1
|
Li Q, Zhao X, Yang H, Zhu X, Sui X, Feng J. Modulating Endoplasmic Reticulum Stress in Gastrointestinal Cancers: Insights from Traditional Chinese Medicine. Pharmaceuticals (Basel) 2024; 17:1599. [PMID: 39770441 PMCID: PMC11676909 DOI: 10.3390/ph17121599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) play critical roles in tumorigenesis, cancer progression, and drug resistance. Persistent activation of the ER stress system enhances the survival capacities of malignant tumor cells, including increased proliferation, invasion, and resistance to treatment. Dysregulation of ER function and the resultant stress is a common cellular response to cancer therapies and may lead to cancer cell death. Currently, growing evidence suggests that Traditional Chinese medicine (TCM), either as a monotherapy or in combination with other treatments, offers significant advantages in preventing cancer, inhibiting tumor growth, reducing surgical complications, improving drug sensitivity, and mitigating drug-induced damage. Some of these natural products have even entered clinical trials as primary or complementary anticancer agents. In this review, we summarize the anticancer effects of TCM monomers/natural products on the gastrointestinal (GI) tumors and explore their mechanisms through ER stress modulation. We believe that ongoing laboratory research and the clinical development of TCM-based cancer therapies hold considerable potential for advancing future cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (Q.L.); (X.Z.); (H.Y.); (X.Z.); (X.S.)
| |
Collapse
|
2
|
Ounjaijean S, Somsak V. Synergistic antimalarial treatment of Plasmodium berghei infection in mice with dihydroartemisinin and Gymnema inodorum leaf extract. BMC Complement Med Ther 2023; 23:20. [PMID: 36690988 PMCID: PMC9869572 DOI: 10.1186/s12906-023-03850-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Chemotherapy is crucial in the fight against malaria. The rise of resistance to most antimalarial medicines has been a serious hurdle to effective treatment. Artemisinin-based combination therapies (ACTs) are currently the most effective antimalarial medication. Malaria parasites are growing more resistant to ACTs, particularly in Southeast Asia. As a result, effective alternative antimalarials are in high demand. The leaf extract of Gymnema inodorum (GIE) has previously shown promise as an effective antimalarial. Therefore, this study evaluated the antimalarial potential of combination dihydroartemisinin (DHA) and GIE therapy against Plasmodium berghei in a mouse model. METHODS The medications were evaluated using the standard 4-day test for determining the 50% effective dosage (ED50) of DHA and GIE on P. berghei ANKA (PbANKA). DHA and GIE were combined using a fixed-ratio approach, with DHA/GIE ED50s of 100/0, 80/20, 60/40, 40/60, 20/80, and 0/100, respectively. RESULTS The ED50 against PbANKA was determined to be 2 mg/kg of DHA and 100 mg/kg of GIE. The 60/40 (DHA/GIE) ratio demonstrated significantly higher antimalarial activity than the other ratios (p < 0.001) against PbANKA, with 88.95% inhibition, suggesting synergistic efficacy (combination index (CI) = 0.68695). Furthermore, this ratio protected PbANKA-infected mice against loss of body weight and packed cell volume decline, leading to a longer survival time over 30 days. CONCLUSION Our results suggest that GIE could be an effective adjuvant to DHA that can enhance the antimalarial effects in the treatment of PbANKA-infected mice.
Collapse
Affiliation(s)
- Sakaewan Ounjaijean
- grid.7132.70000 0000 9039 7662Research Institute for Health Sciences, Chiang Mai University, 50200 Chiang Mai, Thailand ,grid.7132.70000 0000 9039 7662Environmental-Occupational Health Sciences and Non-Communicable Diseases Research Group (EOHS and NCD Research Group), Research Institute for Health Sciences, Chiang Mai University, 50200 Chiang Mai, Thailand
| | - Voravuth Somsak
- grid.412867.e0000 0001 0043 6347School of Allied Health Sciences, Walailak University, 80160 Nakhon Si Thammarat, Thailand ,grid.412867.e0000 0001 0043 6347Research Excellence Center for Innovation and Health Products, Walailak University, 80160 Nakhon Si Thammarat, Thailand
| |
Collapse
|
3
|
Wong KH, Yang D, Chen S, He C, Chen M. Development of Nanoscale Drug Delivery Systems of Dihydroartemisinin for Cancer Therapy: A Review. Asian J Pharm Sci 2022; 17:475-490. [PMID: 36105316 PMCID: PMC9459003 DOI: 10.1016/j.ajps.2022.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/20/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022] Open
|
4
|
Selective Inhibition of Plasmodium falciparum ATPase 6 by Artemisinins and Identification of New Classes of Inhibitors after Expression in Yeast. Antimicrob Agents Chemother 2022; 66:e0207921. [PMID: 35465707 PMCID: PMC9112895 DOI: 10.1128/aac.02079-21] [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] [Indexed: 01/24/2023] Open
Abstract
Treatment failures with artemisinin combination therapies (ACTs) threaten global efforts to eradicate malaria. They highlight the importance of identifying drug targets and new inhibitors and of studying how existing antimalarial classes work. Here, we report the successful development of a heterologous expression-based compound-screening tool. The validated drug target Plasmodium falciparum ATPase 6 (PfATP6) and a mammalian orthologue (sarco/endoplasmic reticulum calcium ATPase 1a [SERCA1a]) were functionally expressed in Saccharomyces cerevisiae, providing a robust, sensitive, and specific screening tool. Whole-cell and in vitro assays consistently demonstrated inhibition and labeling of PfATP6 by artemisinins. Mutations in PfATP6 resulted in fitness costs that were ameliorated in the presence of artemisinin derivatives when studied in the yeast model. As previously hypothesized, PfATP6 is a target of artemisinins. Mammalian SERCA1a can be mutated to become more susceptible to artemisinins. The inexpensive, low-technology yeast screening platform has identified unrelated classes of druggable PfATP6 inhibitors. Resistance to artemisinins may depend on mechanisms that can concomitantly address multitargeting by artemisinins and fitness costs of mutations that reduce artemisinin susceptibility.
Collapse
|
5
|
Youssef HMK, Radi DA, Abd El-Azeem MA. Expression of TSP50, SERCA2 and IL-8 in Colorectal Adenoma and Carcinoma: Correlation to Clinicopathological Factors. Pathol Oncol Res 2021; 27:1609990. [PMID: 34744521 PMCID: PMC8566330 DOI: 10.3389/pore.2021.1609990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022]
Abstract
Background: Colorectal cancer (CRC) is the third most common type of cancer, it is considered a genetically heterogeneous disease with different molecular pathways being involved in its initiation and progression. Testes-specific protease 50 (TSP50) gene is a member of cancer/testis antigens that encodes for threonine protease enzyme. Overexpression of TSP50 was found to enhance the progression and invasion of breast cancer and other malignant tumors. SERCA2 is widely expressed in several body tissues; its aberrant expression has been involved in many cancers. IL-8 is an inflammatory cytokine. Alongside its role in inflammation, its expression was reported to induce the migration of tumor cells. Aim: Study the expression of TSP50, SERCA2 and IL-8 in colorectal adenoma (CRA), CRC and normal colonic tissues to compare the expression of these biomarkers in relation to clinicopathological parameters and prognostic factors. Results: TSP50, SERCA2 and IL-8 expression varied between normal colonic tissues, CRA and CRC. Significant statistical association was detected between the three biomarkers' overexpression and degree of dysplasia in CRA. Also, significant statistical relation was found between the three biomarkers' overexpression and presence of lympho-vascular invasion, advanced TNM staging and high intra-tumoral inflammatory infiltrate. Multivariable analysis showed that the overexpression of the three biomarkers is significantly associated with worse prognosis. Conclusion: The expression of TSP50, SERCA2 and IL-8 was different between the normal tissue and neoplastic colorectal tissue on one hand and between CRA and CRC on the other. Increased expression of these biomarkers in neoplastic epithelial cells of colorectal carcinoma is associated with adverse prognostic factors and could be considered as independent prognostic factors.
Collapse
Affiliation(s)
- Heba M K Youssef
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dina A Radi
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | |
Collapse
|
6
|
Zhang G, Li N, Tong Y, Li P, Han H, Song Q, Yang B, Cui L. Artemisinin derivatives inhibit adipogenic differentiation of 3T3-L1 preadipocytes through upregulation of CHOP. Biochem Biophys Res Commun 2021; 557:309-315. [PMID: 33894419 DOI: 10.1016/j.bbrc.2021.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/02/2021] [Indexed: 10/21/2022]
Abstract
Artemisinin derivatives could inhibit adipogenic differentiation of 3T3-L1 preadipocytes and prevent obesity in mice. However, the molecular mechanism remains largely unclear. Our research was designed to investigate the specific molecular target of artemisinin derivatives in adipogenic differentiation of 3T3-L1 preadipocytes. Here, we revealed that in response to dihydroartemisinin (DHA) or artesunate (ATS), intracellular lipid was decreased in a concentration dependent manner as shown by BODIPY staining. Quantitative PCR analysis showed that expression of Cebpa, Pparg, Fabp4 and Plin was significantly decreased by DHA treatment in a concentration and time dependent manner. Also, DHA treatment remarkably downregulated expression of CCAAT/enhancer-binding protein α (C/EBPα) and nuclear receptor peroxisome proliferation-activated receptor γ (PPARγ) of adipogenic induced 3T3-L1 cells as assayed by western blotting. RNA-seq analysis identified thousands of differential expression genes (DEGs), among which CHOP expression was significantly improved in DHA treated cells. Upregulation of CHOP was verified by quantitative PCR and western blotting, respectively. Knockdown of CHOP by the specific shRNA revealed that the inhibition of adipogenesis by DHA was strongly blocked, resulting in restored lipid accumulation and expression of adipogenic molecules. In conclusions, the inhibitory effect of DHA on adipogenic differentiation of 3T3-L1 preadipocytes was exerted in a concentration and time dependent manner, which was mediated by expression of CHOP.
Collapse
Affiliation(s)
- Guoying Zhang
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Na Li
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yongjuan Tong
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Peilin Li
- Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Haotian Han
- Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Qihan Song
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Bin Yang
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Lei Cui
- Department of Central Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
7
|
Chen K, Hua H, Zhu Z, Wu T, Jia Z, Liu Q. Artemisinin and dihydroartemisinin promote β-cell apoptosis induced by palmitate via enhancing ER stress. Apoptosis 2021; 25:192-204. [PMID: 31894447 DOI: 10.1007/s10495-019-01587-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Artemisinin (ART) and dihydroartemisinin (DHA) are first-line antimalarial drugs and have been reported to have anti-obesity effects. Hyperlipidemia is associated with β-cell damage in obese subjects, which could contribute to the pathogenesis of type 2 diabetes. In addition to their anti-obesity effects, ART and DHA also have protective roles in some diseases. Thus, we investigated the effects of ART and DHA in palmitate-induced β-cell apoptosis and the underlying mechanism. In this study, the rat pancreatic β-cell line INS-1 and mouse pancreatic β-cell line MIN6 were cultured with palmitate (PA) (0.1 mM) to induce cell apoptosis in the presence or absence of ART or DHA. Cell apoptosis was investigated by using flow cytometry, and the expression of ER stress markers, including CHOP, GRP78 and PDI, was detected by Western blotting and/or qRT-PCR. The results showed that ART and DHA significantly increased the apoptosis of β-cells induced by PA and exacerbated the ER stress caused by PA. An inhibitor of ER stress, 4-phenylbutyric acid (4-PBA), significantly ameliorated cell apoptosis caused by ART and DHA in PA-treated β-cells, consistent with the inhibition of ER stress. Together, the findings from the current study suggested that ART and DHA may promote lipid disorder-associated β-cell injury via enhancing ER stress when they were used to treat obesity.
Collapse
Affiliation(s)
- Ke Chen
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
| | - Hu Hua
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ziyang Zhu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
| | - Tong Wu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China.
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China.
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing, 210008, China.
| |
Collapse
|
8
|
Wang F, Gao Q, Yang J, Wang C, Cao J, Sun J, Fan Z, Fu L. Artemisinin suppresses myocardial ischemia-reperfusion injury via NLRP3 inflammasome mechanism. Mol Cell Biochem 2020; 474:171-180. [PMID: 32729005 DOI: 10.1007/s11010-020-03842-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/17/2020] [Indexed: 01/28/2023]
Abstract
Artemisinin is known for its pharmaceutical effect against malaria and received increased attention for its other potential function. Mounting evidence suggest that artemisinin could also exert cardioprotective effects while the understanding of its regulatory mechanism is still limited. This study is designed to investigate the role of artemisinin in myocardial ischemia/reperfusion (I/R) injury and the involvement of NLRP3 inflammasome. Artemisinin was administrated for 14 consecutive days intragastrically before I/R injury. Cardiac function was assessed by echocardiography. Infarct area was observed through HE and TTC staining. Apoptosis and autophagy were assessed by TUNEL and Western blotting. The artemisinin-treated myocardial I/R rats demonstrated less severe myocardial I/R injury (smaller infarct size and lower CK-MB, LDH), significant inhibition of cardiac autophagy (decreased LC3II/I and increased p62), improved mitochondrial electron transport chain activity, concomitant with decreased activation of NLRP3 inflammasome (decreased NLRP3, ASC, cleaved caspase-1, IL-1β). In conclusion, our findings further confirmed that activation of the NLRP3 inflammasome pathway is involved in myocardial I/R injury, whereas artemisinin preconditioning could effectively protect against myocardial I/R injury through suppression of NLRP3 inflammasome activation. Therefore, the NLRP3 inflammasome might serve as a promising therapeutic target providing new mechanisms for understanding the effect of artemisinin during the evolution of myocardial infarction.
Collapse
Affiliation(s)
- Fengyue Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Qianping Gao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Jing Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Can Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Junxian Cao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Junfeng Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Zhixin Fan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Lu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
9
|
Luo Y, Guo Q, Zhang L, Zhuan Q, Meng L, Fu X, Hou Y. Dihydroartemisinin exposure impairs porcine ovarian granulosa cells by activating PERK-eIF2α-ATF4 through endoplasmic reticulum stress. Toxicol Appl Pharmacol 2020; 403:115159. [PMID: 32721431 DOI: 10.1016/j.taap.2020.115159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022]
Abstract
Dihydroartemisinin (DHA) is an artemisinin derivative commonly used in malaria therapy, and a growing number of studies have focused on the potent anticancer activity of DHA. However, the reproductive toxicity of anticancer drugs is a major concern for young female cancer patients. Previous studies have suggested that DHA can cause embryonic damage and affect oocyte maturation. Here, we explored the side effects of DHA exposure on ovarian somatic cells. We exposed porcine granulosa cells to 5 μM and 40 μM DHA for 24 h or 48 h in vitro. DHA inhibited granulosa cell viability in a dose-dependent manner and, in the 48 h treatment group, DHA enhanced the apoptotic rate. We observed that the levels of intracellular calcium, mitochondrial calcium, and ATP concentration were elevated with DHA treatment. In granulosa cells exposed to DHA, the mRNA levels of endoplasmic reticulum stress-related genes GRP78 and ATF4 were increased. Furthermore, analysis of the unfolded protein response signaling pathway showed that the protein levels of P-PERK, P-eIF2α, and ATF4 were upregulated by DHA exposure. These results demonstrate that in granulosa cells, DHA exposure induces endoplasmic reticulum stress that then activates the PERK/eIF2α/ATF4 signaling pathway, thus providing insight into the mechanism underlying DHA-induced reproductive toxicity, and giving reference to DHA use in females.
Collapse
Affiliation(s)
- Yan Luo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qing Guo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Luyao Zhang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qingrui Zhuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lin Meng
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yunpeng Hou
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
10
|
Elhassanny AEM, Soliman E, Marie M, McGuire P, Gul W, ElSohly M, Van Dross R. Heme-Dependent ER Stress Apoptosis: A Mechanism for the Selective Toxicity of the Dihydroartemisinin, NSC735847, in Colorectal Cancer Cells. Front Oncol 2020; 10:965. [PMID: 32626657 PMCID: PMC7313430 DOI: 10.3389/fonc.2020.00965] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/15/2020] [Indexed: 01/05/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death in the United States. Artemisinin derivatives, including the dihydroartemisinin (DHA) monomers, are widely used as clinical agents for the treatment of malaria. Numerous studies demonstrate that these molecules also display antineoplastic activity with minimal toxicity. Of interest, dimeric DHA molecules are more active than their monomeric counterparts. Our previous data showed that the DHA dimer, NSC735847, was a potent inducer of death in different cancer cell types. However, the mechanism of action and activity of NSC735847 in colon cancer cells was not explored. The present study investigated the anticancer activity of NSC735847 and four structurally similar analog in human tumorigenic (HT-29 and HCT-116) and non-tumorigenic (FHC) colon cell lines. NSC735847 was more cytotoxic toward tumorigenic than non-tumorigenic colonocytes. In addition, NSC735847 exhibited greater cytotoxicity and tumor selectivity than the NSC735847 derivatives. To gain insight into mechanisms of NSC735847 activity, the requirement for endoplasmic reticulum (ER) stress and oxidative stress was tested. The data show that ER stress played a key role in the cytotoxicity of NSC735847 while oxidative stress had little impact on cell fate. In addition, it was observed that the cytotoxic activity of NSC735847 required the presence of heme, but not iron. The activity of NSC735847 was then compared to clinically utilized CRC therapeutics. NSC735847 was cytotoxic toward colon tumor cells at lower concentrations than oxaliplatin (OX). In addition, cell death was achieved at lower concentrations in colon cancer cells that were co-treated with folinic acid (Fol), 5-FU (F), and NSC735847 (FolFNSC), than Fol, F, and OX (FolFOX). The selective activity of NSC735847 and its ability to induce cytotoxicity at low concentrations suggest that NSC735847 may be an alternative for oxaliplatin in the FolFOX regimen for patients who are unable to tolerate its adverse effects.
Collapse
Affiliation(s)
- Ahmed E M Elhassanny
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Eman Soliman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mona Marie
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Paul McGuire
- Medical Doctor Program, Brody School of Medicine, Greenville, NC, United States
| | - Waseem Gul
- ElSohly Laboratories Inc., Oxford, MS, United States.,National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS, United States
| | - Mahmoud ElSohly
- ElSohly Laboratories Inc., Oxford, MS, United States.,National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS, United States
| | - Rukiyah Van Dross
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Center for Health Disparities, East Carolina University, Greenville, NC, United States
| |
Collapse
|
11
|
Abstract
Artemisinin (ART) and its derivatives are one of the most important classes of antimalarial agents, originally derived from a Chinese medicinal plant called Artemisia annua L. Beyond their outstanding antimalarial and antischistosomal activities, ART and its derivatives also possess both in-vitro and in-vivo activities against various types of cancer. Their anticancer effects range from initiation of apoptotic cell death to inhibition of cancer proliferation, metastasis and angiogenesis, and even modulation of the cell signal transduction pathway. This review provides a comprehensive update on ART and its derivatives, their mechanisms of action, and their synergistic effects with other chemicals in targeting leukemia cells. Combined with limited evidence of drug resistance and low toxicity profile, we conclude that ART and its derivatives, including dimers, trimers, and hybrids, might be a potential therapeutic alternative to current chemotherapies in combating leukemia, although more studies are necessary before they can be applied clinically.
Collapse
|
12
|
Yang X, Lou J, Shan W, Hu Y, Du Q, Liao Q, Xie R, Xu J. Pathogenic roles of altered calcium channels and transporters in colon tumorogenesis. Life Sci 2019; 239:116909. [PMID: 31689439 DOI: 10.1016/j.lfs.2019.116909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 01/26/2023]
|
13
|
Shi H, Bi H, Sun X, Dong H, Jiang Y, Mu H, Li W, Liu G, Gao R, Su J. Tubeimoside-1 inhibits the proliferation and metastasis by promoting miR-126-5p expression in non-small cell lung cancer cells. Oncol Lett 2018; 16:3126-3134. [PMID: 30127904 PMCID: PMC6096222 DOI: 10.3892/ol.2018.9051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/12/2018] [Indexed: 12/27/2022] Open
Abstract
Tubeimoside-1 (TBMS1) possesses broad anticancer activities, including the cytostatic and anti-angiogenesis effects in lung cancer. However, the effect of TBMS1 on the metastasis of non-small cell lung cancer (NSCLC) cells and the potential underlying mechanism remain unclear. In the present study, a cell counting kit-8 assay revealed that TBMS1 suppressed the proliferation of NCI-H1299 cells significantly, particularly following 48 h of treatment. Further studies showed that TBMS1 notably enhanced the apoptosis, and inhibited the migration and invasion of NCI-H1299 cells upon treatment for 48 h. A total of 14 NSCLC tissues and 14 normal adjacent tissues were collected, reverse transcription-quantitative polymerase chain reaction revealed decreased expression of microRNA (miR)-126-5p in NSCLC tissues compared with adjacent NSCLC tissues, which was reversed following TBMS1 administration in NCI-H1299 cells. The overexpression of miR-126-5p induced by TBMS1 was demonstrated to target and downregulate vascular endothelial growth factor (VEGF)-A. Simultaneously, the expression of VEGF-R2 was reduced notably, along with a significant declined in the phosphorylation levels of dual specificity mitogen-activated protein kinase kinase 1 and extracellular signal-regulated kinase (ERK)1/2. Overall, the aforementioned results indicated that TBMS1 inhibited the proliferation and metastasis, and promoted the apoptosis of NCI-H1299 cells, which may be mediated by overexpressing miR-126-5p, which inactivates the VEGF-A/VEGFR2/ERK signaling pathway. Therefore, TBMS1 may be a promising drug for prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Hanbing Shi
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Hongxia Bi
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Xingyuan Sun
- Department of Neurology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haiying Dong
- Laboratory Center of Ultrastructural Pathology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yunfei Jiang
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haijun Mu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Wei Li
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Guohua Liu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Ruizhi Gao
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Jiang Su
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
14
|
Toxicity and related mechanisms of dihydroartemisinin on porcine oocyte maturation in vitro. Toxicol Appl Pharmacol 2018; 341:8-15. [DOI: 10.1016/j.taap.2018.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 11/19/2022]
|
15
|
Qu C, Ma J, Liu X, Xue Y, Zheng J, Liu L, Liu J, Li Z, Zhang L, Liu Y. Dihydroartemisinin Exerts Anti-Tumor Activity by Inducing Mitochondrion and Endoplasmic Reticulum Apoptosis and Autophagic Cell Death in Human Glioblastoma Cells. Front Cell Neurosci 2017; 11:310. [PMID: 29033794 PMCID: PMC5626852 DOI: 10.3389/fncel.2017.00310] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 09/19/2017] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) is the most advanced and aggressive form of gliomas. Dihydroartemisinin (DHA) has been shown to exhibit anti-tumor activity in various cancer cells. However, the effect and molecular mechanisms underlying its anti-tumor activity in human GBM cells remain to be elucidated. Our results proved that DHA treatment significantly reduced cell viability in a dose- and time-dependent manner by CCK-8 assay. Further investigation identified that the cell viability was rescued by pretreatment either with Z-VAD-FMK, 3-methyladenine (3-MA) or in combination. Moreover, DHA induced apoptosis of GBM cells through mitochondrial membrane depolarization, release of cytochrome c and activation of caspases-9. Enhanced expression of GRP78, CHOP and eIF2α and activation of caspase 12 were additionally confirmed that endoplasmic reticulum (ER) stress pathway of apoptosis was involved in the cytotoxicity of DHA. DHA-treated GBM cells exhibited the morphological and biochemical changes typical of autophagy. Co-treatment with chloroquine (CQ) significantly induced the above effects. Furthermore, ER stress and mitochondrial dysfunction were involved in the DHA-induced autophagy. Further study revealed that accumulation of reactive oxygen species (ROS) was attributed to the DHA induction of apoptosis and autophagy. The illustration of these molecular mechanisms will present a novel insight for the treatment of human GBM.
Collapse
Affiliation(s)
- Chengbin Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Jing Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Lei Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China.,Liaoning Key Laboratory of Neuro-Oncology, Shenyang, China
| |
Collapse
|
16
|
Chemaly ER, Troncone L, Lebeche D. SERCA control of cell death and survival. Cell Calcium 2017; 69:46-61. [PMID: 28747251 DOI: 10.1016/j.ceca.2017.07.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 12/31/2022]
Abstract
Intracellular calcium (Ca2+) is a critical coordinator of various aspects of cellular physiology. It is increasingly apparent that changes in cellular Ca2+ dynamics contribute to the regulation of normal and pathological signal transduction that controls cell growth and survival. Aberrant perturbations in Ca2+ homeostasis have been implicated in a range of pathological conditions, such as cardiovascular diseases, diabetes, tumorigenesis and steatosis hepatitis. Intracellular Ca2+ concentrations are therefore tightly regulated by a number of Ca2+ handling enzymes, proteins, channels and transporters located in the plasma membrane and in Ca2+ storage organelles, which work in concert to fine tune a temporally and spatially precise Ca2+ signal. Chief amongst them is the sarco/endoplasmic reticulum (SR/ER) Ca2+ ATPase pump (SERCA) which actively re-accumulates released Ca2+ back into the SR/ER, therefore maintaining Ca2+ homeostasis. There are at least 14 different SERCA isoforms encoded by three ATP2A1-3 genes whose expressions are species- and tissue-specific. Altered SERCA expression and activity results in cellular malignancy and induction of ER stress and ER stress-associated apoptosis. The role of SERCA misregulation in the control of apoptosis in various cell types and disease setting with prospective therapeutic implications is the focus of this review. Ca2+ is a double edge sword for both life as well as death, and current experimental evidence supports a model in which Ca2+ homeostasis and SERCA activity represent a nodal point that controls cell survival. Pharmacological or genetic targeting of this axis constitutes an incredible therapeutic potential to treat different diseases sharing similar biological disorders.
Collapse
Affiliation(s)
- Elie R Chemaly
- Division of Nephrology and Hypertension, Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Luca Troncone
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
17
|
Zhang T, Hu Y, Wang T, Cai P. Dihydroartemisinin inhibits the viability of cervical cancer cells by upregulating caveolin 1 and mitochondrial carrier homolog 2: Involvement of p53 activation and NAD(P)H:quinone oxidoreductase 1 downregulation. Int J Mol Med 2017; 40:21-30. [PMID: 28498397 PMCID: PMC5466377 DOI: 10.3892/ijmm.2017.2980] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 04/07/2017] [Indexed: 12/25/2022] Open
Abstract
Dihydroartemisinin (DHA) has been shown to inhibit the viability of various cancer cells. Previous studies have revealed that the mechanisms involved in the inhibitory effects of DHA are based on theactivation of p53 and the mitochondrial-related cell death pathway. However, the exact association between upstream signaling and the activation of cell death pathway remains unclear. In this study, we found that DHA treatment induced the upregulation of caveolin 1 (Cav1) and mitochondrial carrier homolog 2 (MTCH2) in HeLa cells, and this was associated with the DHA-induced inhibition of cell viability and DHA-induced apoptosis. Additionally, the overexpression of Cav1 and MTCH2 in HeLa cells enhanced the inhibitory effects of DHA on cell viability. Moreover, we also found that the upregulation of Cav1 contributed to the DHA-mediated p53 activation and the downregulation of the redox enzyme, NAD(P)H:quinone oxidoreductase 1 (NQO1), which have been reported to contribute to the activation of the cell death pathway. Of note, we also found that DHA induced the nuclear translocation and accumulation of both Cav1 and p53, indicating a novel potential mechanism, namely the regulation of p53 activation by Cav1. On the whole, our study identified Cav1 and MTCH2 as the molecular targets of DHA and revealed a new link between the upstream Cav1/MTCH2 upregulation and the downstream activation of the cell death pathway involved in the DHA-mediated inhibition of cell viability.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Medical Cell Biology and Genetics, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuan Hu
- Department of Anatomy and Histology, School of Medicine, Chengdu University, Chengdu, Sichuan 610106, P.R. China
| | - Ting Wang
- Department of Anatomy and Histology, School of Medicine, Chengdu University, Chengdu, Sichuan 610106, P.R. China
| | - Peiling Cai
- Department of Anatomy and Histology, School of Medicine, Chengdu University, Chengdu, Sichuan 610106, P.R. China
| |
Collapse
|
18
|
Chen BY, Chen D, Lyu JX, Li KQ, Jiang MM, Zeng JJ, He XJ, Hao K, Tao HQ, Mou XZ, Ying YM, Zhang W, Zhu MH, Wang Z. Marsdeniae tenacissimae extract (MTE) suppresses cell proliferation by attenuating VEGF/VEGFR2 interactions and promotes apoptosis through regulating PKC pathway in human umbilical vein endothelial cells. Chin J Nat Med 2017; 14:922-930. [PMID: 28262119 DOI: 10.1016/s1875-5364(17)30017-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 01/13/2023]
Abstract
Marsdeniae tenacissimae extract (MTE), commonly known as Xiao-Ai-Ping in China, is a traditional Chinese herb medicine capable of inhibiting proliferation and metastasis and boosting apoptosis in various cancer cells. However, little is known about the contribution of MTE towards tumor angiogenesis and the underlying mechanism. The present study aimed to evaluate the effects of MTE on the proliferation and apoptosis of human umbilical vein endothelial cells (HUVECs) and the molecular mechanism. 3-(4,5-dimethylthiazol-2-yl)-5(3-carboxymethoxyphenyl)-2-(4-sulfopheny)-2H-tetrazolium, inner salt (MTS) and PI-stained flow cytometry assays revealed that MTE dose-dependently reduced the proliferation of HUVECs by arresting cell cycle at S phase (P < 0.05). Annexin V-FITC/PI-stained flow cytometry confirmed that MTE (160 μL·L-1) enhanced the apoptosis of HUVECs significantly (P < 0.001). Real-time quantitative RT-PCR and Western blot analyses showed an increase in Bax expression and a sharply decline in Bcl-2 expression; caspase-3 was activated simultaneously in a dose-dependent manner (P < 0.05). Further study observed the dose-dependent down-regulation of vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2), P2Y6 receptor (P2Y6R), and chemokine (C-C motif) ligand 2 (CCL-2), along with the activation of PKC Δ and up-regulation of p53 in a dose-dependent manner in MTE-treated selected cells (P < 0.05). Collectively, the results from the present study suggested that MTE suppressed the proliferation by attenuating CCL-2-mediated VEGF/VEGFR2 interactions and promoted the apoptosis through PKCΔ-induced p53-dependent mitochondrial pathway in HUVECs, supporting that MTE may be developed as a potent anti-cancer medicine.
Collapse
Affiliation(s)
- Bing-Yu Chen
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Dong Chen
- Wenzhou Center for Disease Control and Prevention, Wenzhou 325001, China
| | - Jian-Xin Lyu
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Kai-Qiang Li
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Meng-Meng Jiang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jing-Jing Zeng
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xu-Jun He
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Ke Hao
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Hou-Quan Tao
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - You-Min Ying
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Zhang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Meng-Hua Zhu
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| | - Zhen Wang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
19
|
Mokarram P, Albokashy M, Zarghooni M, Moosavi MA, Sepehri Z, Chen QM, Hudecki A, Sargazi A, Alizadeh J, Moghadam AR, Hashemi M, Movassagh H, Klonisch T, Owji AA, Łos MJ, Ghavami S. New frontiers in the treatment of colorectal cancer: Autophagy and the unfolded protein response as promising targets. Autophagy 2017; 13:781-819. [PMID: 28358273 PMCID: PMC5446063 DOI: 10.1080/15548627.2017.1290751] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC), despite numerous therapeutic and screening attempts, still remains a major life-threatening malignancy. CRC etiology entails both genetic and environmental factors. Macroautophagy/autophagy and the unfolded protein response (UPR) are fundamental mechanisms involved in the regulation of cellular responses to environmental and genetic stresses. Both pathways are interconnected and regulate cellular responses to apoptotic stimuli. In this review, we address the epidemiology and risk factors of CRC, including genetic mutations leading to the occurrence of the disease. Next, we discuss mutations of genes related to autophagy and the UPR in CRC. Then, we discuss how autophagy and the UPR are involved in the regulation of CRC and how they associate with obesity and inflammatory responses in CRC. Finally, we provide perspectives for the modulation of autophagy and the UPR as new therapeutic options for CRC treatment.
Collapse
Affiliation(s)
- Pooneh Mokarram
- a Colorectal Research Center and Department of Biochemistry , School of Medicine, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mohammed Albokashy
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Maryam Zarghooni
- c Zabol University of Medical Sciences , Zabol , Iran.,d University of Toronto Alumni , Toronto , ON , Canada
| | - Mohammad Amin Moosavi
- e Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology , Tehran , Iran
| | - Zahra Sepehri
- c Zabol University of Medical Sciences , Zabol , Iran
| | - Qi Min Chen
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | | | | | - Javad Alizadeh
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Adel Rezaei Moghadam
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Mohammad Hashemi
- g Department of Clinical Biochemistry , School of Medicine, Zahedan University of Medical Sciences , Zahedan , Iran
| | - Hesam Movassagh
- h Department of Immunology , Rady Faculty of Health Sciences, College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Thomas Klonisch
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Ali Akbar Owji
- i Department of Clinical Biochemistry , School of Medicine, Shiraz Medical University , Shiraz , Iran
| | - Marek J Łos
- j Małopolska Centre of Biotechnology , Jagiellonian University , Krakow , Poland ; LinkoCare Life Sciences AB , Sweden
| | - Saeid Ghavami
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada.,k Health Policy Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|