1
|
Jurjus A, El Masri J, Ghazi M, El Ayoubi LM, Soueid L, Gerges Geagea A, Jurjus R. Mechanism of Action of Melatonin as a Potential Adjuvant Therapy in Inflammatory Bowel Disease and Colorectal Cancer. Nutrients 2024; 16:1236. [PMID: 38674926 PMCID: PMC11054672 DOI: 10.3390/nu16081236] [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: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammatory bowel disease (IBD), a continuum of chronic inflammatory diseases, is tightly associated with immune system dysregulation and dysbiosis, leading to inflammation in the gastrointestinal tract (GIT) and multiple extraintestinal manifestations. The pathogenesis of IBD is not completely elucidated. However, it is associated with an increased risk of colorectal cancer (CRC), which is one of the most common gastrointestinal malignancies. In both IBD and CRC, a complex interplay occurs between the immune system and gut microbiota (GM), leading to the alteration in GM composition. Melatonin, a neuroendocrine hormone, was found to be involved with this interplay, especially since it is present in high amounts in the gut, leading to some protective effects. Actually, melatonin enhances the integrity of the intestinal mucosal barrier, regulates the immune response, alleviates inflammation, and attenuates oxidative stress. Thereby, the authors summarize the multifactorial interaction of melatonin with IBD and with CRC, focusing on new findings related to the mechanisms of action of this hormone, in addition to its documented positive outcomes on the treatment of these two pathologies and possible future perspectives to use melatonin as an adjuvant therapy.
Collapse
Affiliation(s)
- Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Jad El Masri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
- Faculty of Medical Sciences, Lebanese University, Beirut 6573, Lebanon;
| | - Maya Ghazi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
- Faculty of Medical Sciences, Lebanese University, Beirut 6573, Lebanon;
| | | | - Lara Soueid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Alice Gerges Geagea
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| |
Collapse
|
2
|
Wang L, Peng JL. METTL5 serves as a diagnostic and prognostic biomarker in hepatocellular carcinoma by influencing the immune microenvironment. Sci Rep 2023; 13:10755. [PMID: 37400463 DOI: 10.1038/s41598-023-37807-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/28/2023] [Indexed: 07/05/2023] Open
Abstract
Despite the abnormal expression of 18S rRNA m6A methyltransferase METTL5 being reported in some types of human malignancies, but its effect on hepatocellular carcinoma (HCC) remains to be unclear. This study aims to elucidate the influences of METTL5 on the carcinogenesis and progression of HCC. Expressions of METTL5 gene, transcript, protein, and promoter methylation in HCC were examined through multiple databases, c-BioPortal was used to confirm the genomic alterations of METTL5, the biological functions, target networks of kinases and microRNAs of METTL5, and its interactive differential genes were investigated through LinkedOmics. The possible correlation of METTL5 with the tumor-related infiltration of immune cells for HCC were explored comprehensively by using the online tools of TIMER and TISIDB. Expressions of METTL5 gene, mRNA, and protein were considerably overexpressed in HCC samples in comparison with healthy samples. The high methylation of the METTL5 promoter was observed in HCC tissues. Elevated METTL5 expression exhibited unfavorable survival outcomes in HCC patients. METTL5 expression were enriched in the signaling pathways of ribosome and oxidative phosphorylation, mismatch repair, and spliceosome through the involvement of several cancer-related kinases and miRNAs. The METTL5 expression has a positive correlation with the infiltration degree of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells in HCC. Marker genes of tumor immune-infiltrated cells have strong connection with METTL5. Furthermore, the upregulation of METTL5 was strongly correlated with the immune regulation of immunomodulators, chemokines, and chemokine receptors in the immune microenvironment. The oncogenesis and development of HCC are closely related to METTL5 expression, and the overexpression of METTL5 resulted in the poor survival outcome of HCC patients by regulating tumor immune microenvironment.
Collapse
Affiliation(s)
- Lei Wang
- Department of Rehabilitation Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410001, Hunan Province, China
| | - Jin-Lin Peng
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
3
|
Targhazeh N, Hutt KJ, Winship AL, Reiter R, Yousefi B. Melatonin as an oncostatic agent: Review of the modulation of tumor microenvironment and overcoming multidrug resistance. Biochimie 2022; 202:71-84. [PMID: 36116742 DOI: 10.1016/j.biochi.2022.09.010] [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: 07/13/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
Abstract
Multi drug resistance (MDR) generally limits the efficacy of chemotherapy in cancer patients and can be categorized into primary or acquired resistance. Melatonin (MLT), a lipophilic hormone released from pineal gland, is a molecule with oncostatic effects. Here, we will briefly review the contribution of different microenvironmental components including fibroblasts, immune and inflammatory cells, stem cells and vascular endothelial cells in tumor initiation, progression and development. Then, the mechanisms by which MLT can potentially affect these elements and regulate drug resistance will be presented. Finally, we will explain how different studies have used novel strategies incorporating MLT to suppress cancer resistance against therapeutics.
Collapse
Affiliation(s)
- Niloufar Targhazeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karla J Hutt
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Amy L Winship
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Russel Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA.
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Erdogan CS, Al Hassadi Y, Aru B, Yilmaz B, Gemici B. Combinatorial effects of melatonin and paclitaxel differ depending on the treatment scheme in colorectal cancer in vitro. Life Sci 2022; 308:120927. [PMID: 36063977 DOI: 10.1016/j.lfs.2022.120927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
AIMS Colorectal carcinoma (CRC) is the third most prevalent cancer with high mortality. Besides regulating the circadian rhythm, melatonin (MTN) exerts anticancer activities. Paclitaxel (PTX) is successful against different malignancies, however, acquired resistance and variability in patient response restrict its use. mTOR and MAPK pathways are often deregulated in human cancers. We aimed to investigate whether MTN enhances or sensitizes the chemotherapeutic activity of PTX and if so, determine the underlying possible mechanisms in CRC in vitro. MAIN METHODS Antiproliferative and cytotoxic activities of PTX and MTN were assessed alone and in combination, as well as with different treatment regimens (renewal or replacement of the treatment after 24 h), up to 48 h. Apoptosis, viability and autophagy were assessed by flow cytometry. mTOR and MAPK pathway activities were investigated by immunoblotting. KEY FINDINGS Both drugs reduced cell viability in a dose-dependent manner at 24 and 48 h. Only the highest dose of MTN (500 μM) potentiated the cytotoxicity of PTX (50 nM). Replacement of PTX after 24 h with MTN was superior in reducing cell viability than vice versa via apoptosis induction. Renewal of MTN treatment every 24 h reduced autophagy compared to the control group, while other treatments did not alter the autophagic activity. A 24 h MTN treatment followed by 24 h PTX treatment increased S6 phosphorylation in a mTOR-independent manner and increased Erk1/2 phosphorylation. SIGNIFICANCE The present study suggests that sequential treatment with MTN and PTX distinctly affect apoptosis and cytotoxicity via regulating mTOR and MAPK pathways differentially in CRC.
Collapse
Affiliation(s)
- Cihan Suleyman Erdogan
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Yasmine Al Hassadi
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Basak Aru
- Yeditepe University, Faculty of Medicine, Department of Immunology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Bayram Yilmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Burcu Gemici
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey.
| |
Collapse
|
5
|
Sadoughi F, Dana PM, Homayoonfal M, Sharifi M, Asemi Z. Molecular basis of melatonin protective effects in metastasis: A novel target of melatonin. Biochimie 2022; 202:15-25. [DOI: 10.1016/j.biochi.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022]
|
6
|
Shi X, Yu X, Wang J, Bian S, Li Q, Fu F, Zou X, Zhang L, Bast RC, Lu Z, Guo L, Chen Y, Zhou J. SIK2 promotes ovarian cancer cell motility and metastasis by phosphorylating MYLK. Mol Oncol 2022; 16:2558-2574. [PMID: 35278271 PMCID: PMC9251837 DOI: 10.1002/1878-0261.13208] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/11/2022] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Salt‐inducible kinase 2 (SIK2; also known as serine/threonine‐protein kinase SIK2) is overexpressed in several cancers and has been implicated in cancer progression. However, the mechanisms by which SIK2 regulates cancer cell motility, migration and metastasis in ovarian cancer have not been fully discovered. Here, we identify that SIK2 promotes ovarian cancer cell motility, migration and metastasis in vitro and in vivo. Mechanistically, SIK2 regulated cancer cell motility and migration by myosin light chain kinase, smooth muscle (MYLK)‐meditated phosphorylation of myosin light chain 2 (MYL2). SIK2 directly phosphorylated MYLK at Ser343 and activated its downstream effector MYL2, promoting ovarian cancer cell motility and metastasis. In addition, we found that adipocytes induced SIK2 phosphorylation at Ser358 and MYLK phosphorylation at Ser343, enhancing ovarian cancer cell motility. Moreover, SIK2 protein expression was positively correlated with the expression of MYLK‐pS343 in ovarian cancer cell lines and tissues. The co‐expression of SIK2 and MYLK‐pS343 was associated with reduced median overall survival in human ovarian cancer samples. Taken together, SIK2 positively regulates ovarian cancer motility, migration and metastasis, suggesting that SIK2 is a potential candidate for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiu Shi
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xuejiao Yu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Shimin Bian
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Qiutong Li
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
- Jiangsu Institute of Clinical Immunology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Xinwei Zou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lin Zhang
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Robert C. Bast
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Zhen Lu
- Department of Imaging Department The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
| | - Lingchuan Guo
- Department of Experimental Therapeutics University of Texas M.D. Anderson Cancer Center Houston Texas USA
| | - Youguo Chen
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology The First Affiliated Hospital of Soochow University Suzhou People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology Jiangsu Key Laboratory of Clinical Immunology Soochow University Suzhou People’s Republic of China
| |
Collapse
|
7
|
The interplay of pineal hormones and socioeconomic status leading to colorectal cancer disparity. Transl Oncol 2022; 16:101330. [PMID: 34990909 PMCID: PMC8741600 DOI: 10.1016/j.tranon.2021.101330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in the United States. Despite increased screening options and state-of-art treatments offered in clinics, racial differences remain in CRC. African Americans (AAs) are disproportionately affected by the disease; the incidence and mortality are higher in AAs than Caucasian Americans (CAs). At the time of diagnosis, AAs more often present with advanced stages and aggressive CRCs, primarily accounting for the racial differences in therapeutic outcomes and mortality. The early incidence of CRC in AAs could be attributed to race-specific gene polymorphisms and lifestyle choices associated with socioeconomic status (SES). Altered melatonin-serotonin signaling, besides the established CRC risk factors (age, diet, obesity, alcoholism, and tobacco use), steered by SES, glucocorticoid, and Vitamin D status in AAs could also account for the early incidence in this racial group. This review focuses on how the lifestyle factors, diet, allelic variants, and altered expression of specific genes could lead to atypical serotonin and melatonin signaling by modulating the synthesis, secretion, and signaling of these pineal hormones in AAs and predisposing them to develop more aggressive CRC earlier than CAs. Crosstalk between gut microbiota and pineal hormones and its impact on CRC pathobiology is addressed from a race-specific perspective. Lastly, the status of melatonin-focused CRC treatments, the need to better understand the perturbed melatonin signaling, and the potential of pineal hormone-directed therapeutic interventions to reduce CRC-associated disparity are discussed.
Collapse
|
8
|
Therapeutic potential of melatonin in colorectal cancer: Focus on lipid metabolism and gut microbiota. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166281. [PMID: 34610472 DOI: 10.1016/j.bbadis.2021.166281] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/24/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal malignancies. The occurrence and development of CRC are complicated processes. Obesity and dysbacteriosis have been increasingly regarded as the main risk factors for CRC. Understanding the etiology of CRC from multiple perspectives is conducive to screening for some potential drugs or new treatment strategies to limit the serious side effects of conventional treatment and prolong the survival of CRC patients. Melatonin, a natural indoleamine, is mainly produced by the pineal gland, but it is also abundant in other tissues, including the gastrointestinal tract, retina, testes, lymphocytes, and Harder's glands. Melatonin could participate in lipid metabolism by regulating adipogenesis and lipolysis. Additionally, many studies have focused on the potential beneficial effects of melatonin in CRC, such as promotion of apoptosis; inhibition of cell proliferation, migration, and invasion; antioxidant activity; and immune regulation. Meaningfully, gut microbiota is the main determinant of all aspects of health and disease (including obesity and tumorigenesis). The gut microbiota is of great significance for understanding the relationship between obesity and increased risk of CRC. Although the current understanding of how the melatonin-mediated gut microbiota coordinates a variety of physiological and pathological activities is fairly comprehensive, there are still many unknown topics to be explored in the face of a complex nutritional status and a changeable microbiota. This review summarizes the potential links among melatonin, lipid metabolism, gut microbiota, and CRC to promote the development of melatonin as a preventive and therapeutic agent for CRC.
Collapse
|
9
|
Pourhanifeh MH, Mehrzadi S, Kamali M, Hosseinzadeh A. Melatonin and gastrointestinal cancers: Current evidence based on underlying signaling pathways. Eur J Pharmacol 2020; 886:173471. [PMID: 32877658 DOI: 10.1016/j.ejphar.2020.173471] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Gastrointestinal (GI) cancers, leading causes of cancer-related deaths, have been serious challenging human diseases up to now. Because of high rates of mortality, late-stage diagnosis, metastasis to distant locations, and low effectiveness and adverse events of routine standard therapies, the quality of life and survival time are low in patients with GI cancers. Hence, many efforts need to be done to explore and find novel efficient treatments. Beneficial effects of melatonin have been reported in a wide variety of human diseases. Melatonin has antioxidant, anti-inflammatory, antimicrobial, and anticancer effects. Various studies have showed the regulatory effects of melatonin on apoptotsis, autophagy and angiogenesis; these properties result in the inhibition of invasion, migration, and proliferation of GI cancer cells in vivo and in vitro. Together, this review suggests that melatonin in combination with anticancer agents may improve the efficacy of routine medicine and survival rate of patients with cancer.
Collapse
Affiliation(s)
- Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Kvietkauskas M, Zitkute V, Leber B, Strupas K, Stiegler P, Schemmer P. The role of melatonin in colorectal cancer treatment: a comprehensive review. Ther Adv Med Oncol 2020; 12:1758835920931714. [PMID: 32733605 PMCID: PMC7370547 DOI: 10.1177/1758835920931714] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer worldwide, known as the second leading cause of cancer-related deaths annually. Currently, multimodal treatment strategies, including surgical resection, combined with chemotherapy and radiotherapy, have been used as conventional treatments in patients with CRC. However, clinical outcome of advanced stage disease remains relatively discouraging, due mainly to appearance of CRC chemoresistance, toxicity, and other detrimental side effects. New strategies to overcome these limitations are essential. During the last decades, melatonin (MLT) has been shown to be a potent antiproliferative, anti-metastatic agent with cytotoxic effects on different types of human malignancies, including CRC. Hence, this comprehensive review compiles the available experimental and clinical data analyzing the effects of MLT treatment in CRC patients and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Mindaugas Kvietkauskas
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Viktorija Zitkute
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Bettina Leber
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | | | - Philipp Stiegler
- General, Visceral and Transplant Surgery, Department of Surgery, Transplant Center Graz, Medical University of Graz, Auenbruggerplatz 29, Graz, 8036, Austria
| | - Peter Schemmer
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| |
Collapse
|
11
|
Bjørklund G, Rajib SA, Saffoon N, Pen JJ, Chirumbolo S. Insights on Melatonin as an Active Pharmacological Molecule in Cancer Prevention: What's New? Curr Med Chem 2019; 26:6304-6320. [PMID: 29714136 DOI: 10.2174/0929867325666180501094850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022]
Abstract
Along with playing an important role in circadian rhythm, melatonin is thought to play a significant role in preventing cells from damage, as well as in the inhibition of growth and in triggering apoptosis in malignant cells. Its relationship with circadian rhythms, energetic homeostasis, diet, and metabolism, is fundamental to achieve a better comprehension of how melatonin has been considered a chemopreventive molecule, though very few papers dealing with this issue. In this article, we tried to review the most recent evidence regarding the protective as well as the antitumoral mechanisms of melatonin, as related to diet and metabolic balance. From different studies, it was evident that an intracellular antioxidant defense mechanism is activated by upregulating an antioxidant gene battery in the presence of high-dose melatonin in malignant cells. Like other broad-spectrum antioxidant molecules, melatonin plays a vital role in killing tumor cells, preventing metastasis, and simultaneously keeping normal cells protected from oxidative stress and other types of tissue damage.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | | | - Nadia Saffoon
- Department of Pharmacy and Forensic Science, Faculty of Life Science and Medicine, King's College London, London, United Kingdom
| | - Joeri J Pen
- Diabetes Clinic, Department of Internal Medicine, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Department of Nutrition, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
12
|
Wu H, Liu J, Yin Y, Zhang D, Xia P, Zhu G. Therapeutic Opportunities in Colorectal Cancer: Focus on Melatonin Antioncogenic Action. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9740568. [PMID: 31637261 PMCID: PMC6766109 DOI: 10.1155/2019/9740568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 08/31/2019] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) influences individual health worldwide with high morbidity and mortality. Melatonin, which shows multiple physiological functions (e.g., circadian rhythm, immune modulation, and antioncogenic action), can be present in almost all organisms and found in various tissues including gastrointestinal tract. Notably, melatonin disruption is closely associated with the elevation of CRC incidence, indicating that melatonin is effective in suppressing CRC development and progression. Mechanistically, melatonin favors in activating apoptosis and colon cancer immunity, while reducing proliferation, autophagy, metastasis, and angiogenesis, thereby exerting its anticarcinogenic effects. This review highlights that melatonin can be an adjuvant therapy and be beneficial in treating patients suffering from CRC.
Collapse
Affiliation(s)
- Hucong Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jiaqi Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yi Yin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Dong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Pengpeng Xia
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
13
|
Mirza-Aghazadeh-Attari M, Mohammadzadeh A, Mostavafi S, Mihanfar A, Ghazizadeh S, Sadighparvar S, Gholamzadeh S, Majidinia M, Yousefi B. Melatonin: An important anticancer agent in colorectal cancer. J Cell Physiol 2019; 235:804-817. [PMID: 31276205 DOI: 10.1002/jcp.29049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
Colorectal cancer is one of the most common cancers among the elderly, which is also seen in the forms of hereditary syndromes occurring in younger individuals. Numerous studies have been conducted to understand the molecular and cellular pathobiology underlying colorectal cancer. These studies have found that cellular signaling pathways are at the core of colorectal cancer pathology. Because of this, new agents have been proposed as possible candidates to accompany routine therapy regimens. One of these agents is melatonin, a neuro-hormone known best for its essential role in upholding the circadian rhythm and orchestrating the many physiologic changes it accompanies. Melatonin is shown to be able to modulate many signaling pathways involved in many essential cell functions, which if deregulated cause an accelerated pace towards cancer. More so, melatonin is involved in the regulation of immune function, tumor microenvironment, and acts as an antioxidant agent. Many studies have focused on the beneficial effects of melatonin in colorectal cancers, such as induction of apoptosis, increased sensitivity to chemotherapy agents and radiotherapy, limiting cellular proliferation, migration, and invasion. The present review aims to illustrate the known significance of melatonin in colorectal cancer and to address possible clinical use.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mohammadzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Mostavafi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aynaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saber Ghazizadeh
- Danesh Pey Hadi Co., Health Technology Development Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Chok KC, Ng CH, Koh RY, Ng KY, Chye SM. The potential therapeutic actions of melatonin in colorectal cancer. Horm Mol Biol Clin Investig 2019; 39:hmbci-2019-0001. [DOI: 10.1515/hmbci-2019-0001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/01/2019] [Indexed: 12/24/2022]
Abstract
Abstract
Colorectal cancer (CRC) is the third most common cancer and lethal disease worldwide. Melatonin, an indoleamine produced in pineal gland, shows anticancer effects on a variety of cancers, especially CRC. After clarifying the pathophysiology of CRC, the association of circadian rhythm with CRC, and the relationship between shift work and the incidence of CRC is reviewed. Next, we review the role of melatonin receptors in CRC and the relationship between inflammation and CRC. Also included is a discussion of the mechanism of gene regulation, control of cell proliferation, apoptosis, autophagy, antiangiogenesis and immunomodulation in CRC by melatonin. A review of the drug synergy of melatonin with other anticancer drugs suggests its usefulness in combination therapy. In summary, the information compiled may serve as comprehensive reference for the various mechanisms of action of melatonin against CRC, and as a guide for the design of future experimental research and for advancing melatonin as a therapeutic agent for CRC.
Collapse
Affiliation(s)
- Kian Chung Chok
- School of Health Sciences, International Medical University , Kuala Lumpur , Malaysia
| | - Chew Hee Ng
- School of Pharmacy, International Medical University , Kuala Lumpur , Malaysia
| | - Rhun Yian Koh
- School of Health Sciences, International Medical University , Kuala Lumpur , Malaysia
| | - Khuen Yen Ng
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia , Selangor , Malaysia
| | - Soi Moi Chye
- School of Health Sciences, International Medical University , Kuala Lumpur , Malaysia , Phone: +6032731 7220; Fax: +60386567229
| |
Collapse
|
15
|
Shafabakhsh R, Reiter RJ, Davoodabadi A, Asemi Z. Melatonin as a potential inhibitor of colorectal cancer: Molecular mechanisms. J Cell Biochem 2019; 120:12216-12223. [PMID: 31087705 DOI: 10.1002/jcb.28833] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/17/2019] [Accepted: 01/24/2019] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a prevalent disease and a major cause of mortality in the world. Several factors including population aging, poor dietary habits, obesity, insufficient physical activity, and smoking can explain its increased prevalence. CRC is a heterogeneous disease both histopathologically and in term of its molecular and genetic aspects. Melatonin a derivative of tryptophan, is synthesized and released from pineal gland but it is also found in numerous extrapineal tissues including retina, testes, lymphocytes, Harderian gland, gastrointestinal tract, etc. This molecule has several tasks which enhance physiological functions such as antioxidant, antiaging, immunomodulatory, and tumor inhibition. Multiple immunocytochemical studies reported melatonin in the intestinal mucosa where its concentration is greater than in the blood. These findings suggest that melatonin may have a potential inhibitory role in CRC progression. The purpose of this review is to examine the effects of melatonin in molecular pathogenesis and signaling pathways of CRC.
Collapse
Affiliation(s)
- Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science, Center, San Antonio, Texas
| | - Abdoulhossein Davoodabadi
- Departments of General Surgery, Trauma Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
16
|
Xia N, Cui J, Zhu M, Xing R, Lu Y. Androgen receptor variant 12 promotes migration and invasion by regulating MYLK in gastric cancer. J Pathol 2019; 248:304-315. [PMID: 30737779 DOI: 10.1002/path.5257] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/14/2019] [Accepted: 02/06/2019] [Indexed: 12/30/2022]
Abstract
Androgen receptor (AR) and its variants (AR-Vs) promote tumorigenesis and metastasis in many hormone-related cancers, such as breast, prostate and hepatocellular cancers. However, the expression patterns and underlying molecular mechanisms of AR in gastric cancer (GC) are not fully understood. This study aimed to detect the expression of AR-Vs in GC and explored their role in metastasis of GC. Here, the AR expression form was identified in GC cell lines and tissues by RT-PCR and qPCR. Transwell assays and experimental lung metastasis animal models were used to assess the function of AR in cell migration and invasion. Downstream targets of AR were screened by bioinformatics, and identified by luciferase reporter assays and electrophoretic mobility shift assays. AR-v12 was identified as the main expression form in GC cell lines and tissues. Different from full length of AR, AR-v12 was localized to the nucleus independent of androgen. Upregulation of AR-v12 in primary GC tissues was significantly associated with metastasis. Overexpression of AR-v12 promoted migration and invasion independent of androgen. Knockdown of AR-v12 inhibited migration and invasion in vitro, as well as metastasis in vivo. Furthermore, AR-v12, serving as a transcription factor, promoted metastasis through regulating the promoter activity of MYLK. In AR-v12 overexpressing cells, knockdown of MYLK inhibited cell migration and invasion, while in AR-v12 knocked-down cells, overexpression of MYLK promoted cell migration and invasion. Collectively, our study demonstrates that AR-v12 is highly expressed in GC tissues and promotes migration and invasion through directly regulating MYLK. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nan Xia
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Jiantao Cui
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Min Zhu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Rui Xing
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China.,Department of Medical Oncology, Beijing Hospital, Beijing 100730, PR China
| |
Collapse
|
17
|
Gil-Martín E, Egea J, Reiter RJ, Romero A. The emergence of melatonin in oncology: Focus on colorectal cancer. Med Res Rev 2019; 39:2239-2285. [PMID: 30950095 DOI: 10.1002/med.21582] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/04/2019] [Accepted: 03/16/2019] [Indexed: 12/17/2022]
Abstract
Within the last few decades, melatonin has increasingly emerged in clinical oncology as a naturally occurring bioactive molecule with substantial anticancer properties and a pharmacological profile optimal for joining the currently available pharmacopeia. In addition, extensive experimental data shows that this chronobiotic agent exerts oncostatic effects throughout all stages of tumor growth, from initial cell transformation to mitigation of malignant progression and metastasis; additionally, melatonin alleviates the side effects and improves the welfare of radio/chemotherapy-treated patients. Thus, the support of clinicians and oncologists for the use of melatonin in both the treatment and proactive prevention of cancer is gaining strength. Because of its epidemiological importance and symptomatic debut in advanced stages of difficult clinical management, colorectal cancer (CRC) is a preferential target for testing new therapies. In this regard, the development of effective forms of clinical intervention for the improvement of CRC outcome, specifically metastatic CRC, is urgent. At the same time, the need to reduce the costs of conventional anti-CRC therapy results is also imperative. In light of this status quo, the therapeutic potential of melatonin, and the direct and indirect critical processes of CRC malignancy it modulates, have aroused much interest. To illuminate the imminent future on CRC research, we focused our attention on the molecular mechanisms underlying the multiple oncostatic actions displayed by melatonin in the onset and evolution of CRC and summarized epidemiological evidence, as well as in vitro, in vivo and clinical findings that support the broadly protective potential demonstrated by melatonin.
Collapse
Affiliation(s)
- Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, Vigo, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Laboratory, Research Unit, Hospital Universitario Santa Cristina, Madrid, Spain.,Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas, USA
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
18
|
Salem SM, Hamed AR, Fayez AG, Nour Eldeen G. Non-target Genes Regulate miRNAs-Mediated Migration Steering of Colorectal Carcinoma. Pathol Oncol Res 2018; 25:559-566. [PMID: 30361904 DOI: 10.1007/s12253-018-0502-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) trigger a two-layer regulatory network directly or through transcription factors and their co-regulators. Unlike miR-375, the role of miR-145 and miR-224 in inhibiting or driving cancer cell migration is controversial. This study is a step towards addressing the potential of miR-375, miR-145 and miR-224 expression modulation to inhibit colorectal carcinoma (CRC) cells migration in vitro through regulation of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44. Transwell migration assay results revealed a significant subdue of migration ability of cells transfected with miR-375 and miR-145 mimics and miR-224 inhibitor. Real time PCR data showed that expression of VEGFA, TGFβ1, IGF1, CD105 and CD44 was downregulated as a consequence of exogenous re-expression of miR-375 and inhibition of miR-224. On the other hand, ectopic expression of miR-145 did not affect VEGFA, TGFβ1 and CD44 expression, while it elevated CD105 and suppressed IGF1 expression. MAP4K4, a predicted target of miR-145, was validated as a target that could play a role in miR-145-mediated regulation of migration. At mRNA level, no change was observed in expression of MAP4K4 in cells with restored expression of miR-145, while western blotting analysis revealed a 25% reduction of protein level. By applying luciferase reporter assay, a significant decrease in luciferase activity was observed, supporting that miR-145 directly target 3' UTR of MAP4K4. The study highlighted the involvement of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44 in mediating anti- and pro-migratory effect of miR-375 and miR-224, respectively, and validated MAP4K4 as a direct target of anti-migratory miR-145.
Collapse
Affiliation(s)
- Sohair M Salem
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Ahmed R Hamed
- Phytochemistry Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Biology Unit - Central Laboratory of Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Alaaeldin G Fayez
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Ghada Nour Eldeen
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Stem Cell Research Unit, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
19
|
de Lima RMT, Dos Reis AC, de Menezes AAPM, Santos JVDO, Filho JWGDO, Ferreira JRDO, de Alencar MVOB, da Mata AMOF, Khan IN, Islam A, Uddin SJ, Ali ES, Islam MT, Tripathi S, Mishra SK, Mubarak MS, Melo-Cavalcante AADC. Protective and therapeutic potential of ginger (Zingiber officinale) extract and [6]-gingerol in cancer: A comprehensive review. Phytother Res 2018; 32:1885-1907. [PMID: 30009484 DOI: 10.1002/ptr.6134] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/21/2022]
Abstract
Natural dietary agents have attracted considerable attention due to their role in promoting health and reducing the risk of diseases including cancer. Ginger, one of the most ancient known spices, contains bioactive compounds with several health benefits. [6]-Gingerol constitutes the most pharmacologically active among such compounds. The aim of the present work was to review the literature pertaining to the use of ginger extract and [6]-gingerol against tumorigenic and oxidative and inflammatory processes associated with cancer, along with the underlying mechanisms of action involved in signaling pathways. This will shed some light on the protective or therapeutic role of ginger derivatives in oxidative and inflammatory regulations during metabolic disturbance and on the antiproliferative and anticancer properties. Data collected from experimental (in vitro or in vivo) and clinical studies discussed in this review indicate that ginger extract and [6]-gingerol exert their action through important mediators and pathways of cell signaling, including Bax/Bcl2, p38/MAPK, Nrf2, p65/NF-κB, TNF-α, ERK1/2, SAPK/JNK, ROS/NF-κB/COX-2, caspases-3, -9, and p53. This suggests that ginger derivatives, in the form of an extract or isolated compounds, exhibit relevant antiproliferative, antitumor, invasive, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Rosália Maria Tôrres de Lima
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - Antonielly Campinho Dos Reis
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - Ag-Anne Pereira Melo de Menezes
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - José Victor de Oliveira Santos
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - José Williams Gomes de Oliveira Filho
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - José Roberto de Oliveira Ferreira
- Laboratory of Experimental Cancerology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - Marcus Vinícius Oliveira Barros de Alencar
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - Ana Maria Oliveira Ferreira da Mata
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| | - Ishaq N Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Amirul Islam
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Eunüs S Ali
- Gaco Pharmaceuticals and Research Laboratory, Dhaka-1000, Bangladesh; College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Muhammad Torequl Islam
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Swati Tripathi
- Amity Institute of Microbial Technology, Amity University, Noida, India
| | - Siddhartha Kumar Mishra
- Cancer Biology Laboratory, School of Biological Sciences (Zoology), Dr. Harisingh Gour Central University, Sagar, India
| | | | - Ana Amélia de Carvalho Melo-Cavalcante
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí, Teresina, Brazil
- Laboratory of Genetical Toxicology, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
20
|
Song Y, Miao S, Li Y, Fu H. Ulinastatin attenuates liver injury and inflammation in a cecal ligation and puncture induced sepsis mouse model. J Cell Biochem 2018; 120:417-424. [PMID: 30126034 DOI: 10.1002/jcb.27396] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/10/2018] [Indexed: 12/28/2022]
Abstract
Sepsis is a syndrome of life-threatening multiorgan dysfunction caused by host response dysregulation to infection. Ulinastatin (UTI), a serine protease inhibitor, possesses anti-inflammatory properties and has been suggested to modulate lipopolysaccharide-induced sepsis. However, little is known about the mechanism underlying its effects on sepsis. In the current study, we investigated the protective effect of UTI on liver injury in a cecal ligation and puncture (CLP)-induced sepsis of C57BL/6 mouse model and explored the possible mechanisms. Mice underwent CLP as sepsis models and were randomized into five groups including the sham group, UTI group, CLP group, UTI-L group, and UTI-H group. UTI was intraperitoneally administered at doses of UTI 1500 U/100 g (UTI-L group) or 3000 U/100 g (UTI-H group), before CLP. The mice were killed, and immunohistochemical changes, cytokine levels, and antioxidant enzyme activities were detected. Our results showed that UTI ameliorated CLP-mediated increases in serum aspartate aminotransferase and alanine aminotransferase activities, histological activity index, degenerative region ratio, and infiltrated inflammatory cell numbers. Moreover, UTI also decreased nitrotyrosine and 4-hydroxynonenal, activated caspase-3, and activated poly (ADP-ribose) polymerase (PARP) levels and inhibited the mitogen-activated protein kinase pathway activation in liver tissues. Our results indicated that UTI could inhibit CLP-induced liver injury by suppressing inflammation and oxidation. Our results indicated that UTI may serve as a potential therapeutic agent for sepsis.
Collapse
Affiliation(s)
- Yukang Song
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Taizhou, Zhejiang, China
| | - Shixing Miao
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Taizhou, Zhejiang, China
| | - Yexuzi Li
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Taizhou, Zhejiang, China
| | - Hui Fu
- Department of Medical Intensive Care Unit, The First People's Hospital of Wenling, Taizhou, Zhejiang, China
| |
Collapse
|
21
|
Lin J, He Y, Chen L, Chen X, Zang S, Lin W. MYLK promotes hepatocellular carcinoma progression through regulating cytoskeleton to enhance epithelial-mesenchymal transition. Clin Exp Med 2018; 18:523-533. [PMID: 29855744 DOI: 10.1007/s10238-018-0509-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/24/2018] [Indexed: 11/24/2022]
Abstract
Myosin light chain kinase (MYLK) is found to catalyze the phosphorylation of myosin light chains (MLC) and regulate invasion and metastasis in some malignancies. However, there is little knowledge on the role of MYLK in hepatocellular carcinoma (HCC), and no studies have been conducted to investigate the mechanisms underlying MYLK-mediated promotion of HCC invasion and metastasis until now. In this study, we investigated the expression of MYLK in 50 pairs of human HCC and adjacent liver specimens. High MYLK expression was significantly correlated with aggressive clinicopathological features including tumor encapsulation, microvascular invasion and metastasis. In vitro assays showed that shRNA-induced MYLK knockdown significantly inhibited the wound-healing ability of HCC cells and the ability to migrate and invade through Matrigel. We next uncovered that MYLK knockdown resulted in a reduction in the number of F-actin stress fibers, disorganization of F-actin architectures and morphological alterations of HCC cells. Phosphorylated MLC, rather than total MLC, was found to be markedly reduced in response to downregulation of MYLK expression, and MYLK-regulated actin cytoskeleton through phosphorylating MLC in HCC cells. In addition, Western blotting assay revealed downregulation of the epithelial marker E-cadherin and upregulation of mesenchymal markers Vimentin, N-cadherin and Snail. Taken together, our findings indicate that MYLK promotes HCC progression by altering cytoskeleton to enhance epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Jie Lin
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Yihui He
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Lingfeng Chen
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Xiaoyan Chen
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, 350001, China.,Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Shengbing Zang
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, No.420, Fuma Road, Jinan District, Fuzhou City, 350014, Fujian Province, China. .,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China.
| |
Collapse
|
22
|
Marques TC, da Silva Santos EC, Diesel TO, Leme LO, Martins CF, Dode M, Alves BG, Costa F, de Oliveira EB, Gambarini ML. Melatonin reduces apoptotic cells, SOD2 and HSPB1 and improves the in vitro production and quality of bovine blastocysts. Reprod Domest Anim 2017; 53:226-236. [PMID: 29205523 DOI: 10.1111/rda.13097] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
Effects of adding different concentrations of melatonin (10-7 , 10-9 and 10-11 M) to maturation (Experiment 1; Control, IVM + 10-7 , IVM + 10-9 , IVM + 10-11 ) and culture media (Experiment 2; Control, IVC + 10-7 , IVC + 10-9 , IVC + 10-11 ) were evaluated on in vitro bovine embryonic development. The optimal concentration of melatonin (10-9 M) from Experiments 1-2 was tested in both maturation and/or culture media of Experiment 3 (Control, IVM + 10-9 , IVC + 10-9 , IVM/IVC + 10-9 ). In Experiment 1, maturated oocytes from Control and IVM + 10-9 treatments showed increased glutathione content, mitochondrial membrane potential and percentage of Grade I blastocysts (40.6% and 43%, respectively). In Experiment 2, an increase in the percentage of Grade I blastocysts was detected in IVC + 10-7 (43.5%; 56.7%) and IVC + 10-9 (47.4%; 57.4%). Moreover, a lower number and percentage of apoptotic cells in blastocysts were observed in the IVC + 10-9 group compared to Control (3.8 ± 0.6; 3.6% versus 6.1 ± 0.6; 5.3%). In Experiment 3, the IVC + 10-9 treatment increased percentage of Grade I blastocysts with a lower number of apoptotic cells compared to IVM/IVC + 10-9 group (52.6%; 3.0 ± 0.5 versus 46.0%; 5.4 ± 1.0). The IVC + 10-9 treatment also had a higher mRNA expression of antioxidant gene (SOD2) compared to the Control, as well as the heat shock protein (HSPB1) compared to the IVM + 10-9 . Reactive oxygen species production was greater in the IVM/IVC + 10-9 treatment group. In conclusion, the 10-9 M concentration of melatonin and the in vitro production phase in which it is used directly affected embryonic development and quality.
Collapse
Affiliation(s)
- T C Marques
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - E C da Silva Santos
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - T O Diesel
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - L O Leme
- Embrapa Genetic Resources and Biotechnology, Laboratory of Animal Reproduction, Brasília, DF, Brazil
| | - C F Martins
- Center of Animal Production Systems, Embrapa Cerrados, Brasília, DF, Brazil
| | - Man Dode
- Embrapa Genetic Resources and Biotechnology, Laboratory of Animal Reproduction, Brasília, DF, Brazil
| | - B G Alves
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary, State University of Ceará, Fortaleza, CE, Brazil
| | - Fph Costa
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - E B de Oliveira
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| | - M L Gambarini
- Center for Studies and Research in Animal Reproductive Biology, College of Veterinary and Animal Science, Federal University of Goiás, Goiânia, GO, Brazil
| |
Collapse
|
23
|
Liu Z, Zou D, Yang X, Xue X, Zuo L, Zhou Q, Hu R, Wang Y. Melatonin inhibits colon cancer RKO cell migration by downregulating Rho‑associated protein kinase expression via the p38/MAPK signaling pathway. Mol Med Rep 2017; 16:9383-9392. [PMID: 29152648 PMCID: PMC5779993 DOI: 10.3892/mmr.2017.7836] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/25/2017] [Indexed: 01/05/2023] Open
Abstract
Melatonin is predominately produced and secreted by the pineal gland, and inhibits cell growth in various cancer cell lines such as colorectal cancer. However, the precise mechanisms involved have not been fully elucidated. In the present study, the potential molecular mechanism underlying the efficacy of melatonin on migration in RKO colon cancer cells was investigated. The effects of melatonin and H-1152, a selective inhibitor of Rho-associated protein kinase (ROCK), on the migration of RKO cells were analyzed by an in vitro wound healing assay. The localization of zonula occludens-1 (ZO-1) and occludin were observed by immunofluorescence. Reverse transcription-quantitative polymerase chain reaction (qPCR) was performed to analyze the relative mRNA levels of ROCK, ZO-1 and occludin. In addition, western blot analysis was implemented to examine the expression of ROCK, phospho (p)-myosin phosphatase targeting subunit 1 (MYPT1), p-myosin light chains (MLC) and p-p38. The results revealed that the expression levels of ROCK2, p-MYPT1 and p-MLC in RKO cells were decreased, and the membrane protein expression of ZO-1 and occludin increased when the cells were treated with melatonin. qPCR demonstrated that melatonin downregulated ROCK2 gene expression, and upregulated the expression of the ZO-1 and occludin genes. The levels of ZO-1 and occludin localized in the tight junctions were markedly increased in the immunofluorescence assay. In addition, the phosphorylation levels of p38 were reduced when the cells were treated with melatonin, and treatment with H-1152 downregulated p38 phosphorylation. The results indicated that melatonin may inhibit the migration of RKO colon cancer cells by downregulating ROCK expression via the p38/mitogen-activated protein kinase signaling pathway.
Collapse
Affiliation(s)
- Zhen Liu
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Duobing Zou
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Xiaoping Yang
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiaolong Xue
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Li Zuo
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Qing Zhou
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Ruolei Hu
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
24
|
The Impact of Melatonin on Colon Cancer Cells' Resistance to Doxorubicin in an in Vitro Study. Int J Mol Sci 2017; 18:ijms18071396. [PMID: 28788434 PMCID: PMC5535889 DOI: 10.3390/ijms18071396] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/18/2017] [Accepted: 06/23/2017] [Indexed: 02/06/2023] Open
Abstract
Multi-drug resistance (MDR) is the main cause of low effectiveness of cancer chemotherapy. P-glycoprotein (P-gp) is one of the main factors determining MDR. Some studies indicate the potential role of melatonin (MLT) in MDR. In this study, we examined the effect of MLT on colon cancer cell’s resistance to doxorubicin (DOX). Using the sulforhodamine B (SRB), method the effect of tested substances on the survival of LoVo (colon cancer cells sensitive to DOX) and LoVoDX (colon cancer cells resistant to DOX) was rated. Using immunocytochemistry (ICC), the expression of P-gp in the LoVo and LoVoDX was determined. With the real-time PCR (RT-PCR) technique, the ABCB1 expression in LoVoDX was evaluated. Based on the results, it was found that MLT in some concentrations intensified the cytotoxicity effect of DOX in the LoVoDX cells. In the ICC studies, it was demonstrated that certain concentrations of MLT and DOX cause an increase in the percentage of cells expressing P-gp, which correlates positively with ABCB1 expression (RT-PCR). The mechanism of overcoming resistance by MLT is probably not only associated with the expression of P-gp. It seems appropriate to carry out further research on the use of MLT as the substance supporting cancer chemotherapy.
Collapse
|
25
|
Li Y, Xu B, Xu M, Chen D, Xiong Y, Lian M, Sun Y, Tang Z, Wang L, Jiang C, Lin Y. 6-Gingerol protects intestinal barrier from ischemia/reperfusion-induced damage via inhibition of p38 MAPK to NF-κB signalling. Pharmacol Res 2017; 119:137-148. [PMID: 28167239 DOI: 10.1016/j.phrs.2017.01.026] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/25/2017] [Indexed: 01/08/2023]
Abstract
Intestinal ischemia reperfusion (I/R) injury caused by severe trauma, intestinal obstruction, and operation is one of the tough challenges in clinic. 6-Gingerol (6G), a main active ingredient of ginger, is found to have anti-microbial, anti-inflammatory, anti-oxidative, and anti-cancer activities. The present study was designed to characterize the potential protective effects of 6G on rat intestinal I/R injury and reveal the correlated mechanisms. Rat intestinal I/R model was established with clamping the superior mesenteric artery (SMA) and 6G was intragastrically administered for three consecutive days before I/R injury. Caco-2 and IEC-6 cells were incubated under hypoxia/reoxygenation (H/R) conditions to simulate I/R injury in vitro. The results showed that 6G significantly alleviated intestinal injury in I/R injured rats by reducing the generation of oxidative stress and inhibiting p38 MAPK signaling pathway. 6G significantly reduced MDA level and increased the levels of SOD, GSH, and GSH-Px in I/R injured intestinal tissues. 6G significantly decreased the production of proinflammatory cytokines including TNF-α, IL-1β, and IL-6, and inhibited the expression of inflammatory mediators iNOS/NO in I/R injured intestinal tissues. The impaired intestinal barrier function was restored by using 6G in I/R injured rats and in both Caco-2 and IEC-6 cells characterized by inhibiting p38 MAPK phosphorylation, nuclear translocation of NF-κB, and expression of myosin light chain kinase (MLCK) protein. 6G also reduced the generation of reactive oxygen species (ROS) in both Caco-2 and IEC-6 cells. In vitro transfection of p38 MAPK siRNA mitigated the impact of 6G on NF-κB and MLCK expression, and the results further corroborated the protective effects of 6G on intestinal I/R injury by repressing p38 MAPK signaling. In conclusion, the present study suggests that 6G exerts protective effects against I/R-induced intestinal mucosa injury by inhibiting the formation of ROS and p38 MAPK activation, providing novel insights into the mechanisms of this therapeutic candidate for the treatment of intestinal injury.
Collapse
Affiliation(s)
- Yanli Li
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Bin Xu
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Ming Xu
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Dapeng Chen
- Laboratory Animal Center, Dalian Medical University, Dalian 116044, China
| | - Yongjian Xiong
- Central Laboratory, The First Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Mengqiao Lian
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Yuchao Sun
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Li Wang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Chunling Jiang
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Yuan Lin
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
26
|
Liu S, Liang B, Jia H, Jiao Y, Pang Z, Huang Y. Evaluation of cell death pathways initiated by antitumor drugs melatonin and valproic acid in bladder cancer cells. FEBS Open Bio 2017; 7:798-810. [PMID: 28593135 PMCID: PMC5458469 DOI: 10.1002/2211-5463.12223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023] Open
Abstract
Effective drug combinations have the potential to strengthen therapeutic efficacy and combat drug resistance. Both melatonin and valproic acid (VPA) exhibit antitumor activities in various cancer cells. The aim of this study was to evaluate the cell death pathways initiated by anticancer combinatorial effects of melatonin and VPA in bladder cancer cells. The results demonstrated that the combination of melatonin and VPA leads to significant synergistic growth inhibition of UC3 bladder cancer cells. Gene expression studies revealed that cotreatment with melatonin and VPA triggered the up-regulation of certain genes related to apoptosis (TNFRSF10A and TNFRSF10B), autophagy (BECN, ATG3 and ATG5) and necrosis (MLKL, PARP-1 and RIPK1). The combinatorial treatment increased the expression of endoplasmic reticulum (ER)-stress-related genes ATF6, IRE1, EDEM1 and ERdj4. Cotreatment with melatonin and VPA enhanced the expression of E-cadherin, and decreased the expression of N-cadherin, Fibronectin, Snail and Slug. Furthermore, the Wnt pathway and Raf/MEK/ERK pathway were activated by combinatorial treatment. However, the effects on the expression of certain genes were not further enhanced in cells following combinatorial treatment in comparison to individual treatment of melatonin or VPA. In summary, these findings provided evidence that cotreatment with melatonin and VPA exerted increased cytotoxicity by regulating cell death pathways in UC3 bladder cancer cells, but the clinical significance of combinatorial treatment still needs to be further exploited.
Collapse
Affiliation(s)
- Siwei Liu
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Bilin Liang
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Huiting Jia
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Yuhan Jiao
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Zhongqiu Pang
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Yongye Huang
- College of Life and Health Sciences Northeastern University Shenyang China
| |
Collapse
|
27
|
Khapchaev AY, Shirinsky VP. Myosin Light Chain Kinase MYLK1: Anatomy, Interactions, Functions, and Regulation. BIOCHEMISTRY (MOSCOW) 2017; 81:1676-1697. [PMID: 28260490 DOI: 10.1134/s000629791613006x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This review discusses and summarizes the results of molecular and cellular investigations of myosin light chain kinase (MLCK, MYLK1), the key regulator of cell motility. The structure and regulation of a complex mylk1 gene and the domain organization of its products is presented. The interactions of the mylk1 gene protein products with other proteins and posttranslational modifications of the mylk1 gene protein products are reviewed, which altogether might determine the role and place of MLCK in physiological and pathological reactions of cells and entire organisms. Translational potential of MLCK as a drug target is evaluated.
Collapse
Affiliation(s)
- A Y Khapchaev
- Russian Cardiology Research and Production Center, Moscow, 121552, Russia.
| | | |
Collapse
|
28
|
Zhang S, Chen S, Li Y, Liu Y. Melatonin as a promising agent of regulating stem cell biology and its application in disease therapy. Pharmacol Res 2016; 117:252-260. [PMID: 28042087 DOI: 10.1016/j.phrs.2016.12.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 01/20/2023]
Abstract
Stem cells have emerged as an important approach to repair and regenerate damaged tissues or organs and show great therapeutic potential in a variety of diseases. However, the low survival of engrafted stem cells still remains a major challenge for stem cell therapy. As a major hormone from the pineal gland, melatonin has been shown to play an important role in regulating the physiological and pathological functions of stem cells, such as promoting proliferation, migration and differentiation. Thus, melatonin combined with stem cell transplantation displayed promising application potential in neurodegenerative diseases, liver cirrhosis, wound healing, myocardial infarction, kidney ischemia injury, osteoporosis, etc. It exerts its physiological and pathological functions through its anti-oxidant, anti-inflammatory, anti-apoptosis and anti-ageing properties. Here, we summarize recent advances on exploring the biological role of melatonin in stem cells, and discuss its potential applications in stem cell-based therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Simon Chen
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Yuan Li
- College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Yu Liu
- Department of Clinical Laboratory Diagnosis, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
29
|
Zhang L, Yu C, Chang Q, Luo X, Qiu J, Liu S. Comparison of gene expression profiles in aortic dissection and normal human aortic tissues. Biomed Rep 2016; 5:421-427. [PMID: 27699008 PMCID: PMC5038499 DOI: 10.3892/br.2016.740] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/06/2016] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to compare the gene expression profiles in aortic dissection (AD) and healthy human aortic tissue samples by DNA microarray analysis in order to screen the differential genes. In total, five AD and four healthy aortic specimens were selected; the total RNA was extracted and reverse transcribed into cDNA and in vitro transcribed into aRNA, followed by microarray hybridization for analysis. Thereafter, the transcription levels of six differential genes, myosin light chain kinase (MYLK), polycystin 1, transient receptor potential channel interacting (PKD-1), myosin heavy chain 11 (MYH11), superoxide dismutase 3, extracellular (SOD3), filamin A (FLNA), and transgelin (TAGLN), screened from the expression profiles were quantitatively verified. Compared with the healthy aortic specimens, a total of 1,661 genes in the AD group demonstrated more than 2-fold differential expression, of which 997 genes were upregulated and 664 genes were downregulated. Thereafter, six AD-associated genes that showed downregulation in the microarray assay were selected for quantitatively verifying the gene transcription level using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), which confirmed their downregulation compared with the healthy aortic tissue genes; of the six genes, the expression levels of MYLK, PKD-1, MYH11, SOD3 and TAGLN were significantly downregulated (P<0.05), while the expression of FLNA was not significantly downregulated (P>0.05). Thus, whole genome microarray may be used to screen differentially expressed genes between AD and healthy aortic tissues. When used in combination with RT-qPCR validation, this method may provide novel strategies for investigating AD.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| | - Cuntao Yu
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| | - Qian Chang
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| | - Xinjin Luo
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| | - Juntao Qiu
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| | - Shen Liu
- State Key Laboratory of Cardiovascular Disease, Aortic Surgery Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, P.R. China
| |
Collapse
|