerted a synergistic effect on the inhibition of survivin and vascular endothelial growth factor through mammalian target of rapamycin pathway. Overall, our results revealed that low-dose rapamycin strongly promoted the sensitivity of oral squamous cell carcinoma cell lines to YM155.
Collapse
Affiliation(s)
- Kong-Liang Li
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yu-Fan Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jia-Ruo Qin
- 2 Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Feng Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yong-Tao Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Li-Wu Zheng
- 3 Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ming-Hua Li
- 4 Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jie Kong
- 5 Department of Stomatology, Shenzhen Children's Hospital, Shenzhen, China
| | - Wei Zhang
- 6 Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hong-Yu Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
24
|
Diamantopoulos MA, Kontos CK, Kerimis D, Papadopoulos IN, Scorilas A. Upregulated miR-16 expression is an independent indicator of relapse and poor overall survival of colorectal adenocarcinoma patients. Clin Chem Lab Med 2017; 55:737-747. [PMID: 27930363 DOI: 10.1515/cclm-2016-0756] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/31/2016] [Indexed: 02/01/2023]
Abstract
BACKGROUND Colorectal adenocarcinoma is one of the most common malignant tumors of the gastrointestinal tract and the second leading cause of cancer-related deaths among adults in Western countries. miR-16 is heavily involved in cancer progression. In this study, we examined the potential diagnostic and prognostic utility of miR-16 expression in colorectal adenocarcinoma. METHODS Total RNA was extracted from 182 colorectal adenocarcinoma specimens and 86 non-cancerous colorectal mucosae. After polyadenylation of 2 μg total RNA by poly(A) polymerase and subsequent reverse transcription with an oligo-dT adapter primer, miR-16 expression was determined using an in-house developed reverse transcription quantitative real-time PCR method, based on SYBR Green chemistry. SNORD43 (RNU43) and SNORD48 (RNU48) were used as reference genes. Next, we performed extensive biostatistical analysis. RESULTS miR-16 was shown to be significantly upregulated in colorectal adenocarcinoma specimens compared to non-cancerous colorectal mucosae, suggesting its potential exploitation for diagnostic purposes. Moreover, high miR-16 expression predicts poor disease-free survival (DFS) and overall survival (OS) of colorectal adenocarcinoma patients. Multivariate Cox regression analysis confirmed that miR-16 overexpression is a significant unfavorable prognosticator in colorectal adenocarcinoma, independent of other established prognostic factors, radiotherapy, and chemotherapy. Interestingly, miR-16 overexpression retains its unfavorable prognostic value in patients with advanced yet locally restricted colorectal adenocarcinoma that has not grown through the wall of the colon or rectum (T3) and in those without distant metastasis (M0). CONCLUSIONS Overexpression of the cancer-associated miR-16 predicts poor DFS and OS of colorectal adenocarcinoma patients, independently of clinicopathological factors that are currently used for prognostic purposes.
Collapse
Affiliation(s)
- Marios A Diamantopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens
| | - Dimitrios Kerimis
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens
| | - Iordanis N Papadopoulos
- Fourth Surgery Department, National and Kapodistrian University of Athens, University General Hospital "Attikon", Athens
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701 Athens
| |
Collapse
|
25
|
Lin Q, Ma L, Liu Z, Yang Z, Wang J, Liu J, Jiang G. Targeting microRNAs: a new action mechanism of natural compounds. Oncotarget 2017; 8:15961-15970. [PMID: 28052018 PMCID: PMC5362538 DOI: 10.18632/oncotarget.14392] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/01/2016] [Indexed: 01/22/2023] Open
Abstract
Unlike genetics, epigenetics involves the modification of genome without changes in DNA sequences, including DNA methylation, histone modification, chromatin remodeling and noncoding RNA regulation. MicroRNA (miRNA), a member of noncoding RNAs superfamily, participates in RNA interference through a unique mechanism. Currently, microRNAs have been found to be regulated by some natural compounds. Through altering the expression of miRNAs and influencing the downstream signaling pathways or target genes, several natural compounds exhibit its bioactivity in the prevention, diagnosis, therapy, prognosis and drug resistance of human diseases, such as cancer. In this review, several natural compounds and their studies about miRNA-related action mechanism were summarized. These studies provide a new insight into action mechanism by which natural compound exerts its bioactivity and a novel treatment strategy, demonstrating natural compound a promising remedy for clinical treatments.
Collapse
Affiliation(s)
- Qian Lin
- College of Medicine, Qingdao University, Qingdao, China
| | - Leina Ma
- The Department of Oncology, The First Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zhantao Liu
- College of Medicine, Qingdao University, Qingdao, China
| | - Zhihong Yang
- College of Medicine, Qingdao University, Qingdao, China
| | - Jin Wang
- College of Medicine, Qingdao University, Qingdao, China
| | - Jia Liu
- College of Medicine, Qingdao University, Qingdao, China
| | - Guohui Jiang
- College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Li W, Lee MR, Choi E, Cho MY. Clinicopathologic Significance of Survivin Expression in Relation to CD133 Expression in Surgically Resected Stage II or III Colorectal Cancer. J Pathol Transl Med 2016; 51:17-23. [PMID: 27989099 PMCID: PMC5267540 DOI: 10.4132/jptm.2016.09.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 02/06/2023] Open
Abstract
Background Cancer stem cells have been investigated as new targets for colorectal cancer (CRC) treatment. We recently reported that CD133+ colon cancer cells showed chemoresistance to 5-fluorouracil through increased survivin expression and proposed the survivin inhibitor YM155 as an effective therapy for colon cancer in an in vitro study. Here, we investigate the relationship between survivin and CD133 expression in surgically resected CRC to identify whether the results obtained in our in vitro study are applicable to clinical samples. Methods We performed immunohistochemical staining for survivin and CD133 in surgically resected tissue from 187 stage II or III CRC patients. We also comparatively analyzed apoptosis according to survivin and CD133 expression using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling. Results The results of the Mantel-Haenszel test established a linear association between nuclear survivin and CD133 expression (p = .018), although neither had prognostic significance, according to immunohistochemical expression level. No correlation was found between survivin expression and the following pathological parameters: invasion depth, lymph node metastasis, or histologic differentiation (p > .05). The mean apoptotic index in survivin+ and CD133+ tumors was higher than that in negative tumors: 5.116 ± 4.894 in survivin+ versus 4.103 ± 3.691 in survivin– (p = .044); 5.165 ± 4.961 in CD133+ versus 4.231 ± 3.812 in CD133– (p = .034). Conclusions As observed in our in vitro study, survivin expression is significantly related to CD133 expression. Survivin may be considered as a new therapeutic target for chemoresistant CRC.
Collapse
Affiliation(s)
- Wanlu Li
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mi-Ra Lee
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - EunHee Choi
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mee-Yon Cho
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Institute of Genomic Cohort, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|