1
|
Robledo-Cadena DX, Pacheco-Velázquez SC, Vargas-Navarro JL, Padilla-Flores JA, López-Marure R, Pérez-Torres I, Kaambre T, Moreno-Sánchez R, Rodríguez-Enríquez S. Synergistic celecoxib and dimethyl-celecoxib combinations block cervix cancer growth through multiple mechanisms. PLoS One 2024; 19:e0308233. [PMID: 39325741 PMCID: PMC11426494 DOI: 10.1371/journal.pone.0308233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/15/2024] [Indexed: 09/28/2024] Open
Abstract
OBJECTIVE The synergistic inhibitory effect of celecoxib (CXB) and dimethyl-celecoxib (DMC) plus paclitaxel (PA) or cisplatin (CP) on human cervix HeLa and SiHa cells was assessed at multiple cellular levels in order to elucidate the biochemical mechanisms triggered by the synergistic drug combinations. METHODS The effect of CXB (5 μM)/CP (2 μM) or CXB (5 μM)/PA (15 μM) and DMC (15 μM)/CP (5 μM) or DMC (15 μM)/PA (20 μM) for 24 h was assayed on cancer cell proliferation, energy metabolism, mitophagy, ROS production, glycoprotein-P activity, DNA stability and apoptosis/necrosis. RESULTS Drug combinations synergistically decreased HeLa and SiHa cell proliferation (>75%) and arrested cellular cycle by decreasing S and G2/M phases as well as the Ki67 content (HeLa) by 7.5-30 times. Cell viability was preserved (>90%) and no apparent effects on non-cancer cell growth were observed. Mitochondrial and glycolytic protein contents (44-95%) and ΔΨm (45-50%) in HeLa cells and oxidative phosphorylation and glycolysis fluxes (70-90%) in HeLa and SiHa cells were severely decreased, which in turn promoted a drastic fall in the ATP supply (85-88%). High levels of mitophagy proteins in HeLa cells and active mitochondrial digestion in HeLa and SiHa cells was observed. Mitochondrial fission and microtubule proteins were also affected. Intracellular ROS content (2-2.3-fold) and ROS production was stimulated (2.3-4 times), whereas content and activity of glycoprotein-P (45-85%) were diminished. DNA fragmentation was not observed and apoptosis/necrosis was not detected suggesting that cell death could be mainly associated to mitophagy induction. CONCLUSIONS CXB or DMC combination with canonical chemotherapy may be a promising chemotherapy strategy against cervical cancer growth, because it can selectively block multiple cell processes including inhibition of energy pathways and in consequence ATP-dependent processes such as cell proliferation, glycoprotein-P activity, ROS production and mitophagy, with no apparent effects on non-cancer cells.
Collapse
Affiliation(s)
- Diana Xochiquetzal Robledo-Cadena
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, C.P. 04510, CDMX, México
| | - Silvia Cecilia Pacheco-Velázquez
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States of America
| | - Jorge Luis Vargas-Navarro
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, UNAM, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, México
| | - Joaquín Alberto Padilla-Flores
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, UNAM, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, México
| | - Rebeca López-Marure
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Israel Pérez-Torres
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Rafael Moreno-Sánchez
- Laboratorio de Control Metabólico, Carrera de Biología de la Facultad de Estudios Superiores Iztacala, UNAM, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, México
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Sara Rodríguez-Enríquez
- Laboratorio de Control Metabólico, Carrera de Médico Cirujano de la Facultad de Estudios Superiores Iztacala, UNAM, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, Tlalnepantla, México
| |
Collapse
|
2
|
Wang Y, Wang C, Zhong R, Wang L, Sun L. Research progress of DNA methylation in colorectal cancer (Review). Mol Med Rep 2024; 30:154. [PMID: 38963030 PMCID: PMC11240861 DOI: 10.3892/mmr.2024.13278] [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: 03/28/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024] Open
Abstract
DNA methylation is one of the earliest and most significant epigenetic mechanisms discovered. DNA methylation refers, in general, to the addition of a methyl group to a specific base in the DNA sequence under the catalysis of DNA methyltransferase, with S‑adenosine methionine as the methyl donor, via covalent bonding and chemical modifications. DNA methylation is an important factor in inducing cancer. There are different types of DNA methylation, and methylation at different sites plays different roles. It is well known that the progression of colorectal cancer (CRC) is affected by the methylation of key genes. The present review did not only discuss the potential relationship between DNA methylation and CRC but also discussed how DNA methylation affects the development of CRC by affecting key genes. Furthermore, the clinical significance of DNA methylation in CRC was highlighted, including that of the therapeutic targets and biomarkers of methylation; and the importance of DNA methylation inhibitors was discussed as a novel strategy for treatment of CRC. The present review did not only focus upon the latest research findings, but earlier reviews were also cited as references to older literature.
Collapse
Affiliation(s)
- Yuxin Wang
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Chengcheng Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ruiqi Zhong
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Liang Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lei Sun
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
3
|
Yan L, Shi J, Zhu J. Cellular and molecular events in colorectal cancer: biological mechanisms, cell death pathways, drug resistance and signalling network interactions. Discov Oncol 2024; 15:294. [PMID: 39031216 PMCID: PMC11265098 DOI: 10.1007/s12672-024-01163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide, affecting millions each year. It emerges from the colon or rectum, parts of the digestive system, and is closely linked to both genetic and environmental factors. In CRC, genetic mutations such as APC, KRAS, and TP53, along with epigenetic changes like DNA methylation and histone modifications, play crucial roles in tumor development and treatment responses. This paper delves into the complex biological underpinnings of CRC, highlighting the pivotal roles of genetic alterations, cell death pathways, and the intricate network of signaling interactions that contribute to the disease's progression. It explores the dysregulation of apoptosis, autophagy, and other cell death mechanisms, underscoring the aberrant activation of these pathways in CRC. Additionally, the paper examines how mutations in key molecular pathways, including Wnt, EGFR/MAPK, and PI3K, fuel CRC development, and how these alterations can serve as both diagnostic and prognostic markers. The dual function of autophagy in CRC, acting as a tumor suppressor or promoter depending on the context, is also scrutinized. Through a comprehensive analysis of cellular and molecular events, this research aims to deepen our understanding of CRC and pave the way for more effective diagnostics, prognostics, and therapeutic strategies.
Collapse
Affiliation(s)
- Lei Yan
- Medical Department, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jia Shi
- Department of Obstetrics and Gynecology, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jiazuo Zhu
- Department of Oncology, Xuancheng City Central Hospital, No. 117 Tong Road, Xuancheng, Anhui, China.
| |
Collapse
|
4
|
Motamedzadeh A, Rahmati-Dehkordi F, Heydari H, Behnam M, Rashidi Noshabad FZ, Tamtaji Z, Taheri AT, Nabavizadeh F, Aschner M, Mirzaei H, Tamtaji OR. Therapeutic potential of Phycocyanin in gastrointestinal cancers and related disorders. Mol Biol Rep 2024; 51:741. [PMID: 38874869 DOI: 10.1007/s11033-024-09675-3] [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: 05/18/2023] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Gastrointestinal cancer is the most fatal cancer worldwide. The etiology of gastrointestinal cancer has yet to be fully characterized. Alcohol consumption, obesity, tobacco, Helicobacter pylori and gastrointestinal disorders, including gastroesophageal reflux disease, gastric ulcer, colon polyps and non-alcoholic fatty liver disease are among the several risks factors for gastrointestinal cancers. Phycocyanin which is abundant in Spirulina. Phycocyanin, a member of phycobiliprotein family with intense blue color, is an anti-diabetic, neuroprotective, anti-oxidative, anti-inflammatory, and anticancer compound. Evidence exists supporting that phycocyanin has antitumor effects, exerting its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, cell-cycle arrest, migration and Wnt/β-catenin signaling. Phycocyanin has also been applied in treatment of several gastrointestinal disorders such as, gastric ulcer, ulcerative colitis and fatty liver that is known as a risk factor for progression to cancer. Herein, we summarize various cellular and molecular pathways that are affected by phycocyanin, its efficacy upon combined drug treatment, and the potential for nanotechnology in its gastrointestinal cancer therapy.
Collapse
Affiliation(s)
- Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Rahmati-Dehkordi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoora Heydari
- Student Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Behnam
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Zeinab Tamtaji
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Abdolkarim Talebi Taheri
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
- Department of Physiology, School of Medicine, Tehran University of medical sciences, Tehran, Iran.
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, School of Medicine, Tehran University of medical sciences, Tehran, Iran.
| |
Collapse
|
5
|
Hedayati M, Garousi B, Rezaei Z, Nazerian Y, Yassaghi Y, Tavasol A, Zanjanbar DB, Sharifpour S, Golestani A, Bolideei M, Maleki F. Identifying SCC Lesions Capable of Spontaneous Regression by Using Immunohistochemistry: A Systematic Review and Meta-Analysis. Dermatol Pract Concept 2024; 14:dpc.1402a47. [PMID: 38810039 PMCID: PMC11135932 DOI: 10.5826/dpc.1402a47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 05/31/2024] Open
Abstract
INTRODUCTION Keratoacanthoma (KA) and squamous cell carcinoma (SCC) are two cutaneous conditions with morphological resemblance, which can complicate the diagnosis in some cases. Using immunohistochemistry staining of biomarkers could be beneficial in resolving this obstacle. OBJECTIVES We investigated a variety of biomarkers assessed in different studies in order to find the most important and helpful biomarkers for differentiation between SCC and lesions capable of spontaneous regression. METHODS MEDLINE via PubMed and Google Scholar database were used to identify relevant literature up to 15 June 2022. The aim of our analyses was to determine the capability of biomarkers to distinguish between SCC and lesions capable of spontaneous regression using calculated individual and pooled odds ratios (OR) and 95% confidence intervals (CI) and I2 tests. RESULTS Six potential biomarkers were CD10 with pooled OR= 0.006 (95% CI: 0.001-0.057) and I2=0%; COX-2 with pooled OR=0.089 (95% CI: 0.029-0.269) and I2=17.1%; elastic fibers with pooled OR= 6.69 (95% CI: 2.928-15.281) and I2=0%; IMP-3 with pooled OR=0.145 (95% CI: 0.021-1.001) and I2=44.5%; P53 with pooled OR=0.371 (95% CI: 0.188-0.733) and I2=55.9%; AT1R with OR=0.026 (95% CI: 0.006-0.107). CONCLUSIONS We suggest the utilization of the following IHC biomarkers for discrimination between lesions with spontaneous regression such as KA and SCC: CD10, COX-2, and elastic fibers.
Collapse
Affiliation(s)
| | - Behzad Garousi
- Department of Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Younes Yassaghi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arian Tavasol
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Dorsa Bahrami Zanjanbar
- Pharmaceutical Science Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Amir Golestani
- Students Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mansoor Bolideei
- The Center for Biomedical Research, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Farajolah Maleki
- Non-Communicable Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
6
|
Ubhenin AE, Adefolalu AA, Oriakhi K, Adamude FA, Dingwoke EJ, Ikebuiro JO, Chiwendu BC, Muhammad ML, Omage K. Caesalpinia pulcherrima lowered serum carcinoembryonic antigen and antigen 125 in 7,12-Dimethylbenz[ a]anthracene-induced Mammary Carcinogenesis in Female Albino Rats. Heliyon 2024; 10:e23401. [PMID: 38187255 PMCID: PMC10770447 DOI: 10.1016/j.heliyon.2023.e23401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/07/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Aim This study is aimed at evaluating the anticancer effect of the aqueous extract of Caesalpinia pulcherrima (L.) Sw in 7,12-Dimethlbenz[a]anthracene (DMBA) - induced mammary cancer. Methods Tumors were induced via a single intraperitoneal injection of DMBA (dissolved in olive oil) at a dose of 80 mg/kg body weight to the test rats and allowed to develop for about four months. They were treated with cyclophosphamide and an aqueous extract of Caesalpinia pulcherrima at doses of 10 and 250 mg/kg body weight, respectively, for 28 days. Serum levels of cancer antigen 125 (CA125), carcinoembryonic antigen (CEA) activity, cyclooxygenase-2 (COX-2), and cytochrome p450 oxidase (cytp450) activity, as well as other diagnostic enzymes, were estimated. Results The result revealed that DMBA is associated with a significant (p < 0.05) increase in the serum levels of CA125, CEA, COX-2, cytp450, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) of the rats, thus suggesting tumor-promoting and hepatotoxic effects of DMBA. There was also a significant (p < 0.05) reduction of serum levels of these cancer and liver biomarker enzymes in the groups treated with cyclophosphamide and Caesalpinia pulcherrima compared to the untreated group, thus suggesting anticancer activity of Caesalpinia pulcherrima. The anticancer effect of Caesalpinia pulcherrima was further confirmed by the disappearance of infiltrative fibrous cells and the absence of inflammatory cells from the photomicrographs of the rats treated with Caesalpinia pulcherrima. Conclusion Our findings show that Caesalpinia pulcherrima possesses anticancer activity, and could protect against mammary cancer.
Collapse
Affiliation(s)
- Abraham Ehinomhen Ubhenin
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Adedotun A. Adefolalu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kelly Oriakhi
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Benin, Benin City, Nigeria
| | - Fatima Adis Adamude
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Emeka John Dingwoke
- Department of Tropical Diseases, UNESCO-International Center for Biotechnology, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Joshua Onyeka Ikebuiro
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Benjamin Comfort Chiwendu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Mabruqah Liman Muhammad
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kingsley Omage
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, United States
| |
Collapse
|
7
|
Rodrigues P, Bangali H, Hammoud A, Mustafa YF, Al-Hetty HRAK, Alkhafaji AT, Deorari MM, Al-Taee MM, Zabibah RS, Alsalamy A. COX 2-inhibitors; a thorough and updated survey into combinational therapies in cancers. Med Oncol 2024; 41:41. [PMID: 38165473 DOI: 10.1007/s12032-023-02256-7] [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: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024]
Abstract
Cyclooxygenase (COX) enzymes are pivotal in inflammation and cancer development. COX-2, in particular, has been implicated in tumor growth, angiogenesis, and immune evasion. Recently, COX-2 inhibitors have arisen as potential therapeutic agents in cancer treatment. In addition, combining COX inhibitors with other treatment modalities has demonstrated the potential to improve therapeutic efficacy. This review aims to investigate the effects of COX inhibition, both alone and in combination with other methods, on signaling pathways and carcinogenesis in various cancers. In this study, a literature search of all major academic databases was conducted (PubMed, Scholar google), including the leading research on the mechanisms of COX-2, COX-2 inhibitors, monotherapy with COX-2 inhibitors, and combining COX-2-inhibitors with chemotherapeutic agents in tumors. The study encompasses preclinical and clinical evidence, highlighting the positive findings and the potential implications for clinical practice. According to preclinical studies, multiple signaling pathways implicated in tumor cell proliferation, survival, invasion, and metastasis can be suppressed by inhibiting COX. In addition, combining COX inhibitors with chemotherapy drugs, targeted therapies, immunotherapies, and miRNA-based approaches has enhanced anti-tumor activity. These results suggest that combination therapy has the potential to overcome resistance mechanisms and improve treatment outcomes. However, caution must be exercised when selecting and administering combination regimens. Not all combinations of COX-2 inhibitors with other drugs result in synergistic effects; some may even have unfavorable interactions. Therefore, personalized approaches that consider the specific characteristics of the cancer and the medications involved are crucial for optimizing therapeutic strategies. In conclusion, as monotherapy or combined with other methods, COX inhibition bears promise in modulating signaling pathways and inhibiting carcinogenesis in various cancers. Additional studies and well-designed clinical trials are required to completely elucidate the efficacy of COX inhibition and combination therapy in enhancing cancer treatment outcomes. This narrative review study provides a detailed summary of COX-2 monotherapy and combination targeted therapy in cancer treatment.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Ahmad Hammoud
- Department of Medical and Technical Information Technology, Bauman Moscow State Technical University, Moscow, Russia.
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Mishref Campus, Mubarak Al-Abdullah, Kuwait.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Maha Medha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Rahman S Zabibah
- College of Medical Technique, the Islamic University, Najaf, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| |
Collapse
|
8
|
Zhang H, Dou Z, Bi W, Yang C, Wu X, Wang L. Multi-omics study of sulfur metabolism affecting functional microbial community succession during aerobic solid-state fermentation. BIORESOURCE TECHNOLOGY 2023; 387:129664. [PMID: 37573975 DOI: 10.1016/j.biortech.2023.129664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
Microbial-mediated sulfur metabolism is closely related to carbon and nitrogen metabolism in natural biological systems. In this study, the effects of sulfur metabolism on microbial communities and functional enzyme succession were investigated based on integrated multi-omics by adding sulfur-containing compounds to aerobic fermentation systems. Sulfur powder was oxidized to S2O32- and subsequently to SO42- by the microbial sulfur-oxidizing system, which lowered the pH to 7.5 on day 7. The decrease in pH resulted in Planifilum (secreted S8, M17 and M32 proteases) losing its competitive advantage, whereas Novibacillus (secreted M14 and M19 metalloproteases) became dominant. Structural proteomics indicated that the surface of Novibacillus proteases has more negatively charged amino acid residues that help maintain protein stability at low pH. These findings aid understanding of the effects of sulfur metabolism on fermentation and the mechanism of microbial adaptation after pH reduction, providing new perspectives on the optimization of fermentation processes.
Collapse
Affiliation(s)
- Hong Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Zhixin Dou
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Wenhui Bi
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China; Faculty of Food Science and Engineering, Shandong Agricultural and Engineering University, Jinan, Shandong 250100, China
| | - Chuanlun Yang
- Shandong Chambroad Holding Group Co., Ltd., Boxing 256599, China
| | - Xiuyun Wu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
9
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Yu C, He S, Zhu W, Ru P, Ge X, Govindasamy K. Human cytomegalovirus in cancer: the mechanism of HCMV-induced carcinogenesis and its therapeutic potential. Front Cell Infect Microbiol 2023; 13:1202138. [PMID: 37424781 PMCID: PMC10327488 DOI: 10.3389/fcimb.2023.1202138] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. Human cytomegalovirus (HCMV), a well-studied herpesvirus, has been implicated in malignancies derived from breast, colorectal muscle, brain, and other cancers. Intricate host-virus interactions are responsible for the cascade of events that have the potential to result in the transformed phenotype of normal cells. The HCMV genome contains oncogenes that may initiate these types of cancers, and although the primary HCMV infection is usually asymptomatic, the virus remains in the body in a latent or persistent form. Viral reactivation causes severe health issues in immune-compromised individuals, including cancer patients, organ transplants, and AIDS patients. This review focuses on the immunologic mechanisms and molecular mechanisms of HCMV-induced carcinogenesis, methods of HCMV treatment, and other studies. Studies show that HCMV DNA and virus-specific antibodies are present in many types of cancers, implicating HCMV as an important player in cancer progression. Importantly, many clinical trials have been initiated to exploit HCMV as a therapeutic target for the treatment of cancer, particularly in immunotherapy strategies in the treatment of breast cancer and glioblastoma patients. Taken together, these findings support a link between HCMV infections and cellular growth that develops into cancer. More importantly, HCMV is the leading cause of birth defects in newborns, and infection with HCMV is responsible for abortions in pregnant women.
Collapse
Affiliation(s)
- Chuan Yu
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang Polytechnic, Luoyang, Henan, China
| | - Suna He
- Department of Pharmaceutical Sciences, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, Henan, China
| | - Wenwen Zhu
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang Polytechnic, Luoyang, Henan, China
| | - Penghui Ru
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang Polytechnic, Luoyang, Henan, China
| | - Xuemei Ge
- School of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, China
| | - Kavitha Govindasamy
- School of Arts and Science, Rutgers, the State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
11
|
Solanes Vilanova F, Hellebuyck T, Chiers K. Histological Variants of Squamous and Basal Cell Carcinoma in Squamates and Chelonians: A Comprehensive Classification. Animals (Basel) 2023; 13:ani13081327. [PMID: 37106890 PMCID: PMC10135371 DOI: 10.3390/ani13081327] [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: 03/21/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
In the present study, the histological characteristics of squamous cell carcinomas (SCCs) and basal cell carcinomas (BCCs) obtained from 22 squamate and 13 chelonian species were retrospectively evaluated. While the examined tissues were originally diagnosed as 28 SCCs and 7 BCCs based on histological evaluation by a specialty diagnostic service, eight SCCs could be re-classified as BCCs and three SCCs proved to be non-neoplastic lesions. In addition, all SCCs and BCCs were classified into distinct histological variants. The SCCs could be categorized as one SCC in situ, three moderately differentiated SCCs, seven well-differentiated SCCs, and six keratoacanthomas. BCCs were classified as five solid BCCs, four infiltrating BCCs, five keratotic BCCs, and one basosquamous cell carcinoma. In addition, the present study reports the occurrence of BCCs in seven reptile species for the first time. In contrast to what has been documented in humans, IHC staining with the commercially available epithelial membrane antigen and epithelial antigen clone Ber-EP4 does not allow differentiation of SCCs from BCCs in reptiles, while cyclooxygenase-2 and E-cadherin staining seem to have discriminating potential. Although the gross pathological features of the examined SCCs and BCCs were highly similar, each tumor could be unequivocally assigned to a distinct histological variant according to the observed histological characteristics. Based on the results of this study, a histopathological classification for SCCs and BCCs is proposed, allowing accurate identification and differentiation of SCCs and BCCs and their histological variants in the examined reptile species. Presumably, BCCs are severely underdiagnosed in squamates and chelonians.
Collapse
Affiliation(s)
- Ferran Solanes Vilanova
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Tom Hellebuyck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Koen Chiers
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| |
Collapse
|
12
|
Takala R, Ramji DP, Choy E. The Beneficial Effects of Pine Nuts and Its Major Fatty Acid, Pinolenic Acid, on Inflammation and Metabolic Perturbations in Inflammatory Disorders. Int J Mol Sci 2023; 24:ijms24021171. [PMID: 36674687 PMCID: PMC9861571 DOI: 10.3390/ijms24021171] [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: 10/19/2022] [Revised: 11/24/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Inflammatory disorders such as atherosclerosis, diabetes and rheumatoid arthritis are regulated by cytokines and other inflammatory mediators. Current treatments for these conditions are associated with significant side effects and do not completely suppress inflammation. The benefits of diet, especially the role of specific components, are poorly understood. Polyunsaturated fatty acids (PUFAs) have several beneficial health effects. The majority of studies on PUFAs have been on omega-3 fatty acids. This review will focus on a less studied fatty acid, pinolenic acid (PNLA) from pine nuts, which typically constitutes up to 20% of its total fatty acids. PNLA is emerging as a dietary PUFA and a promising supplement in the prevention of inflammatory disorders or as an alternative therapy. Some studies have shown the health implications of pine nuts oil (PNO) and PNLA in weight reduction, lipid-lowering and anti-diabetic actions as well as in suppression of cell invasiveness and motility in cancer. However, few reviews have specifically focused on the biological and anti-inflammatory effects of PNLA. Furthermore, in recent bioinformatic studies on human samples, the expression of many mRNAs and microRNAs was regulated by PNLA indicating potential transcriptional and post-transcriptional regulation of inflammatory and metabolic processes. The aim of this review is to summarize, highlight, and evaluate research findings on PNO and PNLA in relation to potential anti-inflammatory benefits and beneficial metabolic changes. In this context, the focus of the review is on the potential actions of PNLA on inflammation along with modulation of lipid metabolism and oxidative stress based on data from both in vitro and in vivo experiments, and human findings, including gene expression analysis.
Collapse
Affiliation(s)
- Rabaa Takala
- Division of Infection and Immunity, Tenovus Building, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Ernest Choy
- Division of Infection and Immunity, Tenovus Building, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- Department of Rheumatology, Heath Park, University Hospital of Wales, Cardiff CF14 4XW, UK
- Correspondence:
| |
Collapse
|
13
|
Zhang A, Zou X, Yang S, Yang H, Ma Z, Li J. Effect of NETs/COX-2 pathway on immune microenvironment and metastasis in gastric cancer. Front Immunol 2023; 14:1177604. [PMID: 37153547 PMCID: PMC10156975 DOI: 10.3389/fimmu.2023.1177604] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background Neutrophil extracellular traps (NETs) are crucial in the progression of several cancers. The formation of NETs is closely related to reactive oxygen species (ROS), and the granule proteins involved in nucleosome depolymerization under the action of ROS together with the loosened DNA compose the basic structure of NETs. This study aims to investigate the specific mechanisms of NETs promoting gastric cancer metastasis in order to perfect the existing immunotherapy strategies. Methods In this study, the cells and tumor tissues of gastric cancer were detected by immunological experiments, real-time polymerase chain reaction and cytology experiments. Besides, bioinformatics analysis was used to analyze the correlation between cyclooxygenase-2 (COX-2) and the immune microenvironment of gastric cancer, as well as its effect on immunotherapy. Results Examination of clinical specimens showed that NETs were deposited in tumor tissues of patients with gastric cancer and their expression was significantly correlated with tumor staging. Bioinformatics analysis showed that COX-2 was involved in gastric cancer progression and was associated with immune cell infiltration as well as immunotherapy. In vitro experiments, we demonstrated that NETs could activate COX-2 through Toll-like receptor 2 (TLR2) and thus enhance the metastatic ability of gastric cancer cells. In addition, in a liver metastasis model of nude mice we also demonstrated the critical role of NETs and COX-2 in the distant metastasis of gastric cancer. Conclusion NETs can promote gastric cancer metastasis by initiating COX-2 through TLR2, and COX-2 may become a target for gastric cancer immunotherapy.
Collapse
Affiliation(s)
- Ange Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Xiaoming Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- *Correspondence: Xiaoming Zou,
| | - Shifeng Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Hao Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Zhen Ma
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Jiacheng Li
- Department of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
14
|
Marangio A, Biccari A, D’Angelo E, Sensi F, Spolverato G, Pucciarelli S, Agostini M. The Study of the Extracellular Matrix in Chronic Inflammation: A Way to Prevent Cancer Initiation? Cancers (Basel) 2022; 14:cancers14235903. [PMID: 36497384 PMCID: PMC9741172 DOI: 10.3390/cancers14235903] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
Bidirectional communication between cells and their microenvironment has a key function in normal tissue homeostasis, and in disease initiation, progression and a patient's prognosis, at the very least. The extracellular matrix (ECM), as an element of all tissues and cellular microenvironment, is a frequently overlooked component implicated in the pathogenesis and progression of several diseases. In the inflammatory microenvironment (IME), different alterations resulting from remodeling processes can affect ECM, progressively inducing cancer initiation and the passage toward a tumor microenvironment (TME). Indeed, it has been demonstrated that altered ECM components interact with a variety of surface receptors triggering intracellular signaling that affect cellular pathways in turn. This review aims to support the notion that the ECM and its alterations actively participate in the promotion of chronic inflammation and cancer initiation. In conclusion, some data obtained in cancer research with the employment of decellularized ECM (dECM) models are described. The reported results encourage the application of dECM models to investigate the short circuits contributing to the creation of distinct IME, thus representing a potential tool to avoid the progression toward a malignant lesion.
Collapse
Affiliation(s)
- Asia Marangio
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Andrea Biccari
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Edoardo D’Angelo
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Francesca Sensi
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Marco Agostini
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Correspondence: ; Tel.: +39-049-964-0160
| |
Collapse
|
15
|
Huang HY, Lu TW, Liang HL, Hsu WH, Sung YW, Lee MY. Antiplatelet agents aspirin and dipyridamole, and the risk of different carcinoma in patients with type 2 diabetes mellitus: A Taiwan retrospective cohort study. Medicine (Baltimore) 2022; 101:e30468. [PMID: 36123870 PMCID: PMC9478216 DOI: 10.1097/md.0000000000030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Studies have shown aspirin decreases the risk of some cancers. However, the evidence reported the association between aspirin and cancer risk in the diabetic population. In this study, we investigate whether aspirin and dipyridamole decrease the risk of cancer in patients with type 2 diabetes. A total of 5308 patients with type 2 diabetes were identified by the National Health Insurance from 1998 to 2000 and followed up until 2013. The demographic characteristics among nondipyridamole nor aspirin, aspirin, and dipyridamole users were analyzed by using the χ(2) test. Cox proportional hazard regression models were used to determine the independent effects of no aspirin nor dipyridamole, aspirin, and dipyridamole users on the risk of different cancer. After adjustment with multiple covariates, both low and high doses of aspirin and dipyridamole decrease liver cancer with risk ratios of 0.56 (95% CI, 0.37-0.83), 0.14 (95% CI, 0.05-0.39), 0.61 (95% CI, 0.38-0.99), and 0.28 (95% CI, 0.12-0.66), respectively. Both low and high doses of aspirin decrease any types of cancer with risk ratios of 0.79 (95% CI, 0.64-0.98) and 0.49 (95% CI, 0.34-0.70), respectively. Therefore, we conclude aspirin may decrease any types of cancer and liver cancer, and dipyridamole may decrease the risk of liver cancer in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Hsing-Yi Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tz-Wen Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiu-Ling Liang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Hao Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Wen Sung
- Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Mei-Yueh Lee, Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan (e-mail: )
| |
Collapse
|
16
|
Yadav M, Abdalla M, Madhavi M, Chopra I, Bhrdwaj A, Soni L, Shaheen U, Prajapati L, Sharma M, Sikarwar MS, Albogami S, Hussain T, Nayarisseri A, Singh SK. Structure-Based Virtual Screening, Molecular Docking, Molecular Dynamics Simulation and Pharmacokinetic modelling of Cyclooxygenase-2 (COX-2) inhibitor for the clinical treatment of Colorectal Cancer. MOLECULAR SIMULATION 2022. [DOI: 10.1080/08927022.2022.2068799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Manasi Yadav
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, PR People’s Republic of China
| | - Maddala Madhavi
- Department of Zoology, Osmania University, Hyderabad, Telangana State, India
| | - Ishita Chopra
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Bioinformatics Research Laboratory, LeGene Biosciences Pvt Ltd, Indore, Madhya Pradesh, India
| | - Anushka Bhrdwaj
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Lovely Soni
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Uzma Shaheen
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Leena Prajapati
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Megha Sharma
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | | | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Tajamul Hussain
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Bioinformatics Research Laboratory, LeGene Biosciences Pvt Ltd, Indore, Madhya Pradesh, India
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
17
|
Wang D, Song X, Zhang N, Guo Y. MiR-1179 represses cell proliferation, migration and invasion of hepatocellular carcinoma through suppression of NUAK2. Am J Transl Res 2022; 14:223-239. [PMID: 35173840 PMCID: PMC8829634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/09/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To investigate the function and mechanism of miR-1179 in the tumorigenesis of hepatocellular carcinoma (HCC). METHODS The levels of miR-1179 and NUAK2 in clinical tissues or cell lines were examined using quantitative Real-time PCR (qRT-PCR). Cell Counting Kit-8 (CCK-8), EdU assay, colony formation, wound healing, Transwell assays and flow cytometry assays were conducted to examine the impact of miR-1179 on HCC cells. The protein expression of NUAK2 was detected using Western blotting assay. Bioinformatics analysis and luciferase reporter assays were conducted to reveal the association of miR-1179 with NUAK2. RESULTS MiR-1179 expression was significantly downregulated in HCC specimens and cell lines compared to normal samples and cells. The miR-1179 overexpression inhibited HCC cell migration, invasion and proliferation through targeting NUAK2. Overexpression of NUAK2 can reverse the effect of miR-1179 on hepatocellular carcinoma cells. CONCLUSION miR-1179 suppresses HCC developmen through targeting NUAK2. Which can be used as a potent HCC diagnostic marker.
Collapse
Affiliation(s)
- Dejun Wang
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University Nanjing, Jiangsu Province, China
| | - Xue Song
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University Nanjing, Jiangsu Province, China
| | - Nan Zhang
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University Nanjing, Jiangsu Province, China
| | - Yesong Guo
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University Nanjing, Jiangsu Province, China
| |
Collapse
|
18
|
Alkan U, Bachar G, Nachalon Y, Zlotogorsky A, Gal Levin E, Kaplan I. Proliferative verrucous leukoplakia: a clinicopathological comparative study. Int J Oral Maxillofac Surg 2022; 51:1027-1033. [DOI: 10.1016/j.ijom.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 12/26/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
|
19
|
Hong S, Dia VP, Zhong Q. Synergistic anti-inflammatory activity of apigenin and curcumin co-encapsulated in caseins assessed with lipopolysaccharide-stimulated RAW 264.7 macrophages. Int J Biol Macromol 2021; 193:702-712. [PMID: 34717976 DOI: 10.1016/j.ijbiomac.2021.10.153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
Dietary polyphenols are potential anti-inflammatory agents, and their combinations with enhanced biological activities may lower toxicity and side effects. The objective of this work was to investigate the potential synergistic anti-inflammatory activities of apigenin and curcumin co-nanoencapsulated in sodium caseinate, with comparison to unencapsulated polyphenol combinations. Non-toxic concentrations of apigenin, curcumin, and their combinations in the free and co-encapsulated forms were studied in lipopolysaccharide-stimulated RAW 264.7 macrophage cells. Combinations of free polyphenols produced stronger inhibition of nitric oxide (NO) production, more significant at a higher proportion of curcumin, which was further enhanced after co-encapsulation. The enhanced reduction of NO was concomitant with the decreased expression of iNOS, the enhanced inhibition of pro-inflammatory cytokines of IL-6 and TNF-α, and the reduced production of intracellular reactive oxygen species. The potential multi-target effects and the enhanced solubility, proximity, and bioavailability of AP and CUR after co-encapsulation contributed to the synergistic activities. These results demonstrated that co-nanoencapsulation of apigenin and curcumin may enable the practical application utilizing the synergistic anti-inflammation effects to improve health.
Collapse
Affiliation(s)
- Shan Hong
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA.
| | - Vermont P Dia
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA
| | - Qixin Zhong
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
20
|
Zhang X, Zhu Y, Li J, Zhu P, Liang B. Exploring dynamics and associations of dominant lignocellulose degraders in tomato stalk composting. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2021; 294:113162. [PMID: 34214942 DOI: 10.1016/j.jenvman.2021.113162] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
The plant residues of tomato bring pressures to the environment and composting provides a feasible method to treat such agricultural waste. However, little is known about the succession and associations of the dominant lignocellulose degraders in the compost system. To further accelerate the process by inoculating key functional microorganisms, a compost pile composed of tomato stalk with maize straw addition was constructed, and the whole community structure and functions of the dominant were investigated by applying the integrated mata-omics. Results showed that Actinobacteria, Firmicutes, and Ascomycota dominated and drove the assembly of the co-occurrence network. In the thermophilic stage, Thermobifida was the exclusive degrader of cellulose, and Thermobifida fusca was the most important cellulolytic actinomycete. Saccharomonospora viridis, Planifilum fulgidum, Thermobacillus sp. and the dominant ascomycota of Aspergillus sclerotialis participated in hemicellulose decomposing. In the cooling phase, functional microorganisms became more diverse, with Nocardiopsis flavescens, Glycomyces artemisiae, Glycomyces sambucus, Streptomyces rubrolavendulae and Streptomyces vietnamensis joining the cellulose-degrading rank, and Chaetomium thermophilum emerging as the main hemicellulose degrader. More than two thirds of the bacteria-bacteria interactions and all the fungi-fungi associations were positive, while, both competition (for the same substrate of hemicellulose) and synergy (preference for cellulose and hemicellulose) coexisted in the bacteria-fungi interactions. In conclusion, these findings provide useful information for understanding the biodegradation of tomato plant residues better, and effects of the functional agents identified on composting process should be further studied.
Collapse
Affiliation(s)
- Xiaomei Zhang
- College of Resource and Environment, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yi Zhu
- College of Resource and Environment, Qingdao Agricultural University, Qingdao, 266109, China
| | - Junliang Li
- College of Resource and Environment, Qingdao Agricultural University, Qingdao, 266109, China
| | - Pengcheng Zhu
- Institute of Ecology and Biodiversity, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Bin Liang
- College of Resource and Environment, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
21
|
Planifilum fulgidum Is the Dominant Functional Microorganism in Compost Containing Spent Mushroom Substrate. SUSTAINABILITY 2021. [DOI: 10.3390/su131810002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The extensive accumulation of spent mushroom substrate (SMS) owing to the large-scale production of edible fungi is causing environmental problems that cannot be ignored. Co-composting is a promising method for agricultural and animal husbandry waste disposal. In this study, the composition and function of microbial communities in the process of cattle manure–maize straw composting with SMS addition were compared through an integrated meta-omics approach. The results showed that irrespective of SMS addition, the predominant fungi were Ascomycota, while the dominant bacteria were Firmicutes, Proteobacteria, Actinobacteria, and Bacteroidetes. High temperature promoted the evolution from Gram-negative bacteria (Bacteroides, Proteobacteria) to Gram-positive bacteria (Firmicutes, Actinomycetes). The composting process was accelerated by SMS addition, and the substrate was effectively degraded in 14 days. Metaproteomics results showed that the dominant microorganism, Planifilum fulgidum, secreted large amounts of S8, M17, and M32 proteases that could degrade macromolecular protein substrates in the presence of SMS. Planifilum fulgidum, along with Thermobifida fusca and Melanocarpus albomyces, synergistically degraded hemicellulose, cellulose, and protein. In addition, the dominant microorganisms related to the initial raw materials such as Pichia, Lactobacillus in the microbial agent and Hypsizygus in SMS could not adapt to the high-temperature environment (>60 °C) and were replaced by thermophilic bacteria after 5 days of composting.
Collapse
|
22
|
Zhang Y, Xia Q, Wu T, He Z, Li Y, Li Z, Hou X, He Y, Ruan S, Wang Z, Sun J, Feng N. A novel multi-functionalized multicellular nanodelivery system for non-small cell lung cancer photochemotherapy. J Nanobiotechnology 2021; 19:245. [PMID: 34391438 PMCID: PMC8364713 DOI: 10.1186/s12951-021-00977-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A red blood cell membrane (RBCm)-derived drug delivery system allows prolonged circulation of an antitumor treatment and overcomes the issue of accelerated blood clearance induced by PEGylation. However, RBCm-derived drug delivery systems are limited by low drug-loading capacities and the lack of tumor-targeting ability. Thus, new designs of RBCm-based delivery systems are needed. RESULTS Herein, we designed hyaluronic acid (HA)-hybridized RBCm (HA&RBCm)-coated lipid multichambered nanoparticles (HA&RBCm-LCNPs) to remedy the limitations of traditional RBCm drug delivery systems. The inner core co-assembled with phospholipid-regulated glycerol dioleate/water system in HA&RBCm-LCNPs met the required level of blood compatibility for intravenous administration. These newly designed nanocarriers had a honeycomb structure with abundant spaces that efficiently encapsulated paclitaxel and IR780 for photochemotherapy. The HA&RBCm coating allowed the nanocarriers to overcome the reticuloendothelial system barrier and enhanced the nanocarriers specificity to A549 cells with high levels of CD44. These properties enhanced the combinatorial antitumor effects of paclitaxel and IR780 associated with microtubule destruction and the mitochondrial apoptotic pathway. CONCLUSIONS The multifunctional HA&RBCm-LCNPs we designed expanded the functionality of RBCm and resulted in a vehicle for safe and efficient antitumor treatment.
Collapse
Affiliation(s)
- Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qing Xia
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Wu
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zehui He
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yanyan Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhe Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xuefeng Hou
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuanzhi He
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuyao Ruan
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi Wang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Sun
- Teaching Experiment Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
23
|
Preliminary evaluation of anticancer efficacy of pioglitazone combined with celecoxib for the treatment of non-small cell lung cancer. Invest New Drugs 2021; 40:1-9. [PMID: 34341904 DOI: 10.1007/s10637-021-01158-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Among the lung cancer types, non-small cell lung cancer (NSCLC) is prominent and less responsive to chemotherapy. The current chemotherapeutics for NSCLC are associated with several dose-limiting side effects like bone-marrow suppression, neurotoxicity, nephrotoxicity, and ototoxicity, etc. which are causing non-compliance in patients. Many tumors, including breasts, lung, ovarian, etc. overexpress PPAR-γ receptors and COX-2 enzymes, which play a crucial role in tumor progression, angiogenesis, and metastasis. Lack of PPAR-γ activation and overproduction of prostaglandins, result in uncontrolled activation of Ras/Raf/Mek ultimately, NF-κB mediated tumor proliferation. This study aimed to investigate the anti-cancer potential of PPAR-γ agonist Pioglitazone combined with COX-2 inhibitor Celelcoxib in NSCLC. METHODS Sixty adult Balb/C male mice were classified into sham control, disease control, and treatment groups. Mice were treated with Nicotine-derived nitrosamine ketone (NNK) (10 mg/kg), pioglitazone (10 & 20 mg/kg) and celecoxib (25 & 50 mg/kg). Weekly body weight, food intake, mean survival time & % increased life span were determined. Tumor weight and histopathological analysis were performed at the end of the study. RESULTS The significant tumor reducing potential of pioglitazone combined with celecoxib was observed (p < 0.05). The treatment groups (treated with pioglitazone and celecoxib) showed a remarkable decrease in lung tumor weight, improved life span and mean survival time (p < 0.05). Histopathological studies confirm that treatment groups (treated with pioglitazone and celecoxib) reframed the lung architecture compared to disease control. CONCLUSION Preliminary results revealed that pioglitazone adjunacy with celecoxib may be an effective chemo-preventive agent against NNK induce NSCLC.
Collapse
|
24
|
Zhang H, Chi J, Hu J, Ji T, Luo Z, Zhou C, Huang L, Dai Z, Li J, Wang G, Wang L, Wang Z. Intracellular AGR2 transduces PGE2 stimuli to promote epithelial-mesenchymal transition and metastasis of colorectal cancer. Cancer Lett 2021; 518:180-195. [PMID: 34216690 DOI: 10.1016/j.canlet.2021.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023]
Abstract
Human anterior gradient homolog 2 (AGR2) reportedly acts as an oncogene in multiple types of cancers. As a secreted protein, the oncogenic roles of extracellular AGR2 have been the focus of the increasing number of studies. In contrast, the oncological functions of intracellular AGR2 (iAGR2) remain elusive. Here, we report that intracellular AGR2 (iAGR2) is sufficient to promote CRC metastasis. iAGR2 binds to KDEL receptors (KDELRs) via its KTEL motif to activate downstream Gs-PKA signaling. Activated PKA upregulates the expression of NF-κB subunit c-Rel (REL) and acetylates histone H3 at lysine 9 (H3K9ac) to promote the transcription of SNAIL and SLUG. AGR2 can be upregulated by prostaglandin E2 (PGE2) via EP4-PI3K-AKT pathway and is indispensable for PGE2-induced CRC metastasis. AGR2 knockdown enhances therapeutic effects of a COX-2 inhibitor, celecoxib, in CRC metastasis. Collectively, our study reveals a promoting role and molecular mechanisms of iAGR2 in CRC metastasis and uncovers a new tumor microenvironment signal regulating AGR2 expression, which may provide new targets for treating metastatic CRC.
Collapse
Affiliation(s)
- Hongyan Zhang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jiangyang Chi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jia Hu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Tiantian Ji
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zhen Luo
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Caihong Zhou
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Lifeng Huang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zheng Dai
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jing Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China; Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
25
|
Cao W, Ma X, Fischer JV, Sun C, Kong B, Zhang Q. Immunotherapy in endometrial cancer: rationale, practice and perspectives. Biomark Res 2021; 9:49. [PMID: 34134781 PMCID: PMC8207707 DOI: 10.1186/s40364-021-00301-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy has attracted more and more attention nowadays, and multiple clinical trials have confirmed its effect in a variety of solid tumors. Immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), and lymphocyte-promoting cytokines are the main immunotherapy methods. Endometrial cancer (EC) is one of the most frequent tumors in women and the prognosis of recurrent or metastatic EC is poor. Since molecular classification has been applied to EC, immunotherapy for different EC subtypes (especially POLE and MSI-H) has gradually attracted attention. In this review, we focus on the expression and molecular basis of the main biomarkers in the immunotherapy of EC firstly, as well as their clinical application significance and limitations. Blocking tumor immune checkpoints is one of the most effective strategies for cancer treatment in recent years, and has now become the focus in the field of tumor research and treatment. We summarized clinical date of planned and ongoing clinical trials and introduced other common immunotherapy methods in EC, such as cancer vaccine and ACT. Hormone aberrations, metabolic syndrome (MetS) and p53 mutant and that affect the immunotherapy of endometrial cancer will also be discussed in this review.
Collapse
Affiliation(s)
- Wenyu Cao
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Xinyue Ma
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Jean Victoria Fischer
- Department of Pathology, Northwestern Medicine, Gynecologic Pathology Fellow, Chicago, Illinois, USA
| | - Chenggong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
26
|
Arang N, Gutkind JS. G Protein-Coupled receptors and heterotrimeric G proteins as cancer drivers. FEBS Lett 2021; 594:4201-4232. [PMID: 33270228 DOI: 10.1002/1873-3468.14017] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/09/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) and heterotrimeric G proteins play central roles in a diverse array of cellular processes. As such, dysregulation of GPCRs and their coupled heterotrimeric G proteins can dramatically alter the signalling landscape and functional state of a cell. Consistent with their fundamental physiological functions, GPCRs and their effector heterotrimeric G proteins are implicated in some of the most prevalent human diseases, including a complex disease such as cancer that causes significant morbidity and mortality worldwide. GPCR/G protein-mediated signalling impacts oncogenesis at multiple levels by regulating tumour angiogenesis, immune evasion, metastasis, and drug resistance. Here, we summarize the growing body of research on GPCRs and their effector heterotrimeric G proteins as drivers of cancer initiation and progression, and as emerging antitumoural therapeutic targets.
Collapse
Affiliation(s)
- Nadia Arang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - J Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Deb J, Lakshman TR, Ghosh I, Jana SS, Paine TK. Mechanistic studies of in vitro anti-proliferative and anti-inflammatory activities of the Zn(ii)-NSAID complexes of 1,10-phenanthroline-5,6-dione in MDA-MB-231 cells. Dalton Trans 2021; 49:11375-11384. [PMID: 32766641 DOI: 10.1039/d0dt01721c] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Two zinc(ii)-NSAID complexes [(phendione)ZnII(NPR)2(H2O)2] (1) and [(phendione)ZnII(MFN)2] (2) (HNPR = naproxen and HMFN = mefenamic acid) of 1,10-phenanthroline-5,6-dione (phendione) were isolated and characterized to evaluate their potential as anti-cancer agents. Each of the complexes contains two equivalents of NSAID per zinc(ii)-phendione unit. The complexes are stable in solution under cell culture conditions. Cytotoxic assay on the human breast cancer cell line (MDA-MB-231) reveals that the anti-proliferative activity of phendione is retained in both the complexes. The anti-inflammatory properties of NSAIDs are also preserved in the metal complexes as evident from the PGE2 assay. Both 1 and 2 exhibit selective COX-1 inhibition at a low concentration. Furthermore, the zinc(ii)-naproxen complex (1) disrupts the intercellular bridges displaying in vitro delay in cellular migration and down-regulation of EMT-related genes. The mechanistic studies indicate that the ternary complexes are more active compared to cisplatin and have the potential to overcome cisplatin resistance in MDA MB 231 cells. These findings demonstrate that the zinc(ii)-NSAID complexes are worthy of further in vivo studies for their promising anti-tumor potential.
Collapse
Affiliation(s)
- Jolly Deb
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Triloke Ranjan Lakshman
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Ivy Ghosh
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Siddhartha Sankar Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Tapan Kanti Paine
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
28
|
Hernández-Valdés D, Sadeghi S. Electrochemical Radiofluorination of Small Molecules: New Advances. CHEM REC 2021; 21:2397-2410. [PMID: 34010479 DOI: 10.1002/tcr.202100086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Indexed: 12/14/2022]
Abstract
The development of new 18 F-based radiopharmaceuticals constantly demands innovations in the search for new radiofluorination methods. [18 F]fluoride is the simplest and most convenient chemical form of the isotope for the synthesis of 18 F-based radiopharmaceuticals. The ease of production and handling, as well as the possibility of obtaining high molar activities, makes it the preferred choice for radiofluorination. However, the use of [18 F]fluoride in late-stage radiofluorination comes with challenges, especially for the radiolabeling of electron-rich molecules where SN 2 and SN Ar reactions are not suitable. New developments in fluorination chemistry have been extensively studied to overcome these difficulties. Selective electrochemical oxidation of precursors, using a controlled potential, is one method to create reactive intermediates and overcome the activation energy required for nucleophilic fluorination of electron-rich moieties. This method has been used for years in cold fluorination of organic molecules and more recently has been adapted as an alternative to traditional radiofluorination methods. Although relatively young, this field stands out as a promising route for the synthesis of new PET probes as well as fluorinated pharmaceuticals. This review focuses on recent advances in electrochemical radiofluorination as an alternative for the late-stage radiolabeling of organic molecules.
Collapse
Affiliation(s)
- Daniel Hernández-Valdés
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 L8, Canada
| | - Saman Sadeghi
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 L8, Canada.,Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Yang S, An J, Park S, Lee J, Chae H, Lee K, Song W, Youn H. Enhanced expression of cyclooxygenase-2 related multi-drug resistance gene in melanoma and osteosarcoma cell lines by TSG-6 secreted from canine adipose-derived mesenchymal stem/stromal cells. Vet Med Sci 2021; 7:968-978. [PMID: 33570264 PMCID: PMC8136926 DOI: 10.1002/vms3.442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/09/2020] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multiple drug resistance (MDR) of cancer cells is the main cause of intrinsic or acquired desensitization to chemotherapy in many cancers. A number of studies have found high expression of COX-2 to be a factor for expression of MDR gene in several cancer. Furthermore, adipose tissue derived mesenchymal stem/stromal cells (ADSC) have been found to increase cyclo-oxygenase-2 (COX-2) expression in some tumour cells. The mechanism for this, however, is not yet clear and needs further study. OBJECTIVE The purpose of this study was to determine whether tumour necrosis factor-alpha stimulated gene/protein 6 (TSG-6) secreted from ADSCs is associated with an increase in MDR genes by inducing COX-2 gene expression in melanoma and osteosarcoma cell lines. METHODS ADSCs were transfected with TSG-6 siRNA or Control RNA respected, and cancer cell line were transfected with COX-2 siRNA or Control RNA respected. Using trans well coculture system, the interactions of ADSCs with tumour cells were investigated. RESULTS Increased COX-2 expression was observed in cancer cell co-cultured with ADSCs. Additionally, we identified that COX-2 expression was related to drug resistance genes (P-glycoprotein, multidrug resistance-associated protein). Transfecting canine ADSCs with small interfering RNA, TSG-6 secreted from ADSCs was found to be a major factor in the regulation of COX-2 expression and drug resistance genes in osteosarcoma and melanoma cell lines. CONCLUSION TSG-6 mediated COX-2 up-regulation is a possible mechanism of chemoresistance development induced by ADSCs. These findings provide better understanding about the mechanism associated with ADSC-induced chemoresistance in cancer.
Collapse
Affiliation(s)
- Se‐Jin Yang
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Ju‐Hyun An
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Su‐Min Park
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Jeong‐Hwa Lee
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Hyung‐Kyu Chae
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Kyung‐Mi Lee
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Woo‐Jin Song
- Department of Veterinary Internal MedicineCollege of Veterinary MedicineJeju National UniversityJeju‐SiKorea
| | - Hwa‐Young Youn
- Laboratory of Veterinary Internal MedicineDepartment of Veterinary Clinical ScienceCollege of Veterinary MedicineSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
30
|
Guo S, Wu X, Zheng J, Smith SA, Dong P, Xiao H. Identification of 4'-Demethyltangeretin as a Major Urinary Metabolite of Tangeretin in Mice and Its Anti-inflammatory Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:4381-4391. [PMID: 33787243 DOI: 10.1021/acs.jafc.0c06334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The present study showed that oral administration of tangeretin (TAN) in mice resulted in the production of 4'-demethyltangeretin (4DT) as a major urinary metabolite. The anti-inflammatory efficacy of TAN and 4DT was determined in RAW 264.7 macrophages stimulated by lipopolysaccharides (LPS). 4DT produced considerably stronger inhibition on the overproduction of prostaglandin E2 and nitric oxide than TAN did at the same concentrations. Western blot and quantitative polymerase chain reaction analyses indicated that 4DT exerted more potent suppressive activity on the over-expression of interleukin-1β, inducible nitric oxide synthase, and cyclooxygenase-2 than TAN. Treatments with TAN and 4DT diminished LPS-stimulated nuclear factor κB (NFκB) translocation via suppressing the degradation of inhibitor κB (IκBα). Furthermore, both compounds attenuated mitogen-activated protein kinases (MAPKs) and Akt signaling upregulated by LPS. Overall, our findings showed that TAN and 4DT inhibited the LPS-stimulated inflammatory response in macrophages by suppressing Akt/MAPKs/NFκB proinflammatory pathways, while 4DT showed more potent activity than TAN, its parent compound.
Collapse
Affiliation(s)
- Shanshan Guo
- Department of Food Science, University of Massachusetts Amherst, Amherst 01003, Massachusetts, United States
- Department of Food Science and Nutrition, University of Jinan, Jinan 250022, Shandong, China
| | - Xian Wu
- Department of Food Science, University of Massachusetts Amherst, Amherst 01003, Massachusetts, United States
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford 45056, Ohio, United States
| | - Jinkai Zheng
- Department of Food Science, University of Massachusetts Amherst, Amherst 01003, Massachusetts, United States
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Sarah A Smith
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford 45056, Ohio, United States
| | - Ping Dong
- Department of Food Science, University of Massachusetts Amherst, Amherst 01003, Massachusetts, United States
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, Shandong, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst, Amherst 01003, Massachusetts, United States
| |
Collapse
|
31
|
Moussa N, Hassan A, Gharaghani S. Pharmacophore model, docking, QSAR, and molecular dynamics simulation studies of substituted cyclic imides and herbal medicines as COX-2 inhibitors. Heliyon 2021; 7:e06605. [PMID: 33889764 PMCID: PMC8047494 DOI: 10.1016/j.heliyon.2021.e06605] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/22/2021] [Accepted: 03/24/2021] [Indexed: 01/09/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) enzyme inhibitors have not eliminated the necessity for developed drugs not only in the nonsteroidal anti-inflammatory drug (NSAIDs) area, but also in other therapeutic applications including prevention of cancer and Alzheimer's disease. A series of novel substituted cyclic imides have been reported as selective COX-2 inhibitors. To understand the structural features responsible for their activity, a 3D validated pharmacophore and quantitative structure−activity relationship (QSAR) model have been developed. The values of enrichment factor (EF), goodness of hit score (GH), area under the ROC curve (AUC), sensitivity, and specificity refer to the good ability of the pharmacophore model to identify active compounds. Multiple linear regression (MLR) produced statistically significant QSAR model with (R2training = 0.763, R2test = 0.96) and predictability (Q2training = 0.66, Q2test = 0.84). Then, using the pharmacophore and QSAR models, eight authenticated botanicals in two herbal medicines and the ZINC compounds database, were virtually screened for ligands to COX-2. The retrieved hits which also obey lipinski's rule of five (RO5) were docked in the COX-2 3D structure to investigate their binding mode and affinity. Finally, based on the docking results, nine molecules were prioritized as promising hits that could be used as leads to discover novel COX-2 inhibitors. COX-2 inhibition of most of these hits has not been reported previously. Ten-nanosecond molecular dynamics simulation (10-ns MD) was performed on the initial structure COX-2 complex with ZINC000113253375 and ZINC000043170560 resulted from the docking. Our utilization of the 3D pharmacophore model, QSAR, molecular docking, and molecular dynamics simulation trials can be a potent strategy to successfully predict activity, efficiently design drugs, and screen large numbers of new compounds as active drug candidates.
Collapse
Affiliation(s)
- Nathalie Moussa
- Department of Pharmaceutical Chemistry and Quality Control of Medicaments, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - Ahmad Hassan
- Department of Pharmaceutical Chemistry and Quality Control of Medicaments, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
32
|
Prostaglandin E2 Receptor 4 (EP4) as a Therapeutic Target to Impede Breast Cancer-Associated Angiogenesis and Lymphangiogenesis. Cancers (Basel) 2021; 13:cancers13050942. [PMID: 33668160 PMCID: PMC7956318 DOI: 10.3390/cancers13050942] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
The formation of new blood (angiogenesis) and lymphatic (lymphangiogenesis) vessels are major events associated with most epithelial malignancies, including breast cancer. Angiogenesis is essential for cancer cell survival. Lymphangiogenesis is critical in maintaining tumoral interstitial fluid balance and importing tumor-facilitatory immune cells. Both vascular routes also serve as conduits for cancer metastasis. Intratumoral hypoxia promotes both events by stimulating multiple angiogenic/lymphangiogenic growth factors. Studies on tumor-associated lymphangiogenesis and its exploitation for therapy have received less attention from the research community than those on angiogenesis. Inflammation is a key mediator of both processes, hijacked by many cancers by the aberrant expression of the inflammation-associated enzyme cyclo-oxygenase (COX)-2. In this review, we focus on breast cancer and showed that COX-2 is a major promoter of both events, primarily resulting from the activation of prostaglandin (PG) E receptor EP4 on tumor cells, tumor-infiltrating immune cells, and endothelial cells; and the induction of oncogenic microRNAs. The COX-2/EP4 pathway also promotes additional events in breast cancer progression, such as cancer cell migration, invasion, and the stimulation of stem-like cells. Based on a combination of studies using multiple breast cancer models, we show that EP4 antagonists hold a major promise in breast cancer therapy in combination with other modalities including immune check-point inhibitors.
Collapse
|
33
|
He L, Chang H, Qi Y, Zhang B, Shao Q. ceRNA Networks: The Backbone Role in Neoadjuvant Chemoradiotherapy Resistance/Sensitivity of Locally Advanced Rectal Cancer. Technol Cancer Res Treat 2021; 20:15330338211062313. [PMID: 34908512 PMCID: PMC8689620 DOI: 10.1177/15330338211062313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022] Open
Abstract
Approximately 40% of rectal cancers during initial diagnosis are identified as locally advanced rectal cancers (LARCs), for which the standardized treatment scenario is total mesorectal excision following neoadjuvant chemoradiotherapy (nCRT). nCRT can lead to discernible reductions in local relapse rate and distant metastasis rate in LARC patients, in whom previously inoperable tumors may potentially be surgically removed. However, only 4% to 20% cases can attain pathological complete response, and the remaining patients who are unresponsive to nCRT have to suffer from the side effects plus toxicities and may encounter poor survival outcomes due to the late surgical intervention. As such, employing potential biomarkers to differentiate responders from nonresponders before nCRT implementation appears to be the overarching goal. Well-defined competing endogenous RNA (ceRNA) networks include long noncoding RNA (lncRNA)-microRNA (miRNA)-mRNA and circRNA-miRNA-mRNA networks. As ceRNAs, lncRNAs, and circRNAs sponge miRNAs to indirectly suppress miRNAs downstream of oncogenic mRNAs or tumor-suppressive mRNAs. The abnormal expression of mRNAs regulates the nCRT-induced DNA damage repair process through pluralistic carcinogenic signaling pathways, thereby bringing about alterations in the nCRT resistance/sensitivity of tumors. Moreover, many molecular mechanisms relevant to cell proliferation, metastasis, or apoptosis of cancers (eg, epithelial-mesenchymal transition and caspase-9-caspase-3 pathway) are influenced by ceRNA networks. Herein, we reviewed a large group of abnormally expressed mRNAs and noncoding RNAs that are associated with nCRT resistance/sensitivity in LARC patients and ultimately pinpointed the backbone role of ceRNA networks in the molecular mechanisms of nCRT resistance/sensitivity.
Collapse
Affiliation(s)
- Lin He
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Hao Chang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Yuhong Qi
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Bing Zhang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Qiuju Shao
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| |
Collapse
|
34
|
Non-Steroidal Anti-Inflammatory Drugs Increase Cisplatin, Paclitaxel, and Doxorubicin Efficacy against Human Cervix Cancer Cells. Pharmaceuticals (Basel) 2020; 13:ph13120463. [PMID: 33333716 PMCID: PMC7765098 DOI: 10.3390/ph13120463] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022] Open
Abstract
This study shows that the non-steroidal anti-inflammatory drug (NSAID) celecoxib and its non-cyclooxygenase-2 (COX2) analogue dimethylcelecoxib (DMC) exert a potent inhibitory effect on the growth of human cervix HeLa multi-cellular tumor spheroids (MCTS) when added either at the beginning (“preventive protocol”; IC50 = 1 ± 0.3 nM for celecoxib and 10 ± 2 nM for DMC) or after spheroid formation (“curative protocol”; IC50 = 7.5 ± 2 µM for celecoxib and 32 ± 10 µM for DMC). These NSAID IC50 values were significantly lower than those attained in bidimensional HeLa cells (IC50 = 55 ± 9 µM celecoxib and 48 ± 2 µM DMC) and bidimensional non-cancer cell cultures (3T3 fibroblasts and MCF-10A mammary gland cells with IC50 from 69 to >100 µM, after 24 h). The copper-based drug casiopeina II-gly showed similar potency against HeLa MCTS. Synergism analysis showed that celecoxib, DMC, and casiopeinaII-gly at sub-IC50 doses increased the potency of cisplatin, paclitaxel, and doxorubicin to hinder HeLa cell proliferation through a significant abolishment of oxidative phosphorylation in bidimensional cultures, with no apparent effect on non-cancer cells (therapeutic index >3.6). Similar results were attained with bidimensional human cervix cancer SiHa and human glioblastoma U373 cell cultures. In HeLa MCTS, celecoxib, DMC and casiopeina II-gly increased cisplatin toxicity by 41–85%. These observations indicated that celecoxib and DMC used as adjuvant therapy in combination with canonical anti-cancer drugs may provide more effective alternatives for cancer treatment.
Collapse
|
35
|
Meireson A, Devos M, Brochez L. IDO Expression in Cancer: Different Compartment, Different Functionality? Front Immunol 2020; 11:531491. [PMID: 33072086 PMCID: PMC7541907 DOI: 10.3389/fimmu.2020.531491] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a cytosolic haem-containing enzyme involved in the degradation of tryptophan to kynurenine. Although initially thought to be solely implicated in the modulation of innate immune responses during infection, subsequent discoveries demonstrated IDO1 as a mechanism of acquired immune tolerance. In cancer, IDO1 expression/activity has been observed in tumor cells as well as in the tumor-surrounding stroma, which is composed of endothelial cells, immune cells, fibroblasts, and mesenchymal cells. IDO1 expression/activity has also been reported in the peripheral blood. This manuscript reviews available data on IDO1 expression, mechanisms of its induction, and its function in cancer for each of these compartments. In-depth study of the biological function of IDO1 according to the expressing (tumor) cell can help to understand if and when IDO1 inhibition can play a role in cancer therapy.
Collapse
Affiliation(s)
- Annabel Meireson
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Michael Devos
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Lieve Brochez
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
36
|
Liao X, Gao Y, Liu J, Tao L, Xie J, Gu Y, Liu T, Wang D, Xie D, Mo S. Combination of Tanshinone IIA and Cisplatin Inhibits Esophageal Cancer by Downregulating NF-κB/COX-2/VEGF Pathway. Front Oncol 2020; 10:1756. [PMID: 33014864 PMCID: PMC7511800 DOI: 10.3389/fonc.2020.01756] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/05/2020] [Indexed: 11/13/2022] Open
Abstract
Cisplatin (DDP) represents one of the common drugs used for esophageal squamous cell carcinoma (ESCC), but side effects associated with DDP and drug resistance lead to the failure of treatment. This study aimed to understand whether tanshinone IIA (tan IIA) and DDP could generate a synergistic antitumor effect on ESCC cells. Tan IIA and DDP are demonstrated to restrain ESCC cell proliferation in a time- and dose-dependent mode. Tan IIA and DDP at a ratio of 2:1 present a synergistic effect on ESCC cells. The combination suppresses cell migration and invasion abilities, arrests the cell cycle, and causes apoptosis in HK and K180 cells. Molecular docking indicates that tan IIA and DDP could be docked into active sites with the tested proteins. In all treated groups, the expression levels of E-cadherin, β-catenin, Bax, cleaved caspase-9, P21, P27, and c-Fos were upregulated, and the expression levels of fibronectin, vimentin, Bcl-2, cyclin D1, p-Akt, p-ERK, p-JNK, P38, COX-2, VEGF, IL-6, NF-κB, and c-Jun proteins were downregulated. Among these, the combination induced the most significant difference. Our results suggest that tan IIA could be a novel treatment for combination therapy for ESCC.
Collapse
Affiliation(s)
- Xiaozhong Liao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Gao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanting Tao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Xie
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yueyu Gu
- The Second Clinical College, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Taoli Liu
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dongmei Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Suilin Mo
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
May S, Parry C, Parry L. Berry chemoprevention: Do berries decrease the window of opportunity for tumorigenesis. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.32] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Stephanie May
- European Cancer Stem Cell Research Institute School of Biosciences Cardiff University Cardiff UK
| | - Connor Parry
- European Cancer Stem Cell Research Institute School of Biosciences Cardiff University Cardiff UK
| | - Lee Parry
- European Cancer Stem Cell Research Institute School of Biosciences Cardiff University Cardiff UK
| |
Collapse
|
38
|
Fox EF, Lamb MC, Mellentine SQ, Tootle TL. Prostaglandins regulate invasive, collective border cell migration. Mol Biol Cell 2020; 31:1584-1594. [PMID: 32432969 PMCID: PMC7521797 DOI: 10.1091/mbc.e19-10-0578] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
While prostaglandins (PGs), short-range lipid signals, regulate single cell migration, their roles in collective migration remain unclear. To address this, we use Drosophila border cell migration, an invasive, collective migration that occurs during Stage 9 of oogenesis. Pxt is the Drosophila cyclooxygenase-like enzyme responsible for PG synthesis. Loss of Pxt results in both delayed border cell migration and elongated clusters, whereas somatic Pxt knockdown causes delayed migration and compacted clusters. These findings suggest PGs act in both the border cells and nurse cells, the substrate on which the border cells migrate. As PGs regulate the actin bundler Fascin, and Fascin is required for on-time migration, we assessed whether PGs regulate Fascin to promote border cell migration. Coreduction of Pxt and Fascin results in delayed migration and elongated clusters. The latter may be due to altered cell adhesion, as loss of Pxt or Fascin, or coreduction of both, decreases integrin levels on the border cell membranes. Conversely, integrin localization is unaffected by somatic knockdown of Pxt. Together these data lead to the model that PG signaling controls Fascin in the border cells to promote migration and in the nurse cells to maintain cluster cohesion.
Collapse
Affiliation(s)
- Emily F Fox
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Maureen C Lamb
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Samuel Q Mellentine
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Tina L Tootle
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
39
|
Santoro A, Bufo P, Russo G, Cagiano S, Papagerakis S, Bucci P, Aquino G, Longo F, Feola A, Giordano A, Di Carlo A, Di Domenico M, Pannone G. Expression and clinical implication of cyclooxygenase-2 and E-cadherin in oral squamous cell carcinomas. Cancer Biol Ther 2020. [PMID: 26218314 PMCID: PMC7537792 DOI: 10.1080/15384047.2015.1071741] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epithelial-Mesenchymal Transition (EMT) and angiogenesis are crucial events for development of aggressive and often fatal Oral Squamous Cell Carcinomas (OSCCs). Both promote cancer progression and metastasis development, but while the former induces the loss of E-cadherin expression and, hence cadherin switching; the latter produces hematic blood vessel neo-formation and contribute to OSCC cell growth, tumor mass development, and dissemination. Cyclooxygenase-2 (COX-2) has an important role, not only in angiogenic mechanisms, but also in favoring cancer invasion. Indeed it decreases the expression of E-cadherin and leads to phenotypic changes in epithelial cells (EMT) enhancing their carcinogenic potential. Our aim is to evaluate the interplay between E-cadherin cytoplasmic delocalization, COX-2 up-regulation and COX-2 induced neo-angiogenesis in 120 cases of OSCC. We have analyzed the distribution and the number of neo-formed endothelial buds surrounding infiltrating cells that express COX-2, as well as the neo-formed vessels in chronic inflammatory infiltrate, which surround the tumor. A double immunostaining method was employed in order to verify co-localization of endothelial cell marker (CD34) and COX-2. IHC has also been used to assess E-cadherin expression. Our data demonstrate that the OSCC cells, which lose membranous E-cadherin staining, acquiring a cytoplasmic delocalization, overexpress COX-2. Moreover, we find a new CD34+ vessel formation (sprouting angiogenesis). Only basaloid type of OSCC showes low level of COX-2 expression together with very low level of neo-angiogenesis and consequent tumor necrosis. The well-known anti-metastatic effect of certain COX-2 inhibitors suggests that these molecules might have clinical utility in the management of advanced cancers.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Anatomic Pathology, 'Giovanni Paolo II' Foundation-UCSC , Campobasso, Italy
| | - Pantaleo Bufo
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Giuseppe Russo
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Simona Cagiano
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| | - Silvana Papagerakis
- Laboratory of Head and Neck Cancer Invasion and Metastasis, Department of Otolaryngology, Head and Neck Oncology, University of Michigan, Medical School , Ann Arbor, MI, USA
| | - Paolo Bucci
- Department of Neuroscience, Reproductive and Odontostomatologies Science, University of Naples 'Federico II' , Naples, Italy
| | - Gabriella Aquino
- Section of Pathological Anatomy, National Cancer Institute 'G. Pascale Foundation' , Naples, Italy
| | - Francesco Longo
- Head and Neck Medical Oncology Unit, National Cancer Institute 'G. Pascale Foundation' , Naples, Italy
| | - Antonia Feola
- Department of Biology, University of Naples , Naples, Italy.,Department of Biochemistry, Biophisic and General Pathology Second University of Naples , Naples, Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena , Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia, PA, USA
| | - Angelina Di Carlo
- Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome , Rome, Italy
| | - Marina Di Domenico
- Department of Biochemistry, Biophisic and General Pathology Second University of Naples , Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia, PA, USA
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, Institute of Pathological Anatomy, University of Foggia , Foggia, Italy
| |
Collapse
|
40
|
Small Ones to Fight a Big Problem-Intervention of Cancer Metastasis by Small Molecules. Cancers (Basel) 2020; 12:cancers12061454. [PMID: 32503267 PMCID: PMC7352875 DOI: 10.3390/cancers12061454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Metastasis represents the most lethal attribute of cancer and critically limits successful therapies in many tumor entities. The clinical need is defined by the fact that all cancer patients, who have or who will develop distant metastasis, will experience shorter survival. Thus, the ultimate goal in cancer therapy is the restriction of solid cancer metastasis by novel molecularly targeted small molecule based therapies. Biomarkers identifying cancer patients at high risk for metastasis and simultaneously acting as key drivers for metastasis are extremely desired. Clinical interventions targeting these key molecules will result in high efficiency in metastasis intervention. In result of this, personalized tailored interventions for restriction and prevention of cancer progression and metastasis will improve patient survival. This review defines crucial biological steps of the metastatic cascade, such as cell dissemination, migration and invasion as well as the action of metastasis suppressors. Targeting these biological steps with tailored therapeutic strategies of intervention or even prevention of metastasis using a wide range of small molecules will be discussed.
Collapse
|
41
|
Grbčić P, Sedić M. Sphingosine 1-Phosphate Signaling and Metabolism in Chemoprevention and Chemoresistance in Colon Cancer. Molecules 2020; 25:E2436. [PMID: 32456134 PMCID: PMC7287727 DOI: 10.3390/molecules25102436] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal carcinoma (CRC) is the leading cause of cancer-related deaths worldwide. Despite advances in prevention and treatment modalities for CRC, rapidly developing resistance to chemotherapy limits its effectiveness. For that reason, it is important to better understand the mechanisms that undergird the process of chemoresistance to enable design of novel anticancer agents specifically targeting malignant properties of cancer cells. Over recent decades, bioactive sphingolipid species have come under the spotlight for their recognized role in cancer development and progression, and the evidence has surfaced to support their role as regulators of anti-cancer drug resistance. Colon cancer is characterized by a shift in sphingolipid balance that favors the production and accumulation of oncogenic species such as sphingosine 1-phosphate (S1P). S1P is known to govern the processes that facilitate cancer cell growth and progression including proliferation, survival, migration, invasion and inflammation. In this review paper, we will give a comprehensive overview of current literature findings on the molecular mechanisms by which S1P turnover, transport and signaling via receptor-dependent and independent pathways shape colon cancer cell behavior and influence treatment outcome in colon cancer. Combining available modulators of S1P metabolism and signaling with standard chemotherapy drugs could provide a rational approach to achieve enhanced therapeutic response, diminish chemoresistance development and improve the survival outcome in CRC patients.
Collapse
Affiliation(s)
| | - Mirela Sedić
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia;
| |
Collapse
|
42
|
Kosmas K, Mitropoulou G, Stamoulas M, Marouga A. Expression of cyclooxygenase-2 in imprint smears of endometrial carcinoma. Diagn Cytopathol 2020; 48:1086-1092. [PMID: 32433831 DOI: 10.1002/dc.24481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/16/2020] [Accepted: 05/08/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Worldwide, endometrial carcinoma is one of the most frequently diagnosed cancers among women and a considerable cause of death. The aims of this study were to determine the expression of cyclooxygenase-2 (COX-2) in endometrial adenocarcinoma in imprint smears as an alternative technique and to correlate the results with clinicopathologic parameters of primary untreated endometrial cancer patients. METHODS One hundred twenty-six patients with endometrial carcinoma were evaluated with samples freshly resected after a total abdominal hysterectomy during a 29-month period. The cytologic imprint smears were obtained by touching the cut surface of cancer tissues and the expression of COX-2 was assessed by immunocytochemistry. RESULTS The positive expression of COX-2 in malignant cells, was accompanied by morphologic features of more aggressiveness (pathogenetic type II, advanced clinical stage, mainly high grade, deep myometrial involvement >1/2) tumors and the affected from the disease lymph nodes cases showed higher positivity (41.2%) than the non-affected (13.4%). CONCLUSION Immunocytochemical findings from COX-2 stain in cancer cells could be a predictor of prognosis in most cases in endometrial cytology with imprint smears. Furthermore, positive expression of COX-2 in cancer cells was related to morphologic features of more aggressiveness tumors.
Collapse
Affiliation(s)
- Konstantinos Kosmas
- Department of Cytopathology, General Chest Diseases Hospital of Athens "SOTIRIA", Athens, Greece
| | - Georgia Mitropoulou
- Department of Pathology, "Agia Sofia" Children's Hospital of Athens, Athens, Greece
| | - Marios Stamoulas
- Department of Obstetrics and Gynaecology, Chesterfield Royal Hospital, Derbyshire, UK
| | - Anna Marouga
- Department of Cytopathology, Athens University, Medical School, "Attikon" University Hospital, Athens, Greece
| |
Collapse
|
43
|
Hidalgo-Estévez AM, Stamatakis K, Jiménez-Martínez M, López-Pérez R, Fresno M. Cyclooxygenase 2-Regulated Genes an Alternative Avenue to the Development of New Therapeutic Drugs for Colorectal Cancer. Front Pharmacol 2020; 11:533. [PMID: 32410997 PMCID: PMC7201075 DOI: 10.3389/fphar.2020.00533] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common and recurrent types of cancer, with high mortality rates. Several clinical trials and meta-analyses have determined that the use of pharmacological inhibitors of cyclooxygenase 2 (COX-2), the enzyme that catalyses the rate-limiting step in the synthesis of prostaglandins (PG) from arachidonic acid, can reduce the incidence of CRC as well as the risk of recurrence of this disease, when used together with commonly used chemotherapeutic agents. These observations suggest that inhibition of COX-2 may be useful in the treatment of CRC, although the current drugs targeting COX-2 are not widely used since they increase the risk of health complications. To overcome this difficulty, a possibility is to identify genes regulated by COX-2 activity that could give an advantage to the cells to form tumors and/or metastasize. The modulation of those genes as effectors of COX-2 may cancel the beneficial effects of COX-2 in tumor transformation and metastasis. A review of the available databases and literature and our own data have identified some interesting molecules induced by prostaglandins or COX-2 that have been also described to play a role in colon cancer, being thus potential pharmacological targets in colon cancer. Among those mPGES-1, DUSP4, and 10, Programmed cell death 4, Trop2, and many from the TGFβ and p53 pathways have been identified as genes upregulated in response to COX-2 overexpression or PGs in colon carcinoma lines and overexpressed in colon tumor tissue. Here, we review the available evidence of the potential roles of those molecules in colon cancer in the context of PG/COX signaling pathways that could be critical mediators of some of the tumor growth and metastasis advantage induced by COX-2. At the end, this may allow defining new therapeutic targets/drugs against CRC that could act specifically against tumor cells and would be effective in the prevention and treatment of CRC, lacking the unwanted side effects of COX-2 pharmacological inhibitors, providing alternative approaches in colon cancer.
Collapse
Affiliation(s)
| | - Konstantinos Stamatakis
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| | - Marta Jiménez-Martínez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo López-Pérez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| |
Collapse
|
44
|
Tiwari A, Saraf S, Jain A, Panda PK, Verma A, Jain SK. Basics to advances in nanotherapy of colorectal cancer. Drug Deliv Transl Res 2020; 10:319-338. [PMID: 31701486 DOI: 10.1007/s13346-019-00680-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer existing across the globe. It begins with the formation of polyps leading to the development of metastasis, especially in advanced stage patients, who necessitate intensive chemotherapy that usually results in a poor response and high morbidity owing to multidrug resistance and severe untoward effects to the non-cancerous cells. Advancements in the targeted drug delivery permit the targeting of tumor cells without affecting the non-tumor cells. Various nanocarriers such as liposomes, polymeric nanoparticles, carbon nanotubes, micelles, and nanogels, etc. are being developed and explored for effective delivery of cytotoxic drugs to the target site thereby enhancing the drug distribution and bioavailability, simultaneously subduing the side effects. Moreover, immunotherapy for CRC is being explored for last few decades. Few clinical trials have even potentially benefited patients suffering from CRC, still immunotherapy persists merely an experimental alternative. Assessment of the ongoing and completed trials is to be warranted for effective treatment of CRC. Scientists are paying efforts to develop novel carrier systems that may enhance the targeting potential of low therapeutic index chemo- and immune-therapeutics. Several preclinical studies have revealed the superior efficacy of nanotherapy in CRC as compared to conventional approaches. Clinical trials are being recruited to ascertain the safety and efficacy of CRC therapies. The present review discourses in a nutshell the molecular interventions including the genetics, signaling pathways involved in CRC, and advances in various strategies explored for the treatment of CRC with a special emphasis on nanocarriers based drug targeting.
Collapse
Affiliation(s)
- Ankita Tiwari
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India
| | - Shivani Saraf
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India
| | - Ankit Jain
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India
- Institute of Pharmaceutical Research, GLA University, NH-2, Mathura-Delhi Road, Mathura, 281 406 (U.P.), India
| | - Pritish K Panda
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India
| | - Amit Verma
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India
| | - Sanjay K Jain
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar, 470 003 (M.P.), India.
| |
Collapse
|
45
|
Venkatachalam K, Vinayagam R, Arokia Vijaya Anand M, Isa NM, Ponnaiyan R. Biochemical and molecular aspects of 1,2-dimethylhydrazine (DMH)-induced colon carcinogenesis: a review. Toxicol Res (Camb) 2020; 9:2-18. [PMID: 32440334 DOI: 10.1093/toxres/tfaa004] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/20/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
1,2-dimethylhydrazine (DMH) is a member in the class of hydrazines, strong DNA alkylating agent, naturally present in cycads. DMH is widely used as a carcinogen to induce colon cancer in animal models. Exploration of DMH-induced colon carcinogenesis in rodent models provides the knowledge to perceive the biochemical, molecular, and histological mechanisms of different stages of colon carcinogenesis. The procarcinogen DMH, after a series of metabolic reactions, finally reaches the colon, there produces the ultimate carcinogen and reactive oxygen species (ROS), which further alkylate the DNA and initiate the development of colon carcinogenesis. The preneolpastic lesions and histopathological observations of DMH-induced colon tumors may provide typical understanding about the disease in rodents and humans. In addition, this review discusses about the action of biotransformation and antioxidant enzymes involved in DMH intoxication. This understanding is essential to accurately identify and interpret alterations that occur in the colonic mucosa when evaluating natural or pharmacological compounds in DMH-induced animal colon carcinogenesis.
Collapse
Affiliation(s)
- Karthikkumar Venkatachalam
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, UAE University, Al Ain-17666, United Arab Emirates
| | - Ramachandran Vinayagam
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore, Tamilnadu 632 115, India
| | | | - Nurulfiza Mat Isa
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, 43400 Seri Kembangan, Selangor, Malaysia
| | - Rajasekar Ponnaiyan
- Department of Zoology, Jamal Mohamed College, Tiruchirappalli, Tamil Nadu 620020, India
| |
Collapse
|
46
|
MicroRNA binding site polymorphism in inflammatory genes associated with colorectal cancer: literature review and bioinformatics analysis. Cancer Gene Ther 2020; 27:739-753. [PMID: 32203060 DOI: 10.1038/s41417-020-0172-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/21/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Inflammation, among environmental risk factors, is one of the most important contributors to colorectal cancer (CRC) development. In this way, studies revealed that the incidence of CRC in inflammatory bowel disease patients is up to 60% higher than the general population. MicroRNAs (miRNAs), small noncoding RNA molecules, have attracted excessive attention due to their fundamental role in various aspects of cellular biology, such as inflammation by binding to the 3'-untranslated regions (3'-UTR) of pro and anti-inflammatory genes. Based on multiple previous studies, SNPs at 3'-UTR can affect miRNA recognition elements by changing the thermodynamic features and secondary structure. This effect can be categorized, based on the number of changes, into four groups, including break, decrease, create, and enhance. In this paper, we will focus on functional variants in miRNA binding sites in inflammatory genes, which can modulate the risk of CRC by both investigating previous studies, regarding miRSNPs in inflammatory genes associated with CRC and recruiting in silico prediction algorithms to report putative miRSNPs in 176 inflammatory genes. In our analysis, we achieved 110 miRSNPs in 3'-UTR of 67 genes that seem good targets for future researches.
Collapse
|
47
|
Xiang L, Wang W, Zhou Z, Lv M, Tao L, Ni T, Deng J, Masatara S, Liu Y, Zhou Y. COX-2 promotes metastasis and predicts prognosis in gastric cancer via regulating mTOR. Biomark Med 2020; 14:421-432. [PMID: 32175764 DOI: 10.2217/bmm-2019-0357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: Gastric cancer (GC) is one of the most common malignant tumors in the world. It is important to find accurate and reliable biomarkers in order to decrease whole morbidity and mortality. Results: We examined the expression of COX-2 and mTOR on GC tissue microarrays by immunohistochemistry. Multivariate COX regression analysis showed that the expression of COX-2 or mTOR was an independent factor in the prognosis of GC patients. In addition, COX-2 and mTOR have a potentially synergistic effect on predicting the prognosis of GC. Conclusion: The combined expression of COX-2 and mTOR could serve as efficient prognostic indicators and COX-2 could suppress GC metastasis via regulating mTOR.
Collapse
Affiliation(s)
- Liangliang Xiang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Weimin Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China.,Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, 214200, PR China
| | - Zhen Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Mengying Lv
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Li Tao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Tengyang Ni
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Jianliang Deng
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, 214200, PR China
| | - Sunagawa Masatara
- Department of Physiology, School of Medicine, Showa University, Tokyo 142, Japan
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China.,Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, 214200, PR China
| | - Yan Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China.,Traditional Chinese Medicine Administration, The Key Laboratory of Syndrome Differentiation & Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China.,Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, 214200, PR China
| |
Collapse
|
48
|
Li H, Yao Q, Xu F, Li Y, Kim D, Chung J, Baek G, Wu X, Hillman PF, Lee EY, Ge H, Fan J, Wang J, Nam SJ, Peng X, Yoon J. An Activatable AIEgen Probe for High-Fidelity Monitoring of Overexpressed Tumor Enzyme Activity and Its Application to Surgical Tumor Excision. Angew Chem Int Ed Engl 2020; 59:10186-10195. [PMID: 32155310 DOI: 10.1002/anie.202001675] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Indexed: 12/17/2022]
Abstract
Monitoring fluctuations in enzyme overexpression facilitates early tumor detection and excision. An AIEgen probe (DQM-ALP) for the imaging of alkaline phosphatase (ALP) activity was synthesized. The probe consists of a quinoline-malononitrile (QM) core decorated with hydrophilic phosphate groups as ALP-recognition units. The rapid liberation of DQM-OH aggregates in the presence of ALP resulted in aggregation-induced fluorescence. The up-regulation of ALP expression in tumor cells was imaged using DQM-ALP. The probe permeated into 3D cervical and liver tumor spheroids for imaging spatially heterogeneous ALP activity with high spatial resolution on a two-photon microscopy platform, providing the fluorescence-guided recognition of sub-millimeter tumorigenesis. DQM-ALP enabled differentiation between tumor and normal tissue ex vivo and in vivo, suggesting that the probe may serve as a powerful tool to assist surgeons during tumor resection.
Collapse
Affiliation(s)
- Haidong Li
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Qichao Yao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China
| | - Feng Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China
| | - Yueqing Li
- School of Pharmaceutical Science and Technology, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China
| | - Dayeh Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Jeewon Chung
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Gain Baek
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Xiaofeng Wu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Prima Fitria Hillman
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Eun Young Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Haoying Ge
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China.,Research Institute of Dalian University of Technology in Shenzhen, Gaoxin South fourth Road, Shenzhen, 518057, P. R. China
| | - Jingyun Wang
- School of Life Science and Biotechnology, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, P. R. China.,Research Institute of Dalian University of Technology in Shenzhen, Gaoxin South fourth Road, Shenzhen, 518057, P. R. China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| |
Collapse
|
49
|
Li H, Yao Q, Xu F, Li Y, Kim D, Chung J, Baek G, Wu X, Hillman PF, Lee EY, Ge H, Fan J, Wang J, Nam S, Peng X, Yoon J. An Activatable AIEgen Probe for High‐Fidelity Monitoring of Overexpressed Tumor Enzyme Activity and Its Application to Surgical Tumor Excision. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202001675] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Haidong Li
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Qichao Yao
- State Key Laboratory of Fine ChemicalsDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
| | - Feng Xu
- State Key Laboratory of Fine ChemicalsDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
| | - Yueqing Li
- School of Pharmaceutical Science and TechnologyDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
| | - Dayeh Kim
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Jeewon Chung
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Gain Baek
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Xiaofeng Wu
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | | | - Eun Young Lee
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Haoying Ge
- State Key Laboratory of Fine ChemicalsDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
| | - Jiangli Fan
- State Key Laboratory of Fine ChemicalsDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
- Research Institute of Dalian University of Technology in Shenzhen Gaoxin South fourth Road Shenzhen 518057 P. R. China
| | - Jingyun Wang
- School of Life Science and BiotechnologyDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
| | - Sang‐Jip Nam
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| | - Xiaojun Peng
- State Key Laboratory of Fine ChemicalsDalian University of Technology 2 Linggong Road Dalian 116024 P. R. China
- Research Institute of Dalian University of Technology in Shenzhen Gaoxin South fourth Road Shenzhen 518057 P. R. China
| | - Juyoung Yoon
- Department of Chemistry and NanoscienceEwha Womans University Seoul 03760 Korea
| |
Collapse
|
50
|
He Z, Thorrez L, Siegfried G, Meulemans S, Evrard S, Tejpar S, Khatib AM, Creemers JWM. The proprotein convertase furin is a pro-oncogenic driver in KRAS and BRAF driven colorectal cancer. Oncogene 2020; 39:3571-3587. [PMID: 32139876 DOI: 10.1038/s41388-020-1238-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022]
Abstract
Mutations in KRAS and/or BRAF that activate the ERK kinase are frequently found in colorectal cancer (CRC) and drive resistance to targeted therapies. Therefore, the identification of therapeutic targets that affect multiple signaling pathways simultaneously is crucial for improving the treatment of patients with KRAS or BRAF mutations. The proprotein convertase furin activates several oncogenic protein precursors involved in the ERK-MAPK pathway by endoproteolytic cleavage. Here we show that genetic inactivation of furin suppresses tumorigenic growth, proliferation, and migration in KRAS or BRAF mutant CRC cell lines but not in wild-type KRAS and BRAF cells. In a mouse xenograft model, these KRAS or BRAF mutant cells lacking furin displayed reduced growth and angiogenesis, and increased apoptosis. Mechanistically, furin inactivation prevents the processing of various protein pecursors including proIGF1R, proIR, proc-MET, proTGF-β1 and NOTCH1 leading to potent and durable ERK-MAPK pathway suppression in KRAS or BRAF mutant cells. Furthermore, we identified genes involved in activating the ERK-MAPK pathway, such as PTGS2, which are downregulated in the KRAS or BRAF mutant cells after furin inactivation but upregulated in wild-type KRAS and BRAF cells. Analysis of human colorectal tumor samples reveals a positive correlation between enhanced furin expression and KRAS or BRAF expression. These results indicate that furin plays an important role in KRAS or BRAF-associated ERK-MAPK pathway activation and tumorigenesis, providing a potential target for personalized treatment.
Collapse
Affiliation(s)
- Zongsheng He
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Lieven Thorrez
- Interdisciplinary Research Facility, Department of Development and Regeneration, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Sandra Meulemans
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Serge Evrard
- INSERM, LAMC, UMR, Allée Geoffroy St Hilaire, 1029, Pessac, France.,Institut Bergonié, Bordeaux, France
| | - Sabine Tejpar
- Digestive Oncology Unit, Department of Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - John W M Creemers
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium.
| |
Collapse
|