1
|
Šrámek J, Němcová-Fürstová V, Kovář J. Molecular Mechanisms of Apoptosis Induction and Its Regulation by Fatty Acids in Pancreatic β-Cells. Int J Mol Sci 2021; 22:4285. [PMID: 33924206 PMCID: PMC8074590 DOI: 10.3390/ijms22084285] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic β-cell failure and death contribute significantly to the pathogenesis of type 2 diabetes. One of the main factors responsible for β-cell dysfunction and subsequent cell death is chronic exposure to increased concentrations of FAs (fatty acids). The effect of FAs seems to depend particularly on the degree of their saturation. Saturated FAs induce apoptosis in pancreatic β-cells, whereas unsaturated FAs are well tolerated and are even capable of inhibiting the pro-apoptotic effect of saturated FAs. Molecular mechanisms of apoptosis induction by saturated FAs in β-cells are not completely elucidated. Saturated FAs induce ER stress, which in turn leads to activation of all ER stress pathways. When ER stress is severe or prolonged, apoptosis is induced. The main mediator seems to be the CHOP transcription factor. Via regulation of expression/activity of pro- and anti-apoptotic Bcl-2 family members, and potentially also through the increase in ROS production, CHOP switches on the mitochondrial pathway of apoptosis induction. ER stress signalling also possibly leads to autophagy signalling, which may activate caspase-8. Saturated FAs activate or inhibit various signalling pathways, i.e., p38 MAPK signalling, ERK signalling, ceramide signalling, Akt signalling and PKCδ signalling. This may lead to the activation of the mitochondrial pathway of apoptosis, as well. Particularly, the inhibition of the pro-survival Akt signalling seems to play an important role. This inhibition may be mediated by multiple pathways (e.g., ER stress signalling, PKCδ and ceramide) and could also consequence in autophagy signalling. Experimental evidence indicates the involvement of certain miRNAs in mechanisms of FA-induced β-cell apoptosis, as well. In the rather rare situations when unsaturated FAs are also shown to be pro-apoptotic, the mechanisms mediating this effect in β-cells seem to be the same as for saturated FAs. To conclude, FA-induced apoptosis rather appears to be preceded by complex cross talks of multiple signalling pathways. Some of these pathways may be regulated by decreased membrane fluidity due to saturated FA incorporation. Few data are available concerning molecular mechanisms mediating the protective effect of unsaturated FAs on the effect of saturated FAs. It seems that the main possible mechanism represents a rather inhibitory intervention into saturated FA-induced pro-apoptotic signalling than activation of some pro-survival signalling pathway(s) or metabolic interference in β-cells. This inhibitory intervention may be due to an increase of membrane fluidity.
Collapse
Affiliation(s)
- Jan Šrámek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | - Vlasta Němcová-Fürstová
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | | |
Collapse
|
2
|
Wang H, Wu W, Wang G, Xu W, Zhang F, Wu B, Tian Y. Protective effect of ginsenoside Rg3 on lung injury in diabetic rats. J Cell Biochem 2018; 120:3323-3330. [PMID: 30362612 DOI: 10.1002/jcb.27601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/07/2018] [Indexed: 11/07/2022]
Abstract
Ginsenoside has been used to treat diabetes, while ginsenoside Rg3 is the main active ingredient component of ginseng and is used to study its effects on lung tissue damage in diabetic rats. In this paper, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry were applied to detect the proliferation and apoptosis of BEAS-2B cells treated with different concentrations of Rg3. The inflammatory response and pathological change in the lung tissue of diabetic rats treated with Rg3 were evaluated by enzyme-linked immunosorbent assay, quantative real-time polymerase chain reaction, and hematoxylin and eosin staining immunohistochemistry. Meanwhile, PI3K and MAPK signaling pathway proteins in lung tissue were determined by Western blot analysis. The results showed that ginsenoside Rg3 had no significant influence on the proliferation and apoptosis of BEAS-2B cells. Ginsenoside Rg3 can inhibit inflammatory response and promote the activation of PI3K and MAPK signaling pathways to prevent damages of lung tissues induced by hyperglycemia. The protective effect provided by ginsenoside Rg3 indicates that ginsenoside Rg3 is a potential drug for the treatment of diabetes.
Collapse
Affiliation(s)
- Heyuan Wang
- Departments of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China.,Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Wu
- Departments of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Departments of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Wenzhou Xu
- Departments of Stomatology, Stomatological Hospital of Jilin University, Changchun, Jilin Province, China
| | - Fuqiang Zhang
- Departments of Science and Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bing Wu
- Departments of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.,Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China
| | - Yaping Tian
- Department of Dermatology and Venerology of the First Hospital, Jilin University, Changchun, 130021, China
| |
Collapse
|
3
|
Xu H, Wang Q, Sun Q, Qin Y, Han A, Cao Y, Yang Q, Yang P, Lu J, Liu Q, Xiang Q. In type 2 diabetes induced by cigarette smoking, activation of p38 MAPK is involved in pancreatic β-cell apoptosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:9817-9827. [PMID: 29372523 DOI: 10.1007/s11356-018-1337-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Abstract
Type 2 diabetes (T2D) is a chronic disease caused by pancreatic β-cell dysfunction and insulin resistance. Exposure to smoke is a risk factor for diabetes; however, its mechanisms are unclear. In an epidemiological study, we determined the relationship between cigarette smoking and β-cell function. T2D patients had a history of heavier smoking than people without T2D, and heavy smokers had more abnormal glucose metabolism. For various smoking populations, there was a dose-effect relationship between decreases of homeostatic model assessment (HOMA)-β levels or the increases of HOMA-insulin resistance (IR) levels and amount of smoking (pack-years), which indicated that smoking induced β-cell dysfunction. For MIN6 cells, cigarette smoke extract (CSE) decreased insulin secretion and content; enhanced apoptosis, as illustrated by decreases of BCL-2 levels, increases of BAX and cleaved caspase-3 levels, and an increased apoptotic index; and activated the p38 MAPK pathway. For MIN6 cells, inhibition of p-p38 MAPK by SB203580 prevented enhanced apoptosis and the dysfunction of insulin secretion induced by CSE. In sum, activation of p38 MAPK is involved in the apoptosis of pancreatic β-cells induced by cigarette smoking, which is a possible mechanism for induction of T2D by cigarette smoke.
Collapse
Affiliation(s)
- Hui Xu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Qiushi Wang
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Qian Sun
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Yu Qin
- Institute of Chronic Non-Communicable Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Aohan Han
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Ye Cao
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Qianlei Yang
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Ping Yang
- School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Jiachun Lu
- School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Qizhan Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| | - Quanyong Xiang
- Institute of Chronic Non-Communicable Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Energy depletion and not ROS formation is a crucial step of glucolipotoxicity (GLTx) in pancreatic beta cells. Pflugers Arch 2017; 470:537-547. [PMID: 29218453 DOI: 10.1007/s00424-017-2094-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/30/2017] [Indexed: 12/27/2022]
Abstract
We have shown previously that genetic or pharmacological deletion of KATP channels protect against beta cell dysfunction induced by reactive oxygen species (ROS). Since it is assumed that glucolipotoxicity (GLTx) causes ROS production, we aimed to evaluate whether suppression of KATP channel activity can also prevent beta cell damage evoked by GLTx. We used an in vitro model of GLTx and measured distinct parameters of stimulus-secretion coupling. GLTx gradually induced disturbances of Ca2+ oscillations over 3 days. This impairment in Ca2+ dynamics was partially reversed in beta cells without functional KATP channels (SUR1-/-) and by the sulfonylurea gliclazide but not by tolbutamide. By contrast, the GLTx-induced suppression of glucose-induced insulin secretion could not be rescued by decreased KATP channel activity pointing to a direct interaction of GLTx with the secretory capacity. Accordingly, GLTx also suppressed KCl-induced insulin secretion. GLTx was not accompanied by decisively increased ROS production or enhanced apoptosis. Insulin content of beta cells was markedly reduced by GLTx, an effect not prevented by gliclazide. Since GLTx markedly diminished the mitochondrial membrane potential and cellular ATP content, lack of ATP is assumed to decrease insulin biosynthesis. The deleterious effect of GLTx is therefore caused by direct interference with the secretory capacity whereby reduction of insulin content is one important parameter. These findings deepen our understanding how GLTx damages beta cells and reveal that GLTx is disconnected from ROS formation, a notion important for targeting beta cells in the treatment of diabetes. Overall, GLTx-induced energy depletion may be a primary step in the cascade of events leading to loss of beta cell function in type-2 diabetes mellitus.
Collapse
|
5
|
Triñanes J, Rodriguez-Rodriguez AE, Brito-Casillas Y, Wagner A, De Vries APJ, Cuesto G, Acebes A, Salido E, Torres A, Porrini E. Deciphering Tacrolimus-Induced Toxicity in Pancreatic β Cells. Am J Transplant 2017; 17:2829-2840. [PMID: 28432716 DOI: 10.1111/ajt.14323] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 04/14/2017] [Accepted: 04/14/2017] [Indexed: 01/25/2023]
Abstract
β Cell transcription factors such as forkhead box protein O1 (FoxO1), v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA), pancreatic and duodenal homeobox 1, and neuronal differentiation 1, are dysfunctional in type 2 diabetes mellitus (T2DM). Posttransplant diabetes mellitus resembles T2DM and reflects interaction between pretransplant insulin resistance and immunosuppressants, mainly calcineurin inhibitors (CNIs). We evaluated the effect of tacrolimus (TAC), cyclosporine A (CsA), and metabolic stressors (glucose plus palmitate) on insulinoma β cells in vitro and in pancreata of obese and lean Zucker rats. Cells were cultured for 5 days with 100 μM palmitate and 22 mM glucose; CsA (250 ng/mL) or TAC (15 ng/mL) were added in the last 48 h. Glucose plus palmitate increased nuclear FoxO1 and decreased nuclear MafA. TAC in addition to glucose plus palmitate magnified these changes in nuclear factors, whereas CsA did not. In addition to glucose plus palmitate, both drugs reduced insulin content, and TAC also affected insulin secretion. TAC withdrawal or conversion to CsA restored these changes. Similar results were observed in pancreata of obese animals on CNIs. TAC and CsA, in addition to glucose plus palmitate, induced comparable inhibition of calcineurin and nuclear factor of activated T cells (NFAT); therefore, TAC potentiates glucolipotoxicity in β cells, possibly by sharing common pathways of β cell dysfunction. TAC-induced β cell dysfunction is potentially reversible. Inhibition of the calcineurin-NFAT pathway may contribute to the diabetogenic effect of CNIs but does not explain the stronger effect of TAC compared with CsA.
Collapse
Affiliation(s)
- J Triñanes
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain.,Division of Nephrology and Leiden Transplant Center, Leiden University Medical Center and Leiden University, Leiden, the Netherlands
| | | | - Y Brito-Casillas
- Unit of Endocrinology and Nutrition, Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - A Wagner
- Unit of Endocrinology and Nutrition, Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - A P J De Vries
- Division of Nephrology and Leiden Transplant Center, Leiden University Medical Center and Leiden University, Leiden, the Netherlands
| | - G Cuesto
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain
| | - A Acebes
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain
| | - E Salido
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain.,Pathology Department, Hospital Universitario de Canarias, La Laguna, Tenerife, Spain.,Centre for Biomedical Research on Rare Diseases (CIBERER), La Laguna, Tenerife, Spain
| | - A Torres
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain.,Research Unit of the University Hospital of the Canary Islands, La Laguna, Tenerife, Spain.,Nephrology Department, Hospital Universitario de Canarias, La Laguna, Tenerife, Spain
| | - E Porrini
- Centre for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna, La Laguna, Tenerife, Spain
| |
Collapse
|
6
|
Kim YJ, Park SM, Jung HS, Lee EJ, Kim TK, Kim TN, Kwon MJ, Lee SH, Rhee BD, Kim MK, Park JH. Ginsenoside Rg3 prevents INS-1 cell death from intermittent high glucose stress. Islets 2016; 8:57-64. [PMID: 27246809 PMCID: PMC4987017 DOI: 10.1080/19382014.2016.1161874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/05/2016] [Accepted: 02/24/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ginsenoside Rg3 has been proposed to mediate anti-diabetic effects, but their direct effect on pancreatic β cell viability and mechanisms are not clearly understood. Recent studies suggest that intermittent high glucose (IHG) could be more harmful to pancreatic β cells than sustained high glucose. There are few reports about the effect of the ginsenosideRg3 to β cell apoptosis and proliferation against IHG. METHODS INS-1 cells were treated with alternative glucose concentration with or without ginsenoside Rg3. Cell apoptosis and viability were detected by Annexin V staining and MTT assay. The activation of mitogen-activated protein kinases (MAPKs) was analyzed by Western blotting using specific antibodies. Quantification of secreted insulin protein was measured using rat/mouse Insulin ELISA kits. Bromodeoxyuridine (BrdU) staining and florescence in situ hybridization (FISH) analysis was performed to compare cell proliferation. RESULT INS-1 cell viability was decreased under IHG and increased with Rg3 treatment.Rg3 significantly reduced the apoptotic INS-1 cells against IHG. The quantification of secreted insulin concentration was increased with Rg3. Rg3 increased INS-1 cell proliferation. ERK and p38 MAPK pathways reduced by IHG were activated by the ginsenoside Rg3. CONCLUSION Ginsenoside Rg3 protected INS-1 cell death from IHG with reducing apoptosis and increasing proliferation.
Collapse
Affiliation(s)
- You Jeong Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Maryknoll Medical Center, Busan, South Korea
| | - Su Min Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Hye Sook Jung
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| | - Eun Ju Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Tae Kyoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Tae-Nyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Min Jeong Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Soon Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Byoung Doo Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
| | - Mi-kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| | - Jeong Hyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, South Korea
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, South Korea
| |
Collapse
|
7
|
Bo J, Xie S, Guo Y, Zhang C, Guan Y, Li C, Lu J, Meng QH. Methylglyoxal Impairs Insulin Secretion of Pancreatic β-Cells through Increased Production of ROS and Mitochondrial Dysfunction Mediated by Upregulation of UCP2 and MAPKs. J Diabetes Res 2016; 2016:2029854. [PMID: 26779540 PMCID: PMC4686727 DOI: 10.1155/2016/2029854] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 06/12/2015] [Accepted: 07/01/2015] [Indexed: 01/07/2023] Open
Abstract
Methylglyoxal (MG) is a highly reactive glucose metabolic intermediate and a major precursor of advanced glycation end products. MG level is elevated in hyperglycemic disorders such as diabetes mellitus. Substantial evidence has shown that MG is involved in the pathogenesis of diabetes and diabetic complications. We investigated the impact of MG on insulin secretion by MIN6 and INS-1 cells and the potential mechanisms of this effect. Our study demonstrates that MG impaired insulin secretion by MIN6 or ISN-1 cells in a dose-dependent manner. It increased reactive oxygen species (ROS) production and apoptosis rate in MIN6 or ISN-1 cells and inhibited mitochondrial membrane potential (MMP) and ATP production. Furthermore, the expression of UCP2, JNK, and P38 as well as the phosphorylation JNK and P38 was increased by MG. These effects of MG were attenuated by MG scavenger N-acetyl cysteine. Collectively, these data indicate that MG impairs insulin secretion of pancreatic β-cells through increasing ROS production. High levels of ROS can damage β-cells directly via JNK/P38 upregulation and through activation of UCP2 resulting in reduced MMP and ATP production, leading to β-cell dysfunction and impairment of insulin production.
Collapse
Affiliation(s)
- Jinshuang Bo
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Shiya Xie
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Yi Guo
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Chunli Zhang
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Yanming Guan
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Chunmei Li
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
| | - Jianxin Lu
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, Zhejiang 325035, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, Zhejiang 325035, China
| | - Qing H. Meng
- Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Wenzhou, Zhejiang 325035, China
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- *Qing H. Meng:
| |
Collapse
|
8
|
Nagayama K, Morino K, Sekine O, Nakagawa F, Ishikado A, Iwasaki H, Okada T, Tawa M, Sato D, Imamura T, Nishio Y, Ugi S, Kashiwagi A, Okamura T, Maegawa H. Duality of n-3 Polyunsaturated Fatty Acids on Mcp-1 Expression in Vascular Smooth Muscle: A Potential Role of 4-Hydroxy Hexenal. Nutrients 2015; 7:8112-26. [PMID: 26402697 PMCID: PMC4586576 DOI: 10.3390/nu7095381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/24/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023] Open
Abstract
N-3 polyunsaturated fatty acids such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) have protective effects against atherosclerosis. Monocyte chemotactic protein (MCP)-1 is a major inflammatory mediator in the progression of atherosclerosis. However, little is known about the regulation of MCP-1 by DHA and EPA in vessels and vascular smooth muscle cells (VSMCs). In this study, we compared the effect of DHA and EPA on the expression of Mcp-1 in rat arterial strips and rat VSMCs. DHA, but not EPA, suppressed Mcp-1 expression in arterial strips. Furthermore, DHA generated 4-hydroxy hexenal (4-HHE), an end product of n-3 polyunsaturated fatty acids (PUFAs), in arterial strips as measured by liquid chromatography-tandem mass spectrometry. In addition, 4-HHE treatment suppressed Mcp-1 expression in arterial strips, suggesting 4-HHE derived from DHA may be involved in the mechanism of this phenomenon. In contrast, Mcp-1 expression was stimulated by DHA, EPA and 4-HHE through p38 kinase and the Keap1-Nuclear factor erythroid-derived 2-like 2 (Nrf2) pathway in VSMCs. In conclusion, there is a dual effect of n-3 PUFAs on the regulation of Mcp-1 expression. Further study is necessary to elucidate the pathological role of this phenomenon.
Collapse
MESH Headings
- Aldehydes/metabolism
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chromatography, Liquid
- Docosahexaenoic Acids/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation
- Eicosapentaenoic Acid/pharmacology
- In Vitro Techniques
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-E2-Related Factor 2/metabolism
- RNA Interference
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Tandem Mass Spectrometry
- Time Factors
- Transfection
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Kohji Nagayama
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Katsutaro Morino
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Osamu Sekine
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Fumiyuki Nakagawa
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Osaka Laboratory, JCL Bioassay Corporation, 5-16-26, Minamisuita, Suita-shi, Osaka 564-0043, Japan.
| | - Atsushi Ishikado
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Sunstar Inc., 3-1 Asahi-machi, Takatsuki, Osaka 569-1195, Japan.
- Joslin Diabetes Centre, Harvard Medical School, MA 02115, USA.
| | - Hirotaka Iwasaki
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Takashi Okada
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Masashi Tawa
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Daisuke Sato
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Takeshi Imamura
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Yoshihiko Nishio
- Department of Diabetes and Endocrine Medicine, Kagoshima University, Kagoshima 890-8580, Japan.
| | - Satoshi Ugi
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Atsunori Kashiwagi
- Kusatsu General Hospital, 1660, Yabase-cho, Kusatsu, Shiga 525-8585, Japan.
| | - Tomio Okamura
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| |
Collapse
|
9
|
Kiss K, Baghy K, Spisák S, Szanyi S, Tulassay Z, Zalatnai A, Löhr JM, Jesenofsky R, Kovalszky I, Firneisz G. Chronic hyperglycemia induces trans-differentiation of human pancreatic stellate cells and enhances the malignant molecular communication with human pancreatic cancer cells. PLoS One 2015; 10:e0128059. [PMID: 26010611 PMCID: PMC4444240 DOI: 10.1371/journal.pone.0128059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/23/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetes mellitus is linked to pancreatic cancer. We hypothesized a role for pancreatic stellate cells (PSC) in the hyperglycemia induced deterioration of pancreatic cancer and therefore studied two human cell lines (RLT-PSC, T3M4) in hyperglycemic environment. METHODOLOGY/PRINCIPAL FINDINGS The effect of chronic hyperglycemia (CHG) on PSCs was studied using mRNA expression array with real-time PCR validation and bioinformatic pathway analysis, and confirmatory protein studies. The stress fiber formation (IC: αSMA) indicated that PSCs tend to transdifferentiate to a myofibroblast-like state after exposure to CHG. The phosphorylation of p38 and ERK1/2 was increased with a consecutive upregulation of CDC25, SP1, cFOS and p21, and with downregulation of PPARγ after PSCs were exposed to chronic hyperglycemia. CXCL12 levels increased significantly in PSC supernatant after CHG exposure independently from TGF-β1 treatment (3.09-fold with a 2.73-fold without TGF-β1, p<0.05). The upregualtion of the SP1 transcription factor in PSCs after CHG exposure may be implicated in the increased CXCL12 and IGFBP2 production. In cancer cells, hyperglycemia induced an increased expression of CXCR4, a CXCL12 receptor that was also induced by PSC's conditioned medium. The receptor-ligand interaction increased the phosphorylation of ERK1/2 and p38 resulting in activation of MAP kinase pathway, one of the most powerful stimuli for cell proliferation. Certainly, conditioned medium of PSC increased pancreatic cancer cell proliferation and this effect could be partially inhibited by a CXCR4 inhibitor. As the PSC conditioned medium (normal glucose concentration) increased the ERK1/2 and p38 phosphorylation, we concluded that PSCs produce other factor(s) that influence(s) pancreatic cancer behaviour. CONCLUSIONS Hyperglycemia induces increased CXCL12 production by the PSCs, and its receptor, CXCR4 on cancer cells. The ligand-receptor interaction activates MAP kinase signaling that causes increased cancer cell proliferation and migration.
Collapse
Affiliation(s)
- Katalin Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Spisák
- Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Szilárd Szanyi
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
- School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Zalatnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - J.-Matthias Löhr
- Karolinska Institutet, Gastrocentrum, Karolinska University Hospital, Stockholm, Sweden
| | - Ralf Jesenofsky
- University of Heidelberg, Medical Campus Mannheim, Dept. of Medicine II, Mannheim, Germany
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Firneisz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|