1
|
Cho D, Huang X, Han Y, Kim M. NPC1L1 rs217434 A > G as a Novel Single Nucleotide Polymorphism Related to Dyslipidemia in a Korean Population. Biochem Genet 2024; 62:4103-4119. [PMID: 38280151 DOI: 10.1007/s10528-023-10649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/25/2023] [Indexed: 01/29/2024]
Abstract
A relationship between cholesterol levels and Niemann-Pick C1-Like 1 (NPC1L1) polymorphisms in diverse populations was found in previous studies. However, relevant research on this association in the Korean population is relatively scarce. Therefore, the current study sought to examine the correlation between the NPC1L1 rs217434 A > G polymorphism and clinical as well as biochemical variables pertaining to dyslipidemia in the Korean population. This cross-sectional single-center study included 1404 Korean subjects aged 20-86 years, grouped based on dyslipidemia presence (normal and dyslipidemia) and genotype (AA or AG). After adjusting for sex and age, it was discovered that the dyslipidemia group's BMI, diastolic blood pressure, glucose-related indicators, lipid profile, high-sensitivity C-reactive protein (hs-CRP), and parameters of oxidative stress were considerably different from the normal group's values. When grouped according to genotype, individuals in the AG group exhibited greater total cholesterol, low-density lipoprotein cholesterol, hs-CRP, and 8-epi-prostaglandin F2α in comparison to those in the AA group. Moreover, individuals with dyslipidemia and the AG genotype exhibited unfavorable outcomes for lipid profiles, markers related to glucose and inflammation, and markers of oxidative stress. This study provided evidence for a relationship between the NPC1L1 rs217434 A > G genotype and dyslipidemia in the Korean population, which highlights the potential of the NPC1L1 rs217434 A > G genotype as an early predictor of dyslipidemia.
Collapse
Affiliation(s)
- Dahyun Cho
- Department of Food and Nutrition, College of Life Science and Nano Technology, Hannam University, Daejeon, 34054, Republic of Korea
| | - Ximei Huang
- Department of Food and Nutrition, College of Life Science and Nano Technology, Hannam University, Daejeon, 34054, Republic of Korea
| | - Youngmin Han
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, 03722, Republic of Korea
| | - Minjoo Kim
- Department of Food and Nutrition, College of Life Science and Nano Technology, Hannam University, Daejeon, 34054, Republic of Korea.
| |
Collapse
|
2
|
Sun S, Ma J, Zuo T, Shi J, Sun L, Meng C, Shu W, Yang Z, Yao H, Zhang Z. Inhibition of PCSK9: A Promising Enhancer for Anti-PD-1/PD-L1 Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0488. [PMID: 39324018 PMCID: PMC11423609 DOI: 10.34133/research.0488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
Immune checkpoint therapy, such as programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) blockade, has achieved remarkable results in treating various tumors. However, most cancer patients show a low response rate to PD-1/PD-L1 blockade, especially those with microsatellite stable/mismatch repair-proficient colorectal cancer subtypes, which indicates an urgent need for new approaches to augment the efficacy of PD-1/PD-L1 blockade. Cholesterol metabolism, which involves generating multifunctional metabolites and essential membrane components, is also instrumental in tumor development. In recent years, inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9), a serine proteinase that regulates cholesterol metabolism, has been demonstrated to be a method enhancing the antitumor effect of PD-1/PD-L1 blockade to some extent. Mechanistically, PCSK9 inhibition can maintain the recycling of major histocompatibility protein class I, promote low-density lipoprotein receptor-mediated T-cell receptor recycling and signaling, and modulate the tumor microenvironment (TME) by affecting the infiltration and exclusion of immune cells. These mechanisms increase the quantity and enhance the antineoplastic effect of cytotoxic T lymphocyte, the main functional immune cells involved in anti-PD-1/PD-L1 immunotherapy, in the TME. Therefore, combining PCSK9 inhibition therapy with anti-PD-1/PD-L1 immunotherapy may provide a novel option for improving antitumor effects and may constitute a promising research direction. This review concentrates on the relationship between PCSK9 and cholesterol metabolism, systematically discusses how PCSK9 inhibition potentiates PD-1/PD-L1 blockade for cancer treatment, and highlights the research directions in this field.
Collapse
Affiliation(s)
- Shengbo Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingxin Ma
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingting Zuo
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Jinyao Shi
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Liting Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong Meng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wenlong Shu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhengyang Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
3
|
Butt E, Günder T, Stürzebecher P, Kowalski I, Schneider P, Buschmann N, Schäfer S, Bender A, Hermanns HM, Zernecke A. Cholesterol uptake in the intestine is regulated by the LASP1-AKT-NPC1L1 signaling pathway. Am J Physiol Gastrointest Liver Physiol 2024; 327:G25-G35. [PMID: 38713618 DOI: 10.1152/ajpgi.00222.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/09/2024]
Abstract
Cholesterol is essential for the stability and architecture of the plasma membrane and a precursor of bile acids and steroid hormones in mammals. Excess dietary cholesterol uptake leads to hypercholesterolemia and atherosclerosis and plays a role in cancer development. The role of actin-binding scaffolding protein LIM and SH3 protein 1 (LASP1) in cholesterol trafficking has not been investigated previously. Cholesterol levels, its uptake, and excretion were studied in mice deficient for low-density lipoprotein receptor and Lasp1 (Ldlr-/-Lasp1-/- mice) upon feeding a high-fat diet, and in LASP1-knockdown, differentiated human intestinal epithelial CaCo-2 cells. When compared with diet-fed Ldlr-/- control mice, Ldlr-/-Lasp1-/- mice displayed a reduction in serum cholesterol levels. Mechanistically, we identified a new role of LASP1 in controlling the translocation of the intestinal cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1) to the apical cell surface, which was limited in LASP1-knockdown human CaCo-2 enterocytes and in the intestine of Ldlr-/- Lasp1-/- compared with Ldlr-/- mice, linked to LASP1-pAKT signaling but not CDC42 activation. In line, a reduction in cholesterol reabsorption was noted in LASP1-knockdown CaCo-2 cells in vitro, and an enhanced cholesterol excretion via the feces was observed in Ldlr-/- Lasp1-/- mice. These data uncover a novel function of Lasp1 in cholesterol trafficking, promoting cholesterol reabsorption in the intestine. Targeting LASP1 locally could thus represent a novel targeting strategy to ameliorate hypercholesterolemia and associated diseases.NEW & NOTEWORTHY We here uncovered LASP1 as a novel regulator of the shuttling of the sterol transporter NPC1L1 to the cell surface in enterocytes to control cholesterol absorption. Accordingly, LASP1-deficient mice displayed lowered serum cholesterol levels under dietary cholesterol supplementation.
Collapse
Affiliation(s)
- Elke Butt
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Günder
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Paulina Stürzebecher
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Isabel Kowalski
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Pia Schneider
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Nils Buschmann
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Sarah Schäfer
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Alicia Bender
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike M Hermanns
- Division of Hepatology, University Hospital Würzburg, Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Zhang Z, Zhang D. Circulating lipids, lipid-lowering drug targets, and breast cancer risk: Comprehensive evidence from Mendelian randomization and summary data-based Mendelian randomization. Cancer Causes Control 2024; 35:983-994. [PMID: 38430374 DOI: 10.1007/s10552-024-01857-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/24/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most common and fatal cancer among women, yet the causal relationship between circulating lipids, lipid-lowering drugs, and BC remains unclear. METHODS Mendelian randomization (MR) and summary data-based MR (SMR) analysis are used to explore the causal relationship between plasma lipids, lipid-lowering drug targets, and BC. RESULTS The result of MR suggested that per mg/dL higher levels of LDL-C (OR = 1.045, FDR = 0.023), HDL-C (OR = 1.079, FDR = 0.003), TC (OR = 1.043, FDR = 0.026), and APOA-I (OR = 1.085, FDR = 2.64E-04) were associated with increased BC risk, while TG was associated with reduced BC risk (OR = 0.926, FDR = 0.003). Per mg/dL higher levels of HDL-C (OR = 1.080, FDR = 0.011) and APOA-I (OR = 1.083, FDR = 0.002) were associated with increased ER+BC risk, while TG was associated with reduced ER+BC risk (OR = 0.909, FDR = 0.002). For every per 1 mg/dL decrease in LDL, HMGCR (OR: 0.839; FDR = 0.016), NPC1L1 (OR: 0.702; FDR = 0.004), and PCSK9 (OR: 0.916; FDR = 0.026) inhibition were associated with reduced BC risk, whereas CETP inhibition (OR: 1.194; FDR = 0.026) was associated with increased BC risk. For every per 1 mg/dL decrease in LDL, HMGCR (OR: 0.822; FDR = 0.023), NPC1L1 (OR: 0.633; FDR = 2.37E-03), and APOB inhibition (OR: 0.816; FDR = 1.98E-03) were associated with decreased ER-BC risk, while CETP inhibition (OR: 1.465; FDR = 0.011) was associated with increased ER-BC risk. SMR analysis indicated that HMGCR was associated with increased BC risk (OR: 1.112; p = 0.044). CONCLUSION Lipids are associated with the BC risk, and lipid-lowering drugs targets HMGCR, NPC1L1, PCSK9, and APOB may be effective strategies for preventing BC. However, lipid-lowering drugs target CETP may potentially increase BC risk.
Collapse
Affiliation(s)
- Zhongxu Zhang
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Daxin Zhang
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Elste J, Cast N, Udawatte S, Adhikari K, Payen SH, Verma SC, Shukla D, Swanson-Mungerson M, Tiwari V. Co-Expression of Niemann-Pick Type C1-Like1 (NPC1L1) with ACE2 Receptor Synergistically Enhances SARS-CoV-2 Entry and Fusion. Biomedicines 2024; 12:821. [PMID: 38672177 PMCID: PMC11048565 DOI: 10.3390/biomedicines12040821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into human embryonic kidney (HEK293T) cells has been shown to be a cholesterol-rich, lipid raft-dependent process. In this study, we investigated if the presence of a cholesterol uptake receptor Niemann-pick type c1-like1 (NPC1L1) impacts SARS-CoV-2 cell entry. Initially, we utilized reporter-based pseudovirus cell entry assays and a spike (S) glycoprotein-mediated cell-to-cell fusion assay. Using Chinese hamster ovary (CHO-K1) cells, which lack endogenous receptors for SARS-CoV-2 entry, our data showed that the co-expression of NPC1L1 together with the ACE2 receptor synergistically increased SARS-CoV-2 pseudovirus entry even more than the cells expressing ACE-2 receptor alone. Similar results were also found with the HEK293T cells endogenously expressing the ACE2 receptor. Co-cultures of effector cells expressing S glycoprotein together with target cells co-expressing ACE-2 receptor with NPC1L1 significantly promoted quantitative cell-to-cell fusion, including syncytia formation. Finally, we substantiated that an elevated expression of NPC1L1 enhanced entry, whereas the depletion of NPC1L1 resulted in a diminished SARS-CoV-2 entry in HEK293T-ACE2 cells using authentic SARS-CoV-2 virus in contrast to their respective control cells. Collectively, these findings underscore the pivotal role of NPC1L1 in facilitating the cellular entry of SARS-CoV-2. Importance: Niemann-Pick type C1-like1 (NPC1L1) is an endosomal membrane protein that regulates intracellular cholesterol trafficking. This protein has been demonstrated to play a crucial role in the life cycle of several clinically important viruses. Although SARS-CoV-2 exploits cholesterol-rich lipid rafts as part of its viral entry process, the role of NPC1L1 in SARS-CoV-2 entry remains unclear. Our research represents the first-ever demonstration of NPC1L1's involvement in facilitating SARS-CoV-2 entry. The observed role of NPC1L1 in human kidney cells is not only highly intriguing but also quite relevant. This relevance stems from the fact that NPC1L1 exhibits high expression levels in several organs, including the kidneys, and the fact that kidney damages are reported during severe cases of SARS-CoV-2. These findings may help us understand the new functions and mechanisms of NPC1L1 and could contribute to the identification of new antiviral targets.
Collapse
Affiliation(s)
- James Elste
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Nicole Cast
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Shalini Udawatte
- School of Chemistry & Biochemistry, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA;
| | - Kabita Adhikari
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Shannon Harger Payen
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Subhash C. Verma
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois, Chicago, IL 60612, USA;
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| |
Collapse
|
6
|
Jiang W, Jin WL, Xu AM. Cholesterol metabolism in tumor microenvironment: cancer hallmarks and therapeutic opportunities. Int J Biol Sci 2024; 20:2044-2071. [PMID: 38617549 PMCID: PMC11008265 DOI: 10.7150/ijbs.92274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/27/2024] [Indexed: 04/16/2024] Open
Abstract
Cholesterol is crucial for cell survival and growth, and dysregulation of cholesterol homeostasis has been linked to the development of cancer. The tumor microenvironment (TME) facilitates tumor cell survival and growth, and crosstalk between cholesterol metabolism and the TME contributes to tumorigenesis and tumor progression. Targeting cholesterol metabolism has demonstrated significant antitumor effects in preclinical and clinical studies. In this review, we discuss the regulatory mechanisms of cholesterol homeostasis and the impact of its dysregulation on the hallmarks of cancer. We also describe how cholesterol metabolism reprograms the TME across seven specialized microenvironments. Furthermore, we discuss the potential of targeting cholesterol metabolism as a therapeutic strategy for tumors. This approach not only exerts antitumor effects in monotherapy and combination therapy but also mitigates the adverse effects associated with conventional tumor therapy. Finally, we outline the unresolved questions and suggest potential avenues for future investigations on cholesterol metabolism in relation to cancer.
Collapse
Affiliation(s)
- Wen Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, P. R. China
| | - A-Man Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- Anhui Public Health Clinical Center, Hefei 230022, P. R. China
| |
Collapse
|
7
|
Xu C, Fu F, She Y, Xu C. NPC1L1 Plays a Novel Role in Nonalcoholic Fatty Liver Disease. ACS OMEGA 2023; 8:48586-48589. [PMID: 38162748 PMCID: PMC10753569 DOI: 10.1021/acsomega.3c07337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Niemann-Pick C1-Like 1 (NPC1L1) is a key protein in the transport of cholesterol, which exists in the brush marginal membrane of the intestinal epithelial cells and the timid duct membrane of the liver. It affects cholesterol absorption and plasma low-density lipoprotein levels. Cholesterol is both an important component of the cell membrane and a precursor of bile acid and steroid hormone synthesis. Abnormal cholesterol metabolism is closely related to nonalcoholic steatohepatitis (NASH). NASH can progress to fibrosis and cirrhosis, with serious consequences. NPC1L1 is involved in the regulation of cholesterol and lipid metabolism and plays an important role in maintaining the balance of cholesterol metabolism in the body. It also plays an important role in some metabolic diseases such as nonalcoholic fatty liver disease, obesity, and hypercholesterolemia. Therefore, it is necessary to elucidate the molecular pathological mechanism of NPC1L1 in the regulation of cholesterol metabolism and the occurrence and development of NASH, which can provide a target for the development of novel drugs for the treatment of NASH and other diseases. More importantly, it helps to accelerate the development of drugs that regulate lipid metabolism at multiple levels and reduce liver steatosis, which is extremely important for the prevention and treatment of NASH and related severe metabolic diseases.
Collapse
Affiliation(s)
- ChongLi Xu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Fengyang Fu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Yuhan She
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - ChongBo Xu
- School
of Biology and Agriculture, Shaoguan University, Shaoguan 512005, PR China
| |
Collapse
|
8
|
Liu X, Lv M, Zhang W, Zhan Q. Dysregulation of cholesterol metabolism in cancer progression. Oncogene 2023; 42:3289-3302. [PMID: 37773204 DOI: 10.1038/s41388-023-02836-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/18/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
Cholesterol homeostasis has been implicated in the regulation of cellular and body metabolism. Hence, deregulated cholesterol homeostasis leads to the development of many diseases such as cardiovascular diseases, and neurodegenerative diseases, among others. Recent studies have unveiled the connection between abnormal cholesterol metabolism and cancer development. Cholesterol homeostasis at the cellular level dynamically circulates between synthesis, influx, efflux, and esterification. Any dysregulation of this dynamic process disrupts cholesterol homeostasis and its derivatives, which potentially contributes to tumor progression. There is also evidence that cancer-related signals, which promote malignant progression, also regulate cholesterol metabolism. Here, we described the relationship between cholesterol metabolism and cancer hallmarks, with particular focus on the molecular mechanisms, and the anticancer drugs that target cholesterol metabolism.
Collapse
Affiliation(s)
- Xuesong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
- Peking University International Cancer Institute, Beijing, 100191, China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
- Peking University International Cancer Institute, Beijing, 100191, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
- Soochow University Cancer Institute, Suzhou, 215127, China.
| |
Collapse
|
9
|
Qusairy Z, Gangloff A, Leung SOA. Dysregulation of Cholesterol Homeostasis in Ovarian Cancer. Curr Oncol 2023; 30:8386-8400. [PMID: 37754524 PMCID: PMC10527727 DOI: 10.3390/curroncol30090609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/28/2023] Open
Abstract
Cholesterol plays an essential role in maintaining the rigidity of cell membranes and signal transduction. Various investigations confirmed empirically that the dysregulation of cholesterol homeostasis positively correlates with tumor progression. More specifically, recent studies suggested the distinct role of cholesterol in ovarian cancer cell proliferation, metastasis and chemoresistance. In this review, we summarize the current findings that suggest the contribution of cholesterol homeostasis dysregulation to ovarian cancer progression and resistance to anti-cancer agents. We also discuss the therapeutic implications of cholesterol-lowering drugs in ovarian cancer.
Collapse
Affiliation(s)
- Zahraa Qusairy
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Anne Gangloff
- CHU de Québec-Université Laval Research Center, Québec City, QC G1V 4G2, Canada;
- Faculty of Medicine, Laval University, Québec City, QC G1V 0A6, Canada
| | - Shuk On Annie Leung
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
10
|
Ben Hassen C, Goupille C, Vigor C, Durand T, Guéraud F, Silvente-Poirot S, Poirot M, Frank PG. Is cholesterol a risk factor for breast cancer incidence and outcome? J Steroid Biochem Mol Biol 2023; 232:106346. [PMID: 37321513 DOI: 10.1016/j.jsbmb.2023.106346] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Cholesterol plays important roles in many physiological processes, including cell membrane structure and function, hormone synthesis, and the regulation of cellular homeostasis. The role of cholesterol in breast cancer is complex, and some studies have suggested that elevated cholesterol levels may be associated with an increased risk of developing breast cancer, while others have found no significant association. On the other hand, other studies have shown that, for total cholesterol and plasma HDL-associated cholesterol levels, there was inverse association with breast cancer risk. One possible mechanism by which cholesterol may contribute to breast cancer risk is as a key precursor of estrogen. Other potential mechanisms by which cholesterol may contribute to breast cancer risk include its role in inflammation and oxidative stress, which have been linked to cancer progression. Cholesterol has also been shown to play a role in signaling pathways regulating the growth and proliferation of cancer cells. In addition, recent studies have shown that cholesterol metabolism can generate tumor promoters such as cholesteryl esters, oncosterone, 27-hydroxycholesterol but also tumor suppressor metabolites such as dendrogenin A. This review summarizes some of the most important clinical studies that have evaluated the role of cholesterol or its derivatives in breast cancer. It also addresses the role of cholesterol and its derivatives at the cellular level.
Collapse
Affiliation(s)
| | - Caroline Goupille
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; Department of Gynecology, CHRU Hôpital Bretonneau, boulevard Tonnellé, 37044 Tours, France
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Françoise Guéraud
- INRAE, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Philippe G Frank
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; SGS Health and Nutrition, Saint Benoît, France.
| |
Collapse
|
11
|
Wüster J, Heiland M, Nahles S, Preissner R, Preissner S. Statin Medication Improves Five-Year Survival Rates in Patients with Head and Neck Cancer: A Retrospective Case-Control Study of about 100,000 Patients. Cancers (Basel) 2023; 15:3093. [PMID: 37370705 DOI: 10.3390/cancers15123093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION The overall survival among head and neck cancer patients is still low, even in a time of new therapy regimes. Regarding cancer patients' survival, statin use has already proven to be associated with favorable survival outcomes. Our objective was to investigate the influence of statin medication on the overall survival of head and neck cancer patients. METHODS Retrospective clinical data of patients diagnosed with head and neck cancer (International Classification of Diseases codes: C00-C14) were retrieved from a real-world evidence database. The initial cohort was divided into patients with statin medication, who were assigned to building cohort I, and subjects without statin medication, who were assigned to cohort II, both matched by age, gender, and risk factors (nicotine and alcohol abuse/dependence). Subsequently, Kaplan-Meier and risk analyses were performed, and odds and hazard ratios were calculated. RESULTS After matching, each cohort contained 48,626 patients (cohort I = females: 15,409; (31.7%), males 33,212 (68.3%); mean age ± standard deviation (SD) at diagnosis 66.3 ± 11.4 years; cohort II = females: 15,432; (31.7%), males 33,187 (68.2%); mean age ± standard deviation (SD) at diagnosis 66.4 ± 11.5 years). Five-year survival was found to be significantly higher for cohort I, with 75.19%, respectively 70.48% for cohort II. These findings were correlated significantly with a risk of death of 15.9% (cohort I) and 17.2% (cohort II); the odds ratio was 0.91 (95% CI: 0.881-0.942) and the hazard ratio 0.80 (0.777-0.827). CONCLUSIONS The results indicate that the five-year survival of head and neck cancer patients is significantly improved by statin medication. As this study was conducted retrospectively, our data must be interpreted with caution, especially since other potential influencing factors and the initial tumor stage were not available.
Collapse
Affiliation(s)
- Jonas Wüster
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Susanne Nahles
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Robert Preissner
- Institute of Physiology and Science-IT, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstr. 12, 10115 Berlin, Germany
| | - Saskia Preissner
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
12
|
Luo L, Guo Y, Chen L, Zhu J, Li C. Crosstalk between cholesterol metabolism and psoriatic inflammation. Front Immunol 2023; 14:1124786. [PMID: 37234169 PMCID: PMC10206135 DOI: 10.3389/fimmu.2023.1124786] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Psoriasis is a chronic autoinflammatory skin disease associated with multiple comorbidities, with a prevalence ranging from 2 to 3% in the general population. Decades of preclinical and clinical studies have revealed that alterations in cholesterol and lipid metabolism are strongly associated with psoriasis. Cytokines (tumor necrosis factor-α (TNF-α), interleukin (IL)-17), which are important in the pathogenesis of psoriasis, have been shown to affect cholesterol and lipid metabolism. Cholesterol metabolites and metabolic enzymes, on the other hand, influence not only the biofunction of keratinocytes (a primary type of cell in the epidermis) in psoriasis, but also the immune response and inflammation. However, the relationship between cholesterol metabolism and psoriasis has not been thoroughly reviewed. This review mainly focuses on cholesterol metabolism disturbances in psoriasis and their crosstalk with psoriatic inflammation.
Collapse
Affiliation(s)
- Lingling Luo
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Youming Guo
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Lihao Chen
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jing Zhu
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Chengrang Li
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
- Department of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Qin S, Su Q, Li X, Shao M, Zhang Y, Yu F, Ni Y, Zhong J. Curcumin suppresses cell proliferation and reduces cholesterol absorption in Caco-2 cells by activating the TRPA1 channel. Lipids Health Dis 2023; 22:6. [PMID: 36641489 PMCID: PMC9840307 DOI: 10.1186/s12944-022-01750-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Curcumin (Cur) is a bioactive dietary polyphenol of turmeric with various biological activities against several cancers. Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths. Intestinal cholesterol homeostasis is associated with CRC. Chemotherapy for CRC is related to varied adverse effects. Therefore, natural products with anti-cancer properties represent a potential strategy for primary prevention of CRC. METHODS The present study used Cur as a therapeutic approach against CRC using the Caco-2 cell line. The cells were treated with different concentrations of Cur for different duration of time and then the proliferation ability of cells was assessed using Cell Counting Kit-8 and 5-Ethynyl-2'-deoxyuridine assays. Oil red O staining and cholesterol assay kit were used to evaluate cellular lipid content and cholesterol outward transportation. Finally, the protein expressions of cholesterol transport-related protein and signal transduction molecules were assessed using Western blot assay. RESULTS Cur inhibited cell proliferation in Caco-2 cells in a dose- and time-dependent manner by activating the transient receptor potential cation channel subfamily A member 1 (TRPA1) channel. Activation of the TRPA1 channel led to increased intracellular calcium, peroxisome proliferator-activated receptor gamma (PPARγ) upregulation, and the subsequent downregulation of the specificity protein-1 (SP-1)/sterol regulatory element-binding protein-2 (SREBP-2)/Niemann-Pick C1-like 1 (NPC1L1) signaling pathway-related proteins, and finally reduced cholesterol absorption in Caco-2 cells. CONCLUSIONS Cur inhibits cell proliferation and reduces cholesterol absorption in Caco-2 cells through the Ca2+/PPARγ/SP-1/SREBP-2/NPC1L1 signaling by activating the TRPA1 channel, suggesting that Cur can be used as a dietary supplement for the primary prevention of CRC. In Caco-2 cells, Cur first stimulates calcium influx by activating the TRPA1 channel, further upregulates PPARγ and downregulates SP-1/SREBP-2/NPC1L1 signaling pathway, and finally inhibits the absorption of cholesterol. TRPA1, transient receptor potential cation channel subfamily A member 1; NPC1L1, Niemann-Pick C1-like 1; PPARγ, peroxisome proliferator-activated receptor gamma; SP-1, specificity protein-1; SREBP-2, sterol regulatory element-binding protein-2; Cur, curcumin.
Collapse
Affiliation(s)
- Si Qin
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Qian Su
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Xiang Li
- grid.11135.370000 0001 2256 9319College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Muqing Shao
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Yindi Zhang
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Fadong Yu
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Yinxing Ni
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| | - Jian Zhong
- grid.203458.80000 0000 8653 0555Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Branch Road, Chongqing, 401120 China
| |
Collapse
|
14
|
Xia W, Wang H, Zhou X, Wang Y, Xue L, Cao B, Song J. The role of cholesterol metabolism in tumor therapy, from bench to bed. Front Pharmacol 2023; 14:928821. [PMID: 37089950 PMCID: PMC10117684 DOI: 10.3389/fphar.2023.928821] [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: 04/26/2022] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Cholesterol and its metabolites have important biological functions. Cholesterol is able to maintain the physical properties of cell membrane, play an important role in cellular signaling, and cellular cholesterol levels reflect the dynamic balance between biosynthesis, uptake, efflux and esterification. Cholesterol metabolism participates in bile acid production and steroid hormone biosynthesis. Increasing evidence suggests a strict link between cholesterol homeostasis and tumors. Cholesterol metabolism in tumor cells is reprogrammed to differ significantly from normal cells, and disturbances of cholesterol balance also induce tumorigenesis and progression. Preclinical and clinical studies have shown that controlling cholesterol metabolism suppresses tumor growth, suggesting that targeting cholesterol metabolism may provide new possibilities for tumor therapy. In this review, we summarized the metabolic pathways of cholesterol in normal and tumor cells and reviewed the pre-clinical and clinical progression of novel tumor therapeutic strategy with the drugs targeting different stages of cholesterol metabolism from bench to bedside.
Collapse
Affiliation(s)
- Wenhao Xia
- Cancer Center of Peking University Third Hospital, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hao Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Xiaozhu Zhou
- Department of Clinical Pharmacy, School of Pharmacy, Capital Medical University, Beijing, China
| | - Yan Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| | - Baoshan Cao
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| | - Jiagui Song
- Cancer Center of Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University as the Third Responsibility Unit of Song Jiagui, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| |
Collapse
|
15
|
Huang J, Li H, Wang X, Liang X, Zhao T, Hu J, Bai H, Ge J, Sun S, He J. Impacts of ezetimibe on risks of various types of cancers: a meta-analysis and systematic review. Eur J Cancer Prev 2023; 32:89-97. [PMID: 35352704 DOI: 10.1097/cej.0000000000000750] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Ezetimibe is a widely used medication to reduce the plasma cholesterol level, particularly low-density lipoprotein level. However, its impact on cancer remains controversial. Here, its impacts on risks of various types of cancers were meta-analyzed. METHODS PubMed and Cochrane Library electronic databases were searched and randomized controlled trials with followed up for at least 24 weeks were selected and included. The experimental group was defined as those patients treated with ezetimibe alone or with other medications, and the control group was defined as those who received a placebo or the matched medication. The number of new cancer cases or cancer-related deaths was extracted. Statistical analysis was performed using Review Manager (version 5.3). RESULTS Nine trials enrolling 35 222 patients were included in the analyses. Compared with the control group, ezetimibe increased the number of new intestine cancer patients [relative risk (RR), 1.30; 95% confidence interval (CI), 1.02-1.67; P = 0.03] and had a trend to increase the number of new breast cancer patients (RR, 1.39; 95% CI, 0.98-1.98; P = 0.07). There was no significant difference in new hepatobiliary cancer, prostate cancer, skin cancer or cancer of other sites. Ezetimibe did not significantly increase the risk of new cancer in total (RR, 1.03; 95% CI, 0.96-1.11; P = 0.38), cancer-related death (RR, 1.11; 95% CI, 0.98-1.26; P = 0.10) or cancer events (RR, 1.04; 95% CI, 0.97-1.12; P = 0.30). In terms of lipid-lowering effect, ezetimibe significantly reduced total cholesterol and low-density lipoprotein cholesterol, increased high-density lipoprotein cholesterol. CONCLUSION Ezetimibe may increase the risk of intestine cancer and has a trend of increasing the risk of breast cancer. There is no evidence to support that it increases or decreases the risk of other types.
Collapse
Affiliation(s)
- Jing Huang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Huijing Li
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Xueqi Wang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Xi Liang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Tianhe Zhao
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Jingnan Hu
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Haiyan Bai
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Jianli Ge
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Shijiang Sun
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
| | - Jianming He
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, People's Republic of China
| |
Collapse
|
16
|
Wang X, Li Q, Sun S, Liang X, Li H, Huang J, Zhao T, Hu J, Liu J, Hu Z, Duan Y, He J. Network meta-analysis and cost-effectiveness analysis of infliximab, cyclosporine and tacrolimus for ulcerative colitis. Medicine (Baltimore) 2022; 101:e31850. [PMID: 36595876 PMCID: PMC9794301 DOI: 10.1097/md.0000000000031850] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Assess the efficiency and cost-effectiveness of infliximab, cyclosporine and tacrolimus for the treatment of ulcerative colitis (UC). METHODS A literature search identified studies that investigated infliximab, cyclosporine or tacrolimus compared with placebo in UC patients. Short-term, long-term remission rates and response rates were employed to assess efficacy. Odds ratios with 95% confidence intervals were analyzed. A Markov model was constructed to simulate the progression in a cohort of patients with UC, with an over 10 years of time horizon, with a discount rate of 3%, and established threshold of €30,000/quality-adjusted life-year (QALY) or ¥82442/QALY. RESULTS Results of network meta-analysis showed that the order was cyclosporine, tacrolimus, infliximab and placebo from high rate to low with regard to short-term clinical response. The comparison between infliximab versus cyclosporine achieved an incremental cost effectiveness ratio (ICER) of €184435/QALY and ¥531607/QALY, with a 0.34893 QALYs difference of efficacy, and an incremental cost of €64355 and ¥185494. Tacrolimus versus cyclosporine reached an ICER of €44236/QALY and ¥57494/QALY, with a difference of 0.40963 QALYs in efficacy, and a raising cost to €18120 and ¥23551. The probabilistic sensitivity analysis shows that cyclosporine would be cost-effective in the 75.8% of the simulations, tacrolimus in the 24.2%, and infliximab for the 0%. CONCLUSION Infliximab, cyclosporine and tacrolimus as salvage therapies are efficacious. For long-term of clinical remission, the order of pharmacological agents was tacrolimus, infliximab and cyclosporine from high efficacy to low while no significant difference is seen. In cost-effectiveness analysis, the cyclosporine versus infliximab or tacrolimus is expected to be at best.
Collapse
Affiliation(s)
- Xueqi Wang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Qiubo Li
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Shijiang Sun
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xi Liang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Huijing Li
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jing Huang
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Tianhe Zhao
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jingnan Hu
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jianxin Liu
- College of Electronic Countermeasure, National University of Defense Technology, Hefei, Anhui, China
| | - Zhenbiao Hu
- College of Electronic Countermeasure, National University of Defense Technology, Hefei, Anhui, China
| | - Yangyang Duan
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jianming He
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, Hebei, China
- * Correspondence: Jianming He, Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China (e-mails: , )
| |
Collapse
|
17
|
Zhang R, Zeng J, Liu W, Meng J, Wang C, Shi L, Yang S, Chang J, Xing D. The role of NPC1L1 in cancer. Front Pharmacol 2022; 13:956619. [PMID: 36034854 PMCID: PMC9399402 DOI: 10.3389/fphar.2022.956619] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Lipid metabolism appears to play significant roles in the development of cancer. Numerous studies have shown that the evolution of malignancies, including breast, prostate, and colorectal cancers, involves cholesterol in a profound manner. A crucial part in the intestinal absorption of cholesterol is played by Niemann–Pick C1-like 1 (NPC1L1), a cholesterol transporter protein that is widely expressed in the small intestine and liver. The importance of NPC1L1 in tumor prognosis has been demonstrated in investigations in the interim. NPC1L1 also has the potential to develop into a new therapeutic target and a cancer marker. There is, however, no comprehensive review that summarizes NPC1L1’s function in cancer. To this end, we outlined NPC1L1’s functions in carcinogenesis and treatment, along with resources that can be used to further comprehend the connection between NPC1L1 and tumors.
Collapse
Affiliation(s)
- Renshuai Zhang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jun Zeng
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenjing Liu
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingsen Meng
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chao Wang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Lingyu Shi
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shanbo Yang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Chang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongming Xing
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Dongming Xing,
| |
Collapse
|
18
|
Gu J, Zhu N, Li HF, Zhang CJ, Gong YZ, Liao DF, Qin L. Ezetimibe and Cancer: Is There a Connection? Front Pharmacol 2022; 13:831657. [PMID: 35924044 PMCID: PMC9340271 DOI: 10.3389/fphar.2022.831657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
The high level of serum cholesterol caused by the excessive absorption of cholesterol can lead to hypercholesteremia, thus promoting the occurrence and development of cancer. Ezetimibe is a drug that reduces cholesterol absorption and has been widely used for the treatment of patients with high circulating cholesterol levels for many years. Mechanistically, ezetimibe works by binding to NPC1L1, which is a key mediator of cholesterol absorption. Accumulating data from preclinical models have shown that ezetimibe alone could inhibit the development and progression of cancer through a variety of mechanisms, including anti-angiogenesis, stem cell suppression, anti-inflammation, immune enhancement and anti-proliferation. In the past decade, there has been heated discussion on whether ezetimibe combined with statins will increase the risk of cancer. At present, more and more evidence shows that ezetimibe does not increase the risk of cancers, which supports the role of ezetimibe in anti-cancer. In this review, we discussed the latest progress in the anti-cancer properties of ezetimibe and elucidated its underlying molecular mechanisms. Finally, we highlighted the potential of ezetimibe as a therapeutic agent in future cancer treatment and prevention.
Collapse
Affiliation(s)
- Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Yong-Zhen Gong
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
- Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Changsha, China
- Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Li Qin,
| |
Collapse
|
19
|
Xi Y, Yani Z, Jing M, Yinhang W, Xiaohui H, Jing Z, Quan Q, Shuwen H. Mechanisms of induction of tumors by cholesterol and potential therapeutic prospects. Biomed Pharmacother 2021; 144:112277. [PMID: 34624674 DOI: 10.1016/j.biopha.2021.112277] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggested that cholesterol is an important integrant of cell membranes, that plays a key role in tumor progression, immune dysregulation, and pathological changes in epigenetic mechanisms. Based on these theories, there is a growing interest on targeting cholesterol in the treatment of cancer. Here, we comprehensively reviewed the major function of cholesterol on oncogenicity, the therapeutic targets of cholesterol and its metabolites in cancer, and provide detailed insight into the essential roles of cholesterol in mediating immune and epigenetic mechanisms of the tumor microenvironment. It is also worth mentioning that the gut microbiome is an indispensable component of cancer mediation because of its role in cholesterol metabolism. Finally, we summarized recent studies on the potential targets of cholesterol and their metabolism, to provide more therapeutic interventions in oncology.
Collapse
Affiliation(s)
- Yang Xi
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Zhou Yani
- Graduate School of Medical College of Zhejiang University, No. 268 Kaixuan Road, Jianggan District, Hangzhou, Zhejiang 310029, China.
| | - Mao Jing
- Graduate School of Medical College of Zhejiang University, No. 268 Kaixuan Road, Jianggan District, Hangzhou, Zhejiang 310029, China.
| | - Wu Yinhang
- Graduate School of Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China.
| | - Hou Xiaohui
- Graduate School of Nursing, Huzhou University, No. 1 Bachelor Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Zhuang Jing
- Department of Nursing, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Qi Quan
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Han Shuwen
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| |
Collapse
|
20
|
Kwon RJ, Park EJ, Lee SY, Lee Y, Hwang C, Kim C, Cho YH. Expression and prognostic significance of Niemann-Pick C1-Like 1 in colorectal cancer: a retrospective cohort study. Lipids Health Dis 2021; 20:104. [PMID: 34511128 PMCID: PMC8436523 DOI: 10.1186/s12944-021-01539-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/31/2021] [Indexed: 11/10/2022] Open
Abstract
Background Colorectal cancer (CRC) is a malignancy of the large intestine, whose development and prognosis have been demonstrated to be associated with altered lipid metabolism. High cholesterol intake is associated with an increased risk of CRC, and elevated serum cholesterol levels are known to be correlated with risk of developing CRC. Niemann-Pick C1-Like 1 (NPC1L1), a target of ezetimibe, plays an essential role in the absorption of intestinal cholesterol. However, whether the altered expression of NPC1L1 affects CRC development and prognosis is currently unknown. Methods Data corresponding to patients with CRC were obtained from The Cancer Genome Atlas (TCAG). Datasets from the Genome Data Analysis Center (GDAC) platform were analyzed to compare the expression of NPC1L1 in normal and CRC tissues using the Mann–Whitney U test and chi-square test. Further, the datasets from the Gene Expression Omnibus (GEO) database were analyzed. The log-rank test and multivariate Cox proportional hazard regression analysis were performed to determine whether NPC1L1 significantly affects the prognosis of CRC. Results The expression of NPC1L1 was found to be upregulated in CRC and was significantly associated with the N and pathological stages but not with the histological type, age, and sex. Increased NPC1L1 expression in CRC was related to poor patient survival, as evidenced by the Kaplan–Meier and multivariate regression analyses. Conclusions As high expression of NPC1L1 was associated with CRC development, pathological stage, and prognosis, NPC1L1 can serve as an independent prognostic marker for CRC.
Collapse
Affiliation(s)
- Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, 50612, Yangsan, Gyeongsangnam-do, South Korea
| | - Eun-Ju Park
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, 50612, Yangsan, Gyeongsangnam-do, South Korea
| | - Sang Yeoup Lee
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, 50612, Yangsan, Gyeongsangnam-do, South Korea
| | - Youngin Lee
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, 50612, Yangsan, Gyeongsangnam-do, South Korea
| | - Chungsu Hwang
- Department of Pathology, Pusan National University School of Medicine, 626-780, Yangsan, South Korea
| | - Choongrak Kim
- Department of Statistics, Pusan National University, 609-735, Busan, South Korea
| | - Young Hye Cho
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, 50612, Yangsan, Gyeongsangnam-do, South Korea.
| |
Collapse
|
21
|
Rana P, Thai P, Dinh T, Ghosh P. Relevant and Non-Redundant Feature Selection for Cancer Classification and Subtype Detection. Cancers (Basel) 2021; 13:cancers13174297. [PMID: 34503106 PMCID: PMC8428340 DOI: 10.3390/cancers13174297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Here we introduce a new feature selection algorithm DTA, which selects important, non-redundant, and relevant features from diverse omics data. DTA selects non-redundant features by maximizing the similarity between each patient pair by an approximate k-cover algorithm. We successfully applied this algorithm to three different biological problems: (a) disease to healthy sample classification, (b) multiclass classification of different disease samples, and (c) disease subtypes detection. DTA outperformed other feature selection techniques in the binary classification of healthy and disease samples and multiclass classification of various diseases. It also improved the performance of a subtype detection algorithm by selecting the important features for few cancer types. Abstract Biologists seek to identify a small number of significant features that are important, non-redundant, and relevant from diverse omics data. For example, statistical methods such as LIMMA and DEseq distinguish differentially expressed genes between a case and control group from the transcript profile. Researchers also apply various column subset selection algorithms on genomics datasets for a similar purpose. Unfortunately, genes selected by such statistical or machine learning methods are often highly co-regulated, making their performance inconsistent. Here, we introduce a novel feature selection algorithm that selects highly disease-related and non-redundant features from a diverse set of omics datasets. We successfully applied this algorithm to three different biological problems: (a) disease-to-normal sample classification; (b) multiclass classification of different disease samples; and (c) disease subtypes detection. Considering the classification of ROC-AUC, false-positive, and false-negative rates, our algorithm outperformed other gene selection and differential expression (DE) methods for all six types of cancer datasets from TCGA considered here for binary and multiclass classification problems. Moreover, genes picked by our algorithm improved the disease subtyping accuracy for four different cancer types over state-of-the-art methods. Hence, we posit that our proposed feature reduction method can support the community to solve various problems, including the selection of disease-specific biomarkers, precision medicine design, and disease sub-type detection.
Collapse
|
22
|
Giacomini I, Gianfanti F, Desbats MA, Orso G, Berretta M, Prayer-Galetti T, Ragazzi E, Cocetta V. Cholesterol Metabolic Reprogramming in Cancer and Its Pharmacological Modulation as Therapeutic Strategy. Front Oncol 2021; 11:682911. [PMID: 34109128 PMCID: PMC8181394 DOI: 10.3389/fonc.2021.682911] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Cholesterol is a ubiquitous sterol with many biological functions, which are crucial for proper cellular signaling and physiology. Indeed, cholesterol is essential in maintaining membrane physical properties, while its metabolism is involved in bile acid production and steroid hormone biosynthesis. Additionally, isoprenoids metabolites of the mevalonate pathway support protein-prenylation and dolichol, ubiquinone and the heme a biosynthesis. Cancer cells rely on cholesterol to satisfy their increased nutrient demands and to support their uncontrolled growth, thus promoting tumor development and progression. Indeed, transformed cells reprogram cholesterol metabolism either by increasing its uptake and de novo biosynthesis, or deregulating the efflux. Alternatively, tumor can efficiently accumulate cholesterol into lipid droplets and deeply modify the activity of key cholesterol homeostasis regulators. In light of these considerations, altered pathways of cholesterol metabolism might represent intriguing pharmacological targets for the development of exploitable strategies in the context of cancer therapy. Thus, this work aims to discuss the emerging evidence of in vitro and in vivo studies, as well as clinical trials, on the role of cholesterol pathways in the treatment of cancer, starting from already available cholesterol-lowering drugs (statins or fibrates), and moving towards novel potential pharmacological inhibitors or selective target modulators.
Collapse
Affiliation(s)
- Isabella Giacomini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Gianfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, VIMM, Padova, Italy
| | | | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Tommaso Prayer-Galetti
- Department of Surgery, Oncology and Gastroenterology - Urology, University of Padova, Padova, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
23
|
Tajiri K, Sudo T, Ishi K, Kawahara A, Nagasu S, Shimomura S, Yuge K, Katagiri M, Yomoda T, Fujiyoshi K, Kenichi K, Ohchi T, Yoshida T, Mizobe T, Fujita F, Akiba J, Akagi Y. Investigation of clinicopathological characters and gene expression features in colorectal signet-ring cell carcinoma utilizing CMS classification. Mol Clin Oncol 2021; 14:98. [PMID: 33767867 PMCID: PMC7976453 DOI: 10.3892/mco.2021.2260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 02/04/2021] [Indexed: 11/26/2022] Open
Abstract
Signet ring cell carcinoma (SRCC) is a rare pathological type of colorectal cancer, of which the clinicopathological features and genetic background have not yet been fully investigated. Previous research has focused on the optimization of colorectal cancer treatment utilizing consensus molecular subtyping (CMS). However, it is not known what type of CMS would be designated to SRCC treatment. In the current study, of 1,350 patients diagnosed with colorectal cancer who underwent surgery, 14 were diagnosed with SRCC. The case-control cohort that fit the clinical background of the SRCC case was constructed. Statistical comparison between the SRCC group and the case-control cohort was performed among clinicopathological variables. SRCC and well to moderately adenocarcinoma case mRNA were submitted to microarray analysis and CMS analysis. Compared with the case-control cohort, the SRCC group was located more in the right-sided colon, the lymphatic invasion was more severe and the peritoneal dissemination was more frequent. The cancer-specific survival and the progression-free survival were significantly worse in the SRCC group compared with the case-control cohort. Microarray and CMS analysis identified that one SRCC case was significantly well assigned in the CMS 4 group and the other case was assigned in the CMS 1 group. Gene set analysis revealed the upregulation of EMT related genes and the downregulation of fatty acid, glycolysis, differentiation, MYC, HNF4A, DNA repair genes. In conclusion, the clinical characteristics of SRCC are severe but there is a possibility of the presence of different phenotypes according to CMS analysis.
Collapse
Affiliation(s)
- Kensuke Tajiri
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan.,Research Center for Innovative Cancer Therapy, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Tomoya Sudo
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan.,Research Center for Innovative Cancer Therapy, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Kazuo Ishi
- Biostatistics Center, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Sachiko Nagasu
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan.,Research Center for Innovative Cancer Therapy, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Susumu Shimomura
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Kotaro Yuge
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Mitsuhiro Katagiri
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan.,Research Center for Innovative Cancer Therapy, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Takato Yomoda
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Kenji Fujiyoshi
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Koshi Kenichi
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Takafumi Ohchi
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Takefumi Yoshida
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Tomoaki Mizobe
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Fumihiko Fujita
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan.,Research Center for Innovative Cancer Therapy, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
24
|
Jia X, Guo R, Hu Z, Liu J, Liu J, Li B, Yang Q, He J. Efficacy of infliximab, cyclosporine and tacrolimus on ulcerative colitis: A meta-analysis. Medicine (Baltimore) 2020; 99:e22894. [PMID: 33126341 PMCID: PMC7598782 DOI: 10.1097/md.0000000000022894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 07/20/2020] [Accepted: 09/20/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Positioning infliximab (IFX), cyclosporine and tacrolimus (TAC) for treating ulcerative colitis (UC) is in great debate. METHODS A literature search identified studies that investigated IFX vs. cyclosporine or IFX vs TAC in UC patients. Short-term remission, short-term, 1-year and 3-year colectomy rate were employed as primary end-points to assess efficacy. Odds ratios (ORs) with 95% confidence intervals (CIs) were analyzed. RESULTS Overall, 15 studies comprised 596 patients in IFX group and 866 in calcineurin inhibitors group (644 received cyclosporine and 222 received TAC). No significant difference was seen between IFX and calcineurin inhibitors with regard to short-term remission. IFX led to a lower short-term (OR: 0.59, 95% CI: 0.43-0.82, P:.001), 1-year (OR: 0.53, 95% CI: 0.38-0.73, P < .001), 3-year colectomy (OR: 0.41, 95% CI: 0.20-0.84, P:.02) than calcineurin inhibitors. IFX led to a lower short-term (OR: 0.51, 95% CI: 0.36-0.71, P < .001), 1-year (OR: 0.53, 95% CI: 0.37-0.74, P:.003) colectomy and a trend of lower 3-year colectomy (OR: 0.49, 95% CI: 0.22-1.06, P:.07) than cyclosporine while no significant difference was seen between IFX and TAC. Results of network meta-analysis showed that the order was cyclosporine, TAC and IFX from high rate to low with regard to short-term and 1-year colectomy. CONCLUSION IFX treatment leads to a lower short-term, 1-year colectomy rate and a trend of lower 3-year colectomy rate in UC patients than cyclosporine while no significant difference is seen between IFX and TAC. TAC may be superior than cyclosporine with regard to efficacy based on indirect comparisons. Randomized trials with fixed protocol are warranted to identify the optimal medical strategy in patients with UC.
Collapse
Affiliation(s)
- Xuemei Jia
- Department of Gastroenterology, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang
| | - Ruitong Guo
- Department of Gastroenterology, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang
| | - Zhenbiao Hu
- College of Electronic Countermeasure, National University of Defense Technology
| | - Jianxin Liu
- College of Electronic Countermeasure, National University of Defense Technology
| | - Jianping Liu
- Department of Gastroenterology, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang
| | - Bolin Li
- Department of Gastroenterology, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang
| | - Qian Yang
- Department of Gastroenterology, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang
| | - Jianming He
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
25
|
Ding X, Zhang W, Li S, Yang H. The role of cholesterol metabolism in cancer. Am J Cancer Res 2019; 9:219-227. [PMID: 30906624 PMCID: PMC6405981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/14/2019] [Indexed: 06/09/2023] Open
Abstract
Cholesterol plays an important role in cancer development. Both clinical and experimental studies have found that hypercholesterolemia and a high-fat high-cholesterol diet can affect cancer development. External cholesterol can directly activate the oncogenic Hedgehog pathway, and internal cholesterol can induce mTORC1 signaling. Cholesterol is a key component of lipid rafts, which are the major platforms for signaling regulation in cancer, and chelating membrane cholesterol is an effective anti-cancer strategy that disrupts the functions of lipid rafts. Cholesterol metabolism is often reprogrammed in cancer cells. Targeting cholesterol metabolism as a new therapeutic approach has received increasing attention. Here, we summarize some key molecular mechanisms supporting the use of anti-cholesterol therapy for cancer treatment.
Collapse
Affiliation(s)
- Xiao Ding
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical UniversityChongqing, China
| | - Weihua Zhang
- Department of Biology and Biochemistry, College of Natural Sciences and Mathematics, University of HoustonTX, USA
| | - Song Li
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical UniversityChongqing, China
| | - Hui Yang
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical UniversityChongqing, China
| |
Collapse
|
26
|
He J, Zhou J, Yang W, Zhou Q, Liang X, Pang X, Li J, Pan F, Liang H. Dexamethasone affects cell growth/apoptosis/chemosensitivity of colon cancer via glucocorticoid receptor α/NF-κB. Oncotarget 2017; 8:67670-67683. [PMID: 28978062 PMCID: PMC5620202 DOI: 10.18632/oncotarget.18802] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/02/2017] [Indexed: 11/25/2022] Open
Abstract
Glucocorticoids are effective to treat lymphoma and leukemia. Their effect in colon cancer remains far from clear. Here, we found that glucocorticoid receptor (GR) α protein level was dramatically lower in colon cancer than in lymphoma. Colon cell lines LoVo and HCT116 were GRα-rich and GRα was not detectable in HT29 or SW480. Dexamethasone significantly inhibited cell growth of GRα-rich cell lines and did not significantly affect GRα-negative cell lines. Dexamethasone induced apoptosis and increased chemosensitivity of GRα-rich cell lines. Knockdown of GRα significantly attenuated dexamethasone effects on cell growth, apoptosis and chemosensitivity. NF-κB p65 significantly correlated with GRα in colon cancer samples. Dexamethasone decreased NF-κB p65 activity. Knockdown of NF-κB p65 increased apoptosis. Our data demonstrate GRα protein level is dramatically lower in colon cancer than in lymphoma. Dexamethasone inhibits cell growth, induces apoptosis and enhances chemosensitivity in colon cancer, at least partly, via GRα and NF-κB.
Collapse
Affiliation(s)
- Jianming He
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.,Department of Radiotherapy, Hebei Provincial Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Jinming Zhou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Weiwen Yang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Qi Zhou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xi Liang
- Department of Radiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Xueli Pang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Feng Pan
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
27
|
Rodriguez-Broadbent H, Law PJ, Sud A, Palin K, Tuupanen S, Gylfe A, Hänninen UA, Cajuso T, Tanskanen T, Kondelin J, Kaasinen E, Sarin AP, Ripatti S, Eriksson JG, Rissanen H, Knekt P, Pukkala E, Jousilahti P, Salomaa V, Palotie A, Renkonen-Sinisalo L, Lepistö A, Böhm J, Mecklin JP, Al-Tassan NA, Palles C, Martin L, Barclay E, Farrington SM, Timofeeva MN, Meyer BF, Wakil SM, Campbell H, Smith CG, Idziaszczyk S, Maughan TS, Kaplan R, Kerr R, Kerr D, Passarelli MN, Figueiredo JC, Buchanan DD, Win AK, Hopper JL, Jenkins MA, Lindor NM, Newcomb PA, Gallinger S, Conti D, Schumacher F, Casey G, Aaltonen LA, Cheadle JP, Tomlinson IP, Dunlop MG, Houlston RS. Mendelian randomisation implicates hyperlipidaemia as a risk factor for colorectal cancer. Int J Cancer 2017; 140:2701-2708. [PMID: 28340513 PMCID: PMC6135234 DOI: 10.1002/ijc.30709] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/26/2017] [Accepted: 02/08/2017] [Indexed: 01/07/2023]
Abstract
While elevated blood cholesterol has been associated with an increased risk of colorectal cancer (CRC) in observational studies, causality is uncertain. Here we apply a Mendelian randomisation (MR) analysis to examine the potential causal relationship between lipid traits and CRC risk. We used single nucleotide polymorphisms (SNPs) associated with blood levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) as instrumental variables (IV). We calculated MR estimates for each risk factor with CRC using SNP-CRC associations from 9,254 cases and 18,386 controls. Genetically predicted higher TC was associated with an elevated risk of CRC (odds ratios (OR) per unit SD increase = 1.46, 95% confidence interval [CI]: 1.20-1.79, p = 1.68 × 10-4 ). The pooled ORs for LDL, HDL, and TG were 1.05 (95% CI: 0.92-1.18, p = 0.49), 0.94 (95% CI: 0.84-1.05, p = 0.27), and 0.98 (95% CI: 0.85-1.12, p = 0.75) respectively. A genetic risk score for 3-hydoxy-3-methylglutaryl-coenzyme A reductase (HMGCR) to mimic the effects of statin therapy was associated with a reduced CRC risk (OR = 0.69, 95% CI: 0.49-0.99, p = 0.046). This study supports a causal relationship between higher levels of TC with CRC risk, and a further rationale for implementing public health strategies to reduce the prevalence of hyperlipidaemia.
Collapse
Affiliation(s)
| | - Philip J. Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Kimmo Palin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Sari Tuupanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Alexandra Gylfe
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Ulrika A. Hänninen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Tatiana Cajuso
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Tomas Tanskanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Johanna Kondelin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Eevi Kaasinen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
| | - Johan G. Eriksson
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Folkhälsan Research Centre, Helsinki, 00250, Finland
- Unit of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, 00014, Finland
| | - Harri Rissanen
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Paul Knekt
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Eero Pukkala
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, 00130, Finland
- School of Health Sciences, University of Tampere, Tampere, 33014, Finland
| | - Pekka Jousilahti
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Laura Renkonen-Sinisalo
- Abdominal Center, Department of Surgery, Helsinki University Hospital, Helsinki, 00029, Finland
| | - Anna Lepistö
- Abdominal Center, Department of Surgery, Helsinki University Hospital, Helsinki, 00029, Finland
| | - Jan Böhm
- Department of Pathology, Central Finland Central Hospital, Jyväskylä, 40620, Finland
| | - Jukka-Pekka Mecklin
- Department of Surgery, Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, 40620, Finland
| | - Nada A. Al-Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Claire Palles
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Lynn Martin
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Ella Barclay
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Susan M. Farrington
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Maria N. Timofeeva
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Brian F. Meyer
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Salma M. Wakil
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Christopher G. Smith
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Shelley Idziaszczyk
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Timothy S. Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Richard Kaplan
- MRC Clinical Trials Unit, Aviation House, London, WC2B 6NH, UK
| | - Rachel Kerr
- Oxford Cancer Centre, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - David Kerr
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Michael N. Passarelli
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Jane C. Figueiredo
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Aung K. Win
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - John L. Hopper
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Mark A. Jenkins
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Noralane M. Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Polly A. Newcomb
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - David Conti
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Fred Schumacher
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lauri A. Aaltonen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Jeremy P. Cheadle
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ian P. Tomlinson
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| |
Collapse
|
28
|
He J, Pei L, Jiang H, Yang W, Chen J, Liang H. Chemoresistance of colorectal cancer to 5-fluorouracil is associated with silencing of the BNIP3 gene through aberrant methylation. J Cancer 2017; 8:1187-1196. [PMID: 28607593 PMCID: PMC5463433 DOI: 10.7150/jca.18171] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022] Open
Abstract
Purpose To investigate the correlation between chemoresistance of colorectal cancer to 5-fluorouracil and BNIP3 and the underlying mechanism. Methods BNIP3 protein in specimens was evaluated using immunohistochemistry. Semi-quantitative reverse transcription PCR and Western blot was employed to assay gene expression. The promoter methylation status of BNIP3 was examined by methylation-specific PCR. Drug sensitivity was assayed using MTT assay. Results Specimens from 81 patients with colorectal cancer receiving 5-fluorouracil-based chemotherapy were analyzed. BNIP3 expression was negative in 42 cancer samples. The mean score of BNIP3 in cancer was 1.8±0.2 and it was 3.7±0.5 in adjacent colorectum (p<0.05). The response rate of the BNIP3 positive group was 63.6% and that of the negative group was 36.4% (p=0.021). The median PFS of the BNIP3 positive group was 9.25 months and that of the BNIP3 negative group was 6.5 months (p=0.011). BNIP3 mRNA was not detectable in 4 of 8 colorectal cell lines and all these 4 cell lines displayed BNIP3 methylated allele only. Other 4 cell lines what expressed detectable BNIP3 displayed BNIP3 unmethylated allele only or both unmethylated and methylated alleles. 5-Aza dramatically increased BNIP3 expression. Knockdown of DNMT1 increased BNIP3. Knockdown of DNMT3B alone did not detectably change BNIP3 expression while knockdown of both DNMT1 and DNMT3B increased BNIP3 expression more than knockdown of DNMT1 alone. Knockdown of BNIP3 decreased chemosensitivity to 5-fluorouracil and increasing BNIP3 through demethylation increased chemosensitivity. Conclusion Chemoresistance of colorectal cancer to 5-fluorouracil is associated with silencing of the BNIP3 gene through aberrant methylation via DNMT1/DNMT3B.
Collapse
Affiliation(s)
- Jianming He
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China.,Department Of Oncology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang 050011, China
| | - Li Pei
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Heng Jiang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Weiwen Yang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Jianfang Chen
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Houjie Liang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| |
Collapse
|
29
|
He J, Wang F, Luo F, Chen X, Liang X, Jiang W, Huang Z, Lei J, Shan F, Xu X. Effects of long term low- and high-dose sodium arsenite exposure in human transitional cells. Am J Transl Res 2017; 9:416-428. [PMID: 28337271 PMCID: PMC5340678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 01/30/2017] [Indexed: 06/06/2023]
Abstract
Epidemiological studies have revealed the association between increased risk of bladder cancer and chronic arsenic exposure. Here, we explored biological effects of arsenic in T24. Microarray analysis was applied to analyze mRNA in T24 following 0, 2 or 5 μM sodium arsenite (As) exposure for 72 hours. Long term (up to 140 days) low-dose (200 nM) and high-dose (1,000 nM) As decreased E-cadherin protein level through different mechanisms because the mRNA levels of E-cadherin increased following low-dose As exposure but decreased following high-dose As exposure. Long term As increased the protein levels of N-cadherin, vimentin, β-catenin, and slug. Low-dose As exposure resulted in a change in the morphology of T24 cells from an epithelial to a mesenchymal-like appearance. Knockdown of E-cadherin increased the protein levels of N-cadherin, vimentin, β-catenin, and slug. Cell proliferation and growth of T24 with or without As exposure for 100 days were assayed using EdU and WST, respectively. Low-dose As exposure increased cell proliferation and growth while high-dose As exposure decreased both. Long term As activated p53 on account of increasing protein levels of p53, p-p53 (Ser15), and mRNA levels of p21. These demonstrate that arsenic exposure exerts multiple effects. Long term low- or high-dose arsenic induces epithelial-mesenchymal transition, likely via downregulation of E-cadherin, activates p53, and differently affects cell proliferation/growth.
Collapse
Affiliation(s)
- Jianming He
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
- Department of Oncology, Hebei Provincial Hospital of Traditional Chinese MedicineShijiazhuang 050011, Hebei Province, China
| | - Feng Wang
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Fen Luo
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Xuedan Chen
- Department of Medical Genetics, Third Military Medical UniversityChongqing 400038, China
| | - Xi Liang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
- Department of Radiology, Hebei Provincial Hospital of Traditional Chinese MedicineShijiazhuang 050011, Hebei Province, China
| | - Wenbin Jiang
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Zhizhong Huang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Jiafan Lei
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Fabo Shan
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Xueqing Xu
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| |
Collapse
|
30
|
Bae JS, Park JM, Lee J, Oh BC, Jang SH, Lee YB, Han YM, Ock CY, Cha JY, Hahm KB. Amelioration of non-alcoholic fatty liver disease with NPC1L1-targeted IgY or n-3 polyunsaturated fatty acids in mice. Metabolism 2017; 66:32-44. [PMID: 27923447 DOI: 10.1016/j.metabol.2016.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 02/06/2023]
Abstract
Patients with non-alcoholic fatty liver disease (NAFLD) have an increased risk for progression to hepatocellular carcinoma in addition to comorbidities such as cardiovascular and serious metabolic diseases; however, the current therapeutic options are limited. Based on our previous report that omega-3 polyunsaturated fatty acids (n-3 PUFAs) can significantly ameliorate high fat diet (HFD)-induced NAFLD, we explored the therapeutic efficacy of n-3 PUFAs and N-IgY, which is a chicken egg yolk-derived IgY specific for the Niemann-Pick C1-Like 1 (NPC1L1) cholesterol transporter, on NAFLD in mice. We generated N-IgY and confirmed its efficient cholesterol transport-blocking activity in HepG2 and Caco-2 cells, which was comparable to the effect of ezetimibe (EZM). C57BL/6 wild type and fat-1 transgenic mice, capable of producing n-3 PUFAs, were fed a high fat diet (HFD) alone or supplemented with N-IgY. Endogenously synthesized n-3 PUFAs combined with N-IgY led to significant decreases in hepatic steatosis, fibrosis, and inflammation (p<0.01). The combination of N-IgY and n-3 PUFAs resulted in significant upregulation of genes involved in cholesterol uptake (LDLR), reverse cholesterol transport (ABCG5/ABCG8), and bile acid metabolism (CYP7A1). Moreover, fat-1 transgenic mice treated with N-IgY showed significant downregulation of genes involved in cholesterol-induced hepatic stellate cell activation (Tgfb1, Tlr4, Col1a1, Col1a2, and Timp2). Collectively, these data suggest that n-3 PUFAs and N-IgY, alone or in combination, represent a promising treatment strategy to prevent HFD-induced fatty liver through the activation cholesterol catabolism to bile acids and by decreasing cholesterol-induced fibrosis.
Collapse
Affiliation(s)
- Jin-Sik Bae
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Min Park
- CHA Cancer Prevention Research Center, CHA Bio Complex, CHA University, Seongnam, Gyunggi-do, 13488, Republic of Korea
| | - Junghoon Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Byung-Chul Oh
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sang-Ho Jang
- Bioceltrand Co., Chuncheon, Gangwon-do, 200161, Republic of Korea
| | - Yun Bin Lee
- CHA Cancer Prevention Research Center, CHA Bio Complex, CHA University, Seongnam, Gyunggi-do, 13488, Republic of Korea; Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Gyunggi-do, 13496, Republic of Korea
| | - Young-Min Han
- CHA Cancer Prevention Research Center, CHA Bio Complex, CHA University, Seongnam, Gyunggi-do, 13488, Republic of Korea
| | - Chan-Young Ock
- CHA Cancer Prevention Research Center, CHA Bio Complex, CHA University, Seongnam, Gyunggi-do, 13488, Republic of Korea
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; Gachon Medical Research Institute, Gil Hospital, Incheon 21565, Republic of Korea.
| | - Ki-Baik Hahm
- CHA Cancer Prevention Research Center, CHA Bio Complex, CHA University, Seongnam, Gyunggi-do, 13488, Republic of Korea; Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Gyunggi-do, 13496, Republic of Korea.
| |
Collapse
|
31
|
He J, Liang X, Luo F, Chen X, Xu X, Wang F, Zhang Z. P53 Is Involved in a Three-Dimensional Architecture-Mediated Decrease in Chemosensitivity in Colon Cancer. J Cancer 2016; 7:900-9. [PMID: 27313779 PMCID: PMC4910581 DOI: 10.7150/jca.14506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/16/2016] [Indexed: 12/26/2022] Open
Abstract
Three-dimensional (3D) culture models represent a better approximation of solid tumor tissue architecture, especially cell adhesion, in vivo than two-dimensional (2D) cultures do. Here, we explored the role of architecture in chemosensitivity to platinum in colon cancer. Under the 3D culture condition, colon cancer cells formed multicellular spheroids, consisting of layers of cells. 3D cultures displayed significantly decreased sensitivity to platinum compared with 2D cultures. Platinum increased p53 in a dose-dependent and time-dependent manner. There was no detectable difference in basal p53 levels between 3D cultures and 2D cultures but cisplatin induced less p53 in both HCT116 3D cultures and LoVo 3D cultures. It was not due to cisplatin concentration because cisplatin induced similar γ-H2AX in 3D vs 2D. Knockdown of p53 significantly decreased sensitivity to platinum in 3D cultures. Knockdown of p53 decreased cleaved caspase 3 and apoptosis induced by cisplatin. These findings indicate that 3D architecture confers decreased chemosensitivity to platinum and p53 is involved in the mechanism. Knockdown of p53 decreased cisplatin's induction of c-Jun N-terminal kinase 1/2 (JNK1/2) activation, whereas inhibition of JNK1/2 activation increased chemosensitivity. Inhibition of p38 activation decreased cisplatin's induction of p53, but no difference in p38 activation by cisplatin was observed between 2D cultures and 3D cultures. Taken together, our results suggest that p53 is involved in a 3D architecture-mediated decrease in chemosensitivity to platinum in colon cancer. Mitogen-activated protein kinases (JNK1/2 and p38) do not play a dominant role in the mechanism.
Collapse
Affiliation(s)
- Jianming He
- 1. Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Xi Liang
- 1. Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China;; 2. Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Fen Luo
- 2. Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Xuedan Chen
- 3. Department Of Medical Genetics, Third Military Medical University, Chongqing, 400038 China
| | - Xueqing Xu
- 2. Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Fengchao Wang
- 4. Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Zhenping Zhang
- 5. Department Of Oncology, First Hospital of Shijiazhuang City, Shijiazhuang, Hebei Province, 050011 China
| |
Collapse
|
32
|
Rai S, Bhatnagar S. Hyperlipidemia, Disease Associations, and Top 10 Potential Drug Targets: A Network View. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:152-68. [DOI: 10.1089/omi.2015.0172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sneha Rai
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| |
Collapse
|
33
|
LIANG XI, XU XUEQING, WANG FENGCHAO, CHEN XUEDAN, LI NI, WANG CANCAN, HE JIANMING. E-cadherin knockdown increases β-catenin reducing colorectal cancer chemosensitivity only in three-dimensional cultures. Int J Oncol 2015; 47:1517-27. [DOI: 10.3892/ijo.2015.3137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/11/2015] [Indexed: 11/05/2022] Open
|
34
|
Liang X, Xu X, Wang F, Li N, He J. E-cadherin increasing multidrug resistance protein 1 via hypoxia-inducible factor-1α contributes to multicellular resistance in colorectal cancer. Tumour Biol 2015. [PMID: 26219897 DOI: 10.1007/s13277-015-3811-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
When cancer cells have been cultured as three-dimensional (3D) cultures or in vivo, they decrease sensitivity to anticancer drugs. This is called multicellular resistance, and the mechanism is not fully understood. Here, we report that E-cadherin increasing multidrug resistance protein 1 (MDR1) via hypoxia-inducible factor-1α (HIF-1α) contributes to multicellular resistance in colorectal cancer. The MDR1 protein level was higher in 3D cultures than in monolayer cells. When dispersed cells from 3D cultures were grown as monolayer cells again, the MDR1 protein level decreased to the similar level of cells maintained as monolayer all through. Knockdown of MDR1 significantly decreased multicellular resistance. Knockdown of E-cadherin decreased MDR1 in 3D cultures but did not detectably change MDR1 in monolayer cells. E-cadherin was expressed uniformly in 3D cultures while the MDR1 protein level was higher in the center of 3D cultures than in the peripheral part. Knockdown of E-cadherin decreased E-cadherin uniformly in 3D cultures but mainly decreased MDR1 at the center of 3D cultures. These suggest that knockdown of E-cadherin decreasing MDR1 may be by an indirect mechanism. HIF-1α was remarkably increased in 3D cultures. Knockdown of E-cadherin decreased intercellular junctions, increased intercellular space, and decreased HIF-1α in 3D cultures. Knockdown of HIF-1α decreased MDR1 in 3D cultures. Knockdown of E-cadherin increased β-catenin uniformly in 3D cultures, and knockdown of β-catenin decreased MDR1 what was opposite to knockdown of E-cadherin decreasing MDR1. Our data reveal that knockdown of E-cadherin decreasing MDR1 via HIF-1α is involved in the mechanism of multicellular resistance in colorectal cancer. Though β-catenin is also involved in the mechanism, it does not play a dominant role.
Collapse
Affiliation(s)
- Xi Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.,Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xueqing Xu
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Fengchao Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Ni Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jianming He
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|