1
|
Holendová B, Stokičová L, Plecitá-Hlavatá L. Lipid Dynamics in Pancreatic β-Cells: Linking Physiology to Diabetes Onset. Antioxid Redox Signal 2024. [PMID: 39495600 DOI: 10.1089/ars.2024.0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Significance: Glucose-induced lipid metabolism is essential for preserving functional β-cells, and its disruption is linked to type 2 diabetes (T2D) development. Lipids are an integral part of the cells playing an indispensable role as structural components, energy storage molecules, and signals. Recent Advances: Glucose presence significantly impacts lipid metabolism in β-cells, where fatty acids are primarily synthesized de novo and/or are transported from the bloodstream. This process is regulated by the glycerolipid/free fatty acid cycle, which includes lipogenic and lipolytic reactions producing metabolic coupling factors crucial for insulin secretion. Disrupted lipid metabolism involving oxidative stress and inflammation is a hallmark of T2D. Critical Issues: Lipid metabolism in β-cells is complex involving multiple simultaneous processes. Exact compartmentalization and quantification of lipid metabolism and its intermediates, especially in response to glucose or chronic hyperglycemia, are essential. Current research often uses non-physiological conditions, which may not accurately reflect in vivo situations. Future Directions: Identifying and quantifying individual steps and their signaling, including redox, within the complex fatty acid and lipid metabolic pathways as well as the metabolites formed during acute versus chronic glucose stimulation, will uncover the detailed mechanisms of glucose-stimulated insulin secretion. This knowledge is crucial for understanding T2D pathogenesis and identifying pharmacological targets to prevent this disease. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Blanka Holendová
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Linda Stokičová
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
2
|
Li Q, Ma Q, Zhou Y, Jiang X, Parales RE, Zhao S, Zhuang Y, Ruan Z. Isolation, identification, and degradation mechanism by multi-omics of mesotrione-degrading Amycolatopsis nivea La24. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134951. [PMID: 38917628 DOI: 10.1016/j.jhazmat.2024.134951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024]
Abstract
Mesotrione is a herbicide used in agricultural production; however, its stability and long-term residues pose ecological risks to soil health and subsequent crops. In this research, the strain Amycolatopsis nivea La24 was identified as capable of completely degrading 50 mg∙L-1 mesotrione within 48 h. It exhibited a broad adaptability to various environment and could degrade three sulfonylurea herbicides (nicosulfuron, chlorimuron-methyl, and cinosulfuron). Non-target metabonomic and mass spectrometry demonstrated that La24 strain broke down the mesotrione parent molecule by targeting the β-diketone bond and nitro group, resulting in the production of five possible degradation products. The differentially expressed genes were significantly enriched in fatty acid degradation, amino acid metabolism, and other pathways, and the differentially metabolites in glutathione metabolism, arginine/proline metabolism, cysteine/methionine metabolism, and other pathways. Additionally, it was confirmed by heterologous expression that nitroreductase was directly involved in the mesotrione degradation, and NDMA-dependent methanol dehydrogenase would increase the resistance to mesotrione. Finally, the intracellular response of La24 during mesotrione degradation was proposed. This work provides insight for a comprehensive understanding of the mesotrione biodegradation mechanism, significantly expands the resources for pollutant degradation, and offers the potential for a more sustainable solution to address herbicide pollution in soil.
Collapse
Affiliation(s)
- Qingqing Li
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China; National Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingyun Ma
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China; National Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiqing Zhou
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xu Jiang
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Rebecca E Parales
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, CA 95616, USA
| | - Shumiao Zhao
- National Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Zhuang
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhiyong Ruan
- State Key Laboratory of Efficient Utilization of Arid and Semi-Arid Arable Land in Northern China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China; CAAS-CIAT Joint Laboratory in Advanced Technologies for Sustainable Agriculture, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
3
|
Zeng C, Sun Y, Lin H, Li Z, Zhang Q, Cai T, Xiang W, Tang J, Yasurin P. D-Limonene Inhibits Pichia kluyveri Y-11519 in Sichuan Pickles by Disrupting Metabolism. Molecules 2024; 29:3561. [PMID: 39124965 PMCID: PMC11314558 DOI: 10.3390/molecules29153561] [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: 06/04/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The Pichia kluyveri, a proliferation commonly found in Sichuan pickles (SCPs), can accelerate the growth and reproduction of spoilage bacteria, causing off-odor development and decay. Although D-limonene, a common natural preservative, effectively restricts P. kluyveri, its inhibitory mechanism remains unclear. This study aimed to elucidate this molecular mechanism by investigating the impact on basic P. kluyveri metabolism. The findings revealed that D-limonene inhibited P. kluyveri growth and disrupted the transcription of the genes responsible for encoding the enzymes involved in cell wall and membrane synthesis, oxidative phosphorylation, glycolysis, and the tricarboxylic acid (TCA) cycle pathway. The results indicated that these events disrupted crucial metabolism such as cell wall and membrane integrity, adenosine triphosphate (ATP) synthesis, and reactive oxygen species (ROS) balance. These insights provided a comprehensive understanding of the inhibitory effect of D-limonene on the growth and reproduction of P. kluyveri while highlighting its potential application in the SCP industry.
Collapse
Affiliation(s)
- Chaoyi Zeng
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Department of Food Biotechnology, Faculty of Biotechnology, Assumption University, Bangkok 10240, Thailand;
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Yue Sun
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Haoran Lin
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Ziyu Li
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Qing Zhang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Ting Cai
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Wenliang Xiang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Jie Tang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China; (Y.S.); (H.L.); (Z.L.); (T.C.); (W.X.); (J.T.)
- Department of Food Biotechnology, Faculty of Biotechnology, Assumption University, Bangkok 10240, Thailand;
- Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Patchanee Yasurin
- Department of Food Biotechnology, Faculty of Biotechnology, Assumption University, Bangkok 10240, Thailand;
| |
Collapse
|
4
|
Tan J, Zhong M, Hu Y, Pan G, Yao J, Tang Y, Duan H, Jiang Y, Shan W, Lin J, Liu Y, Huang J, Zheng H, Zhou Y, Fu G, Li Z, Xu B, Zha J. Ritanserin suppresses acute myeloid leukemia by inhibiting DGKα to downregulate phospholipase D and the Jak-Stat/MAPK pathway. Discov Oncol 2023; 14:118. [PMID: 37392305 DOI: 10.1007/s12672-023-00737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023] Open
Abstract
Refractory or relapsed (R/R) AML is the most challenging form of AML to treat. Due to frequent genetic mutations, therapy alternatives are limited. Here, we identified the role of ritanserin and its target DGKα in AML. Several AML cell lines and primary patient cells were treated with ritanserin and subjected to cell proliferation, apoptosis and gene analyses with CCK-8 assay, Annexin V/PI assay and Western blotting, respectively. We also evaluated the function of the ritanserin target diacylglycerol kinase alpha (DGKα) in AML by bioinformatics. In vitro experiments have revealed that ritanserin inhibits AML progression in a dose- and time-dependent manner, and it shows an anti-AML effect in xenograft mouse models. We further demonstrated that the expression of DGKα was elevated in AML and correlated with poor survival. Mechanistically, ritanserin negatively regulates SphK1 expression through PLD signaling, also inhibiting the Jak-Stat and MAPK signaling pathways via DGKα. These findings suggest that DGKα may be an available therapeutic target and provide effective preclinical evidence of ritanserin as a promising treatment for AML.
Collapse
Affiliation(s)
- Jinshui Tan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Guangchao Pan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuanfang Tang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Hongpeng Duan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Weihang Shan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiaqi Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Yating Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiewen Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361002, Fujian, China
| | - Huijian Zheng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yong Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| |
Collapse
|
5
|
Kaneko YK, Sawatani T, Ishikawa T. Involvement of Diacylglycerol Kinase on the Regulation of Insulin Secretion in Pancreatic β-Cells during Type 2 Diabetes. YAKUGAKU ZASSHI 2022; 142:457-463. [PMID: 35491149 DOI: 10.1248/yakushi.21-00176-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Depression of lipid metabolism in β-cells has been indicated to be one of the causes of impaired insulin secretion in type 2 diabetes. Diacylglycerol (DAG) is an important lipid mediator and is known to regulate insulin secretion in pancreatic β-cells. Intracellular DAG accumulation is involved in β-cell dysfunction in the pathogenesis of type 2 diabetes; thus, the regulation of intracellular DAG levels is likely important for maintaining the β-cell function. We focused on diacylglycerol kinases (DGKs), which strictly regulate intracellular DAG levels, and analyzed the function of type I DGKs (DGKα, γ), which are activated by intracellular Ca2+ and expressed in the cytoplasm, in β-cells. The suppression of the DGKα and γ expression decreased the insulin secretory response, and the decreased expression of DGKα and γ was observed in islets of diabetic model mice. In the pancreatic β-cell line MIN6, 1 μM R59949 (a type I DGK inhibitor) and 10 μM DiC8 (a cell permeable DAG analog) enhanced glucose-induced [Ca2+]i oscillation in a PKC-dependent manner, while 10 μM R59949 and 100 μM DiC8 suppressed [Ca2+]i oscillation and voltage-dependent Ca2+ channel activity in a PKC-independent manner. These results suggest that the intracellular accumulation of DAG by the loss of the DGKα and γ functions regulates insulin secretion in a dual manner depending on the degree of DAG accumulation. The regulation of the insulin secretory response through DAG metabolism by type I DGKs may change depending on the degree of progression of type 2 diabetes.
Collapse
Affiliation(s)
- Yukiko K. Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Toshiaki Sawatani
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
6
|
Lukovic E, Emala C. Diacylglycerol Kinase Puts the Brakes on Airway Smooth Muscle Contraction. Am J Respir Cell Mol Biol 2021; 65:578-580. [PMID: 34375574 PMCID: PMC8641800 DOI: 10.1165/rcmb.2021-0325ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Elvedin Lukovic
- Columbia University, 5798, Anesthesiology, New York, New York, United States
| | - Charles Emala
- Columbia University, 5798, Anesthesiology, New York, New York, United States;
| |
Collapse
|
7
|
Cui H, Huang Y, Wu Y, Ma J, Cao X, Xie J, Zhang Y. The expression of diacylglycerol kinase isoforms α and ζ correlates with the progression of experimental autoimmune encephalomyelitis in rats. Histochem Cell Biol 2021; 156:437-448. [PMID: 34312706 DOI: 10.1007/s00418-021-02011-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is characterized by neuroinflammation and neurodegeneration, whose precise processes are not fully understood. Diacylglycerol kinase (DGK) isozymes of α, β, γ and ζ expressed abundantly in the brain and/or the immune system, may be regulatory targets for MS. In this study, we analyzed the four DGK isozymes along the induction, peak and recovery phases in an experimental autoimmune encephalomyelitis (EAE) rat model of MS. The expression of these DGK isozymes and the diacylglycerol (DAG) pathway in the EAE rat brainstems were analyzed by qRT-PCR, immunohistochemistry, immunofluorescence double staining, western blotting and ELISA. Our results showed that the mRNA content of the four DGK isozymes decreased significantly, and their immunoreactivity in myelin sheathes (DGKα, β) and neurons (DGKγ, ζ) became weaker at the beginning of the induction phase. With the progressive increase in clinical signs, DGKα, DGKγ and DGKζ mRNA increased and DGKβ mRNA decreased, and microglia were involved in the formation of perivascular cuffing. In the peak phase, both DGKα and DGKζ were expressed in neurons and inflammatory cells, and DGKζ was also positive in microglia. During the recovery phase, the mRNA content and immunoreactivity of these DGK isozymes generally reached normal levels. Moreover, our results revealed that changes in DAG accumulation and PKCδ phosphorylation were almost the same as those of DGKα and DGKζ mRNA. In summary, the four DGK isozymes are involved in the EAE process. The predominant and broad presence of DGKα and DGKζ suggests that they may regulate the pathological process by attenuating DAG/PKCδ pathway signaling during EAE evolution.
Collapse
Affiliation(s)
- Huilin Cui
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yige Huang
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ying Wu
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jinfeng Ma
- Department of General Surgery, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Ximei Cao
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jianshan Xie
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yu Zhang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, No.56 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
8
|
Sato T, Ishiwatari C, Kaneko YK, Ishikawa Y, Kimura Y, Watanabe N, Aoshima I, Matsuda Y, Nakayama T, Chiba R, Fujinuki T, Iwata K, Lu Q, Usuki T, Sakane F, Ishikawa T. Diacylglycerol kinase δ functions as a proliferation suppressor in pancreatic β-cells. FASEB J 2021; 35:e21420. [PMID: 33774855 DOI: 10.1096/fj.202001279rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/11/2022]
Abstract
Although an aberrant reduction in pancreatic β-cell mass contributes to the pathogenesis of diabetes, the mechanism underlying the regulation of β-cell mass is poorly understood. Here, we show that diacylglycerol kinase δ (DGKδ) is a key enzyme in the regulation of β-cell mass. DGKδ expression was detected in the nucleus of β-cells. We developed β-cell-specific DGKδ knockout (βDGKδ KO) mice, which showed lower blood glucose, higher plasma insulin levels, and better glucose tolerance compared to control mice. Moreover, an increased number of small islets and Ki-67-positive islet cells, as well as elevated cyclin B1 expression in the islets, were detected in the pancreas of βDGKδ KO mice. DGKδ knockdown in the β-cell line MIN6 induced significant increases in bromodeoxyuridine (BrdU) incorporation and cyclin B1 expression. Finally, we confirmed that streptozotocin-induced hyperglycemia and β-cell loss were alleviated in βDGKδ KO mice. Thus, suppressing the expression or enzymatic activity of DGKδ that functions as a suppressor of β-cell proliferation could be a novel therapeutic approach to increase β-cell mass for the treatment of diabetes.
Collapse
Affiliation(s)
- Taiji Sato
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Chihiro Ishiwatari
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoko Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yuki Kimura
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naoya Watanabe
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Ikumi Aoshima
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukari Matsuda
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takahiro Nakayama
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Rina Chiba
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takahiro Fujinuki
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kai Iwata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Qiang Lu
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Takako Usuki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
9
|
Hernandez-Baixauli J, Puigbò P, Torrell H, Palacios-Jordan H, Ripoll VJR, Caimari A, Del Bas JM, Baselga-Escudero L, Mulero M. A Pilot Study for Metabolic Profiling of Obesity-Associated Microbial Gut Dysbiosis in Male Wistar Rats. Biomolecules 2021; 11:303. [PMID: 33670496 PMCID: PMC7922951 DOI: 10.3390/biom11020303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/06/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is one of the most incident and concerning disease worldwide. Definite strategies to prevent obesity and related complications remain elusive. Among the risk factors of the onset of obesity, gut microbiota might play an important role in the pathogenesis of the disease, and it has received extensive attention because it affects the host metabolism. In this study, we aimed to define a metabolic profile of the segregated obesity-associated gut dysbiosis risk factor. The study of the metabolome, in an obesity-associated gut dysbiosis model, provides a relevant way for the discrimination on the different biomarkers in the obesity onset. Thus, we developed a model of this obesity risk factors through the transference of gut microbiota from obese to non-obese male Wistar rats and performed a subsequent metabolic analysis in the receptor rats. Our results showed alterations in the lipid metabolism in plasma and in the phenylalanine metabolism in urine. In consequence, we have identified metabolic changes characterized by: (1) an increase in DG:34:2 in plasma, a decrease in hippurate, (2) an increase in 3-HPPA, and (3) an increase in o-coumaric acid. Hereby, we propose these metabolites as a metabolic profile associated to a segregated dysbiosis state related to obesity disease.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (P.P.); (A.C.); (L.B.-E.)
| | - Pere Puigbò
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (P.P.); (A.C.); (L.B.-E.)
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Department of Biology, University of Turku, 20014 Turku, Finland
| | - Helena Torrell
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili−EURECAT, 43204 Reus, Spain; (H.T.); (H.P.-J.)
| | - Hector Palacios-Jordan
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili−EURECAT, 43204 Reus, Spain; (H.T.); (H.P.-J.)
| | | | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (P.P.); (A.C.); (L.B.-E.)
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (P.P.); (A.C.); (L.B.-E.)
| | - Laura Baselga-Escudero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (P.P.); (A.C.); (L.B.-E.)
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| |
Collapse
|
10
|
Pandurangan S, Murugesan P, Ramudu KN, Krishnaswamy B, Ayyadurai N. Enhanced Cellular Uptake and Sustained Transdermal Delivery of Collagen for Skin Regeneration. ACS APPLIED BIO MATERIALS 2020; 3:7540-7549. [PMID: 35019495 DOI: 10.1021/acsabm.0c00755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The present study reports a method for transporting high molecular weight collagen for skin regeneration. An independent engineered enzymatic vehicle that has the ability for efficient transdermal delivery of regenerative biomaterial was developed for tissue regeneration. Collagen has been well recognized as a skin regeneration molecule due to its interaction with the extracellular matrix to stimulate skin cell growth, proliferation, and differentiation. However, the transdermal delivery of collagen poses a significant challenge due to its high molecular weight as well as a lack of efficient approaches. Here, to improve the transdermal delivery efficiency, α-1,4-glycosidic hydrolase was engineered with genetically encoded 3,4-dihydroxy-L-phenylalanine, which enhanced its biological activity as revealed by microscale thermophoresis. The remodeled catalytic pocket resulted in enhanced substrate binding activity of the enzyme with a predominant glycosaminoglycan (chondroitin sulfate) present in the extracellular matrix of the skin. The engineered enzyme rapidly opened up the skin extracellular matrix fiber (15 min) to ferry collagen across the wall, without disturbing the cellular bundle architecture. Confocal microscopy indicated that macromolecules had diffused three times deeper into the engineered enzyme-treated skin than the native enzyme-treated skin. Gene expression, histopathology, and hematology analysis also supported the penetration of macromolecules. Cytotoxicity (mammalian cell culture) and in vivo (Caenorhabditis elegans and Rattus noryegicus) studies revealed that the congener enzyme could potentially be used as a penetration enhancer, which is of paramount importance for the multimillion cosmetic industries. Hence, it offers promise as a pharmaceutical enzyme for transdermal delivery bioenhancement and dermatological applications.
Collapse
Affiliation(s)
- Suryalakshmi Pandurangan
- Division of Biochemistry and Biotechnology Council of Scientific and Industrial Research, Central Leather Research Institute, Chennai 600 020, India.,Academy of Scientific and Innovative Research Central Leather Research Institute Campus, Chennai 600 020, India
| | | | - Kamini Numbi Ramudu
- Division of Biochemistry and Biotechnology Council of Scientific and Industrial Research, Central Leather Research Institute, Chennai 600 020, India.,Academy of Scientific and Innovative Research Central Leather Research Institute Campus, Chennai 600 020, India
| | | | - Niraikulam Ayyadurai
- Division of Biochemistry and Biotechnology Council of Scientific and Industrial Research, Central Leather Research Institute, Chennai 600 020, India.,Academy of Scientific and Innovative Research Central Leather Research Institute Campus, Chennai 600 020, India
| |
Collapse
|
11
|
Olivença DV, Voit EO, Pinto FR. ENaC regulation by phospholipids and DGK explained through mathematical modeling. Sci Rep 2020; 10:13952. [PMID: 32811866 PMCID: PMC7435262 DOI: 10.1038/s41598-020-70630-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Cystic fibrosis is a condition caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). It is also thought to increase the activity of epithelial sodium channels (ENaC). The altered function of these ion channels is one of the causes of the thick dehydrated mucus that characterizes the disease and is partially responsible for recurrent pulmonary infections and inflammation events that ultimately destroy the lungs of affected subjects. Phosphoinositides are signaling lipids that regulate numerous cellular processes and membrane proteins, including ENaC. Inhibition of diacylglycerol kinase (DGK), an enzyme of the phosphoinositide pathway, reduces ENaC function. We propose a computational analysis that is based on the combination of two existing mathematical models: one representing the dynamics of phosphoinositides and the other explaining how phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) influences ENaC activity and, consequently, airway surface liquid. This integrated model permits, for the first time, a detailed assessment of the intricate interactions between DGK and ENaC and is consistent with available literature data. In particular, the computational approach allows comparisons of two competing hypotheses regarding the regulation of ENaC. The results strongly suggest that the regulation of ENaC is primarily exerted through the control of PI(4,5)P2 production by type-I phosphatidylinositol-4-phosphate 5-kinase (PIP5KI), which in turn is controlled by phosphatidic acid (PA), the product of the DGK reaction.
Collapse
Affiliation(s)
- Daniel V. Olivença
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Eberhard O. Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Francisco R. Pinto
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
| |
Collapse
|
12
|
Poli A, Fiume R, Mongiorgi S, Zaurito A, Sheth B, Vidalle MC, Hamid SA, Kimber S, Campagnoli F, Ratti S, Rusciano I, Faenza I, Manzoli L, Divecha N. Exploring the controversial role of PI3K signalling in CD4 + regulatory T (T-Reg) cells. Adv Biol Regul 2020; 76:100722. [PMID: 32362560 DOI: 10.1016/j.jbior.2020.100722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/10/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
The immune system is a complex network that acts to protect vertebrates from foreign microorganisms and carries out immunosurveillance to combat cancer. In order to avoid hyper-activation of the immune system leading to collateral damage tissues and organs and to prevent self-attack, the network has the intrinsic control mechanisms that negatively regulate immune responses. Central to this negative regulation are regulatory T (T-Reg) cells, which through cytokine secretion and cell interaction limit uncontrolled clonal expansion and functions of activated immune cells. Given that positive or negative manipulation of T-Regs activity could be utilised to therapeutically treat host versus graft rejection or cancer respectively, understanding how signaling pathways impact on T-Regs function should reveal potential targets with which to intervene. The phosphatidylinositol-3-kinase (PI3K) pathway controls a vast array of cellular processes and is critical in T cell activation. Here we focus on phosphoinositide 3-kinases (PI3Ks) and their ability to regulate T-Regs cell differentiation and function.
Collapse
Affiliation(s)
- Alessandro Poli
- The FIRC Institute of Molecular Oncology (IFOM), 20139, Milan, Italy
| | - Roberta Fiume
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy.
| | - Sara Mongiorgi
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Antonio Zaurito
- Center for Translational Cancer Research (TranslaTUM), Klinikum Rechts der Isar, Technische Universität München, 81675, Munich, Germany
| | - Bhavwanti Sheth
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| | - Magdalena Castellano Vidalle
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| | - Shidqiyyah Abdul Hamid
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| | - ScottT Kimber
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| | - Francesca Campagnoli
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| | - Stefano Ratti
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Isabella Rusciano
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Irene Faenza
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical Sciences (DIBINEM), University of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Nullin Divecha
- Inositide Laboratory, School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building 85, Highfield, Southampton, SO17 1BJ, UK
| |
Collapse
|
13
|
Gu X, Wan G, Chen N, Li J, Chen B, Tang Y, Gu W, Jin C, Meng J, Zhang P, Liu L, Yang Z, Lu C. DGKζ Plays Crucial Roles in the Proliferation and Tumorigenicity of Human glioblastoma. Int J Biol Sci 2019; 15:1872-1881. [PMID: 31523189 PMCID: PMC6743304 DOI: 10.7150/ijbs.35193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/23/2019] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma is one of the most malignant brain cancers in adults, and it is a fatal disease because of its untimely pathogenetic location detection, infiltrative growth, and unfavorable prognosis. Unfortunately, multimodal treatment with maximal safe resection, chemotherapy and radiation has not increased the survival rate of patients with glioblastoma. Gene- and molecular-targeted therapy is considered to be a promising anticancer strategy for glioblastoma. The identification of novel potential targets in glioblastoma is of high importance. In this study, we found that both the mRNA and protein levels of diacylglycerol kinase ζ (DGKζ) were significantly higher in glioblastoma tissues than in precancerous lesions. The silencing of DGKζ by lentivirus-delivered shRNA reduced glioblastoma cell proliferation and induced G0/G1 phase arrest. Moreover, knockdown of DGKζ expression in U251 cells markedly reduced in vitro colony formation and in vivo tumorigenic capability. Further study showed that DGKζ inhibition resulted in decreases in cyclin D1, p-AKT and p-mTOR. Moreover, the rescue or overexpression of DGKζ in glioblastoma cells demonstrated the oncogenic function of DGKζ. In conclusion, these studies suggest that the suppression of DGKζ may inhibit the tumor growth of glioblastoma cells with high DGKζ expression. Thus, DGKζ might be a potential therapeutic target in malignant glioblastoma.
Collapse
Affiliation(s)
- Xuefeng Gu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Guoqing Wan
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Nianhong Chen
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Jinhong Li
- Department of Neurosurgery, Jiangmen Central Hospital, Jiangmen, Guangdong, PR China
| | - Bing Chen
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Yeling Tang
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Wangxian Gu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Cuiting Jin
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Jihong Meng
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Peng Zhang
- College of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Li Liu
- College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Zhifang Yang
- College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Changlian Lu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| |
Collapse
|
14
|
Sawatani T, Kaneko YK, Ishikawa T. Dual effect of reduced type I diacylglycerol kinase activity on insulin secretion from MIN6 β-cells. J Pharmacol Sci 2019; 140:178-186. [PMID: 31279581 DOI: 10.1016/j.jphs.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/03/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023] Open
Abstract
The role of type I diacylglycerol kinases (DGKs) in the regulation of insulin secretion was investigated in MIN6 β-cells. In intracellular Ca2+ concentration ([Ca2+]i) measurement experiments, 1 μM R59949, a type I DGK inhibitor, and 10 μM DiC8, a diacylglycerol (DAG) analog, amplified 22.2 mM glucose-induced [Ca2+]i oscillations in a protein kinase C (PKC)-dependent manner, whereas 10 μM R59949 and 100 μM DiC8 decreased [Ca2+]i independent of PKC. High concentrations of R59949 and DiC8 attenuated voltage-dependent Ca2+ channel currents. According to these results, 22.2 mM glucose-stimulated insulin secretion (GSIS) was potentiated by 1 μM R59949 but suppressed by 10 μM of the same. The DGKα inhibitor R59022 showed a similar dual effect. Conversely, DiC8 at 10 and 100 μM potentiated GSIS, although 100 μM DiC8 decreased [Ca2+]i. These results suggest that DAG accumulated through declined type I DGK activity shows a dual effect on insulin secretion depending on the degree of accumulation; a mild DAG accumulation induces a PKC-dependent stimulatory effect on insulin secretion, whereas an excessive DAG accumulation suppresses it in a PKC-independent manner, possibly via attenuation of VDCC activity.
Collapse
Affiliation(s)
- Toshiaki Sawatani
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka, 422-8526, Japan
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka, 422-8526, Japan.
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka, 422-8526, Japan
| |
Collapse
|
15
|
Zhang XF, Ajeti V, Tsai N, Fereydooni A, Burns W, Murrell M, De La Cruz EM, Forscher P. Regulation of axon growth by myosin II-dependent mechanocatalysis of cofilin activity. J Cell Biol 2019; 218:2329-2349. [PMID: 31123185 PMCID: PMC6605792 DOI: 10.1083/jcb.201810054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/26/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Synergism between myosin II contractility and cofilin activity modulates serotonin-dependent axon growth. Normally, cofilin-dependent decreases in actin density are compensated by increases in point contact density and traction force; however, myosin hyperactivation leads to catastrophic decreases in actin network density and neurite retraction. Serotonin (5-HT) is known to increase the rate of growth cone advance via cofilin-dependent increases in retrograde actin network flow and nonmuscle myosin II activity. We report that myosin II activity is regulated by PKC during 5-HT responses and that PKC activity is necessary for increases in traction force normally associated with these growth responses. 5-HT simultaneously induces cofilin-dependent decreases in actin network density and PKC-dependent increases in point contact density. These reciprocal effects facilitate increases in traction force production in domains exhibiting decreased actin network density. Interestingly, when PKC activity was up-regulated, 5-HT treatments resulted in myosin II hyperactivation accompanied by catastrophic cofilin-dependent decreases in actin filament density, sudden decreases in traction force, and neurite retraction. These results reveal a synergistic relationship between cofilin and myosin II that is spatiotemporally regulated in the growth cone via mechanocatalytic effects to modulate neurite growth.
Collapse
Affiliation(s)
- Xiao-Feng Zhang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT
| | - Visar Ajeti
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT.,Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT
| | - Nicole Tsai
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT.,Department of Ophthalmology, University of California, San Francisco, California, CA
| | - Arash Fereydooni
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT
| | - William Burns
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT
| | - Michael Murrell
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Enrique M De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
| | - Paul Forscher
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT
| |
Collapse
|
16
|
Ma Q, Gabelli SB, Raben DM. Diacylglycerol kinases: Relationship to other lipid kinases. Adv Biol Regul 2019; 71:104-110. [PMID: 30348515 PMCID: PMC6347529 DOI: 10.1016/j.jbior.2018.09.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 04/17/2023]
Abstract
Lipid kinases regulate a wide variety of cellular functions and have emerged as one the most promising targets for drug design. Diacylglycerol kinases (DGKs) are a family of enzymes that catalyze the ATP-dependent phosphorylation of diacylglycerol (DAG) to phosphatidic acid (PtdOH). Despite the critical role in lipid biosynthesis, both DAG and PtdOH have been shown as bioactive lipids mediating a number of signaling pathways. Although there is increasing recognition of their role in signaling systems, our understanding of the key enzyme which regulate the balance of these two lipid messages remain limited. Solved structures provide a wealth of information for understanding the function and regulation of these enzymes. Solving the structures of mammalian DGKs by traditional NMR and X-ray crystallography approaches have been challenging and so far, there are still no three-dimensional structures of these DGKs. Despite this, some insights may be gained by examining the similarities and differences between prokaryotic DGKs and other mammalian lipid kinases. This review focuses on summarizing and comparing the structure of prokaryotic and mammalian DGKs as well as two other lipid kinases: sphingosine kinase and phosphatidylinositol-3-kinase. How these known lipid kinases structures relate to mammalian DGKs will also be discussed.
Collapse
Affiliation(s)
- Qianqian Ma
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sandra B Gabelli
- The Department of Biophysics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel M Raben
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
Merida I, Arranz-Nicolás J, Torres-Ayuso P, Ávila-Flores A. Diacylglycerol Kinase Malfunction in Human Disease and the Search for Specific Inhibitors. Handb Exp Pharmacol 2019; 259:133-162. [PMID: 31227890 DOI: 10.1007/164_2019_221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The diacylglycerol kinases (DGKs) are master regulator kinases that control the switch from diacylglycerol (DAG) to phosphatidic acid (PA), two lipids with important structural and signaling properties. Mammalian DGKs distribute into five subfamilies that regulate local availability of DAG and PA pools in a tissue- and subcellular-restricted manner. Pharmacological manipulation of DGK activity holds great promise, given the critical contribution of specific DGK subtypes to the control of membrane structure, signaling complexes, and cell-cell communication. The latest advances in the DGK field have unveiled the differential contribution of selected isoforms to human disease. Defects in the expression/activity of individual DGK isoforms contribute substantially to cognitive impairment, mental disorders, insulin resistance, and vascular pathologies. Abnormal DGK overexpression, on the other hand, confers the acquisition of malignant traits including invasion, chemotherapy resistance, and inhibition of immune attack on tumors. Translation of these findings into therapeutic approaches will require development of methods to pharmacologically modulate DGK functions. In particular, inhibitors that target the DGKα isoform hold particular promise in the fight against cancer, on their own or in combination with immune-targeting therapies.
Collapse
Affiliation(s)
- Isabel Merida
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Javier Arranz-Nicolás
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain
| | - Pedro Torres-Ayuso
- Laboratory of Cell and Developmental Signaling, National Cancer Institute (NCI-NIH), Frederick, MD, USA
| | - Antonia Ávila-Flores
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain
| |
Collapse
|
18
|
Ratti S, Ramazzotti G, Faenza I, Fiume R, Mongiorgi S, Billi AM, McCubrey JA, Suh PG, Manzoli L, Cocco L, Follo MY. Nuclear inositide signaling and cell cycle. Adv Biol Regul 2018; 67:1-6. [PMID: 29102395 DOI: 10.1016/j.jbior.2017.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 06/07/2023]
Abstract
Phosphatidylinositols (PIs) are responsible for several signaling pathways related to many cellular functions, such as cell cycle regulation at different check-points, cell proliferation, cell differentiation, membrane trafficking and gene expression. PI metabolism is not only present at the cytoplasmic level, but also at the nuclear one, where different signaling pathways affect essential nuclear mechanisms in eukaryotic cells. In this review we focus on nuclear inositide signaling in relation to cell cycle regulation. Many evidences underline the pivotal role of nuclear inositide signaling in cell cycle regulation and cell proliferation associated to different strategic physiopathological mechanisms in several cell systems and diseases.
Collapse
Affiliation(s)
- Stefano Ratti
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Irene Faenza
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Roberta Fiume
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Sara Mongiorgi
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Anna Maria Billi
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, MS#629, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Lucia Manzoli
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Lucio Cocco
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Matilde Y Follo
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
19
|
Sakane F, Mizuno S, Takahashi D, Sakai H. Where do substrates of diacylglycerol kinases come from? Diacylglycerol kinases utilize diacylglycerol species supplied from phosphatidylinositol turnover-independent pathways. Adv Biol Regul 2018; 67:101-108. [PMID: 28918129 DOI: 10.1016/j.jbior.2017.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 06/07/2023]
Abstract
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol (DG) to produce phosphatidic acid (PA). Mammalian DGK comprises ten isozymes (α-κ) and regulates a wide variety of physiological and pathological events, such as cancer, type II diabetes, neuronal disorders and immune responses. DG and PA consist of various molecular species that have different acyl chains at the sn-1 and sn-2 positions, and consequently, mammalian cells contain at least 50 structurally distinct DG/PA species. Because DGK is one of the components of phosphatidylinositol (PI) turnover, the generally accepted dogma is that all DGK isozymes utilize 18:0/20:4-DG derived from PI turnover. We recently established a specific liquid chromatography-mass spectrometry method to analyze which PA species were generated by DGK isozymes in a cell stimulation-dependent manner. Interestingly, we determined that DGKδ, which is closely related to the pathogenesis of type II diabetes, preferentially utilized 14:0/16:0-, 14:0/16:1-, 16:0/16:0-, 16:0/16:1-, 16:0/18:0- and 16:0/18:1-DG species (X:Y = the total number of carbon atoms: the total number of double bonds) supplied from the phosphatidylcholine-specific phospholipase C pathway, but not 18:0/20:4-DG, in high glucose-stimulated C2C12 myoblasts. Moreover, DGKα mainly consumed 14:0/16:0-, 16:0/18:1-, 18:0/18:1- and 18:1/18:1-DG species during cell proliferation in AKI melanoma cells. Furthermore, we found that 16:0/16:0-PA was specifically produced by DGKζ in Neuro-2a cells during retinoic acid- and serum starvation-induced neuronal differentiation. These results indicate that DGK isozymes utilize a variety of DG molecular species derived from PI turnover-independent pathways as substrates in different stimuli and cells. DGK isozymes phosphorylate various DG species to generate various PA species. It was revealed that the modes of activation of conventional and novel protein kinase isoforms by DG molecular species varied considerably. However, PA species-selective binding proteins have not been found to date. Therefore, we next attempted to identify PA species-selective binding proteins from the mouse brain and identified α-synuclein, which has causal links to Parkinson's disease. Intriguingly, we determined that among phospholipids, including several PA species (16:0/16:0-PA, 16:0/18:1-PA, 18:1/18:1-PA, 18:0/18:0-PA and 18:0/20:4-PA); 18:1/18:1-PA was the most strongly bound PA to α-synuclein. Moreover, 18:1/18:1-PA strongly enhanced secondary structural changes from the random coil form to the α-helix form and generated a multimeric and proteinase K-resistant α-synuclein protein. In contrast with the dogma described above, our recent studies strongly suggest that PI turnover-derived DG species and also various DG species derived from PI turnover-independent pathways are utilized by DGK isozymes. DG species supplied from distinct pathways may be utilized by DGK isozymes based on different stimuli present in different types of cells, and individual PA molecular species would have specific targets and exert their own physiological functions.
Collapse
Affiliation(s)
- Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan.
| | - Satoru Mizuno
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Hiromichi Sakai
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| |
Collapse
|
20
|
Poli A, Fiume R, Baldanzi G, Capello D, Ratti S, Gesi M, Manzoli L, Graziani A, Suh PG, Cocco L, Follo MY. Nuclear Localization of Diacylglycerol Kinase Alpha in K562 Cells Is Involved in Cell Cycle Progression. J Cell Physiol 2017; 232:2550-2557. [PMID: 27731506 DOI: 10.1002/jcp.25642] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 12/16/2022]
Abstract
Phosphatidylinositol (PI) signaling is an essential regulator of cell motility and proliferation. A portion of PI metabolism and signaling takes place in the nuclear compartment of eukaryotic cells, where an array of kinases and phosphatases localize and modulate PI. Among these, Diacylglycerol Kinases (DGKs) are a class of phosphotransferases that phosphorylate diacylglycerol and induce the synthesis of phosphatidic acid. Nuclear DGKalpha modulates cell cycle progression, and its activity or expression can lead to changes in the phosphorylated status of the Retinoblastoma protein, thus, impairing G1/S transition and, subsequently, inducing cell cycle arrest, which is often uncoupled with apoptosis or autophagy induction. Here we report for the first time not only that the DGKalpha isoform is highly expressed in the nuclei of human erythroleukemia cell line K562, but also that its nuclear activity drives K562 cells through the G1/S transition during cell cycle progression. J. Cell. Physiol. 232: 2550-2557, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessandro Poli
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy.,Istituto Nazionale Genetica Molecolare "Romeo e Enrica Invernizzi", Milano, Italy.,Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine and Institute for Research and Cure of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| | - Daniela Capello
- Department of Translational Medicine and Institute for Research and Cure of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Marco Gesi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Andrea Graziani
- Department of Translational Medicine and Institute for Research and Cure of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy.,University Vita e Salute San Raffaele, Milan, Italy
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| |
Collapse
|
21
|
Kaneko YK, Ishikawa T. [Regulation of Lipid Metabolism by Diacylglycerol Kinases in Pancreatic β-cells]. YAKUGAKU ZASSHI 2017; 136:461-5. [PMID: 26935087 DOI: 10.1248/yakushi.15-00246-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The appropriate secretion of insulin from pancreatic β-cells is essential for regulating blood glucose levels. Glucose-stimulated insulin secretion (GSIS) involves the following steps: Glucose uptake by pancreatic β-cells is metabolized to produce ATP. Increased ATP levels result in the closure of ATP-sensitive K(+) (KATP) channels, resulting in membrane depolarization that activates voltage-dependent Ca(2+) channels to subsequently trigger insulin secretion. In addition to this primary mechanism through KATP channels, insulin secretion is regulated by cyclic AMP and diacylglycerol (DAG), which mediate the effects of receptor agonists such as GLP-1 and acetylcholine. Glucose by itself can also increase the levels of these second messengers. Recently, we have shown an obligatory role of diacylglycerol kinase (DGK), an enzyme catalyzing the conversion of DAG to phosphatidic acid, in GSIS. Of the 10 known DGK isoforms, we focused on type-I DGK isoforms (i.e., DGKα, DGKβ, and DGKγ), which are activated by Ca(2+). The protein expression of DGKα and DGKγ was detected in mouse pancreatic islets and the pancreatic β-cell line MIN6. Depletion of these DGKs by a specific inhibitor or siRNA decreased both [Ca(2+)]i and insulin secretion in MIN6 cells. Similar [Ca(2+)]i responses were induced by DiC8, a membrane-permeable DAG analog. These results suggest that DGKα and DGKγ play crucial roles in insulin secretion, and that their depletion impairs insulin secretion through DAG accumulation. In this article, we review the current understanding of the roles of DAG- and DGK-signaling in pancreatic β-cells, and discuss their pathophysiological roles in the progression of type-2 diabetes.
Collapse
Affiliation(s)
- Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | | |
Collapse
|
22
|
Iwazaki K, Tanaka T, Hozumi Y, Okada M, Tsuchiya R, Iseki K, Topham MK, Kawamae K, Takagi M, Goto K. DGKζ Downregulation Enhances Osteoclast Differentiation and Bone Resorption Activity Under Inflammatory Conditions. J Cell Physiol 2016; 232:617-624. [DOI: 10.1002/jcp.25461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/15/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Kiyoshi Iwazaki
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
- Department of Orthopaedic; Yamagata University School of Medicine; Yamagata Japan
| | - Toshiaki Tanaka
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
| | - Yasukazu Hozumi
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
| | - Masashi Okada
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
| | - Rieko Tsuchiya
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
| | - Ken Iseki
- Department of Emergency and Critical Care Medicine; Yamagata University School of Medicine; Yamagata Japan
| | - Matthew K. Topham
- Department of Oncological Sciences, Huntsman Cancer Institute; University of Utah; Salt Lake City Utah
| | - Kaneyuki Kawamae
- Department of Anesthesiology; Yamagata University School of Medicine; Yamagata Japan
| | - Michiaki Takagi
- Department of Orthopaedic; Yamagata University School of Medicine; Yamagata Japan
| | - Kaoru Goto
- Department of Anatomy and Cell Biology; Yamagata University School of Medicine; Yamagata Japan
| |
Collapse
|
23
|
Huang HY, Zhao GP, Liu RR, Li QH, Zheng MQ, Li SF, Liang Z, Zhao ZH, Wen J. Brain Natriuretic Peptide Stimulates Lipid Metabolism through Its Receptor NPR1 and the Glycerolipid Metabolism Pathway in Chicken Adipocytes. Biochemistry 2015; 54:6622-30. [PMID: 26463554 DOI: 10.1021/acs.biochem.5b00714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain natriuretic peptide (BNP) is related to lipid metabolism in mammals, but its effect and the molecular mechanisms underlying it in chickens are incompletely understood. We found that the level of natriuretic peptide precursor B (NPPB, which encodes BNP) mRNA expression in high-abdominal-fat chicken groups was significantly higher than that of low-abdominal-fat groups. Partial correlations indicated that changes in the weight of abdominal fat were positively correlated with NPPB mRNA expression level. In vitro, compared with the control group, preadipocytes with NPPB interference showed reduced levels of proliferation, differentiation, and glycerin in media. Treatments of cells with BNP led to enhanced proliferation and differentiation of cells and glycerin concentration, and mRNA expression of its receptor natriuretic peptide receptor 1 (NPR1) was upregulated significantly. In cells exposed to BNP, 482 differentially expressed genes were identified compared with controls without BNP. Four genes known to be related to lipid metabolism (diacylglycerol kinase; lipase, endothelial; 1-acylglycerol-3-phosphate O-acyltransferase 1; and 1-acylglycerol-3-phosphate O-acyltransferase 2) were enriched in the glycerolipid metabolism pathway and expressed differentially. In conclusion, BNP stimulates the proliferation, differentiation, and lipolysis of preadipocytes through upregulation of the levels of expression of its receptor NPR1 and key genes enriched in the glycerolipid metabolic pathway.
Collapse
Affiliation(s)
- H Y Huang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,Institute of Poultry Science, Chinese Academy of Agriculture Sciences , Jiangsu 225125, P. R. China
| | - G P Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,State Key Laboratory of Animal Nutrition , Beijing 100193, P. R. China
| | - R R Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,State Key Laboratory of Animal Nutrition , Beijing 100193, P. R. China
| | - Q H Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,State Key Laboratory of Animal Nutrition , Beijing 100193, P. R. China
| | - M Q Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,State Key Laboratory of Animal Nutrition , Beijing 100193, P. R. China
| | - S F Li
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences , Jiangsu 225125, P. R. China
| | - Z Liang
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences , Jiangsu 225125, P. R. China
| | - Z H Zhao
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences , Jiangsu 225125, P. R. China
| | - J Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193, P. R. China.,State Key Laboratory of Animal Nutrition , Beijing 100193, P. R. China
| |
Collapse
|
24
|
Tanaka T, Tsuchiya R, Hozumi Y, Nakano T, Okada M, Goto K. Reciprocal regulation of p53 and NF-κB by diacylglycerol kinase ζ. Adv Biol Regul 2015; 60:15-21. [PMID: 26521214 DOI: 10.1016/j.jbior.2015.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 11/29/2022]
Abstract
Diacylglycerol kinase (DGK) participates in lipid mediated-signal transduction. It phosphorylates diacylglycerol (DG) to phosphatidic acid (PA), thereby regulating the balanced control of these second messenger actions. Previous reports have described that one DGK family, DGKζ, is closely involved in stress responses under various conditions. Cellular stress response, a physiological process enabling cells to cope with an altered environment, is finely tuned through various signaling cascades and their molecular crosstalk. The major components of stress response are p53 and NF-κB. p53 generally serves as a proapoptotic transcriptional factor, whereas NF-κB promotes resistance to programmed cell death under most circumstances. Recent studies have suggested that DGKζ facilitates p53 degradation in cytoplasm through ubiquitin proteasome system and that DGKζ deletion upregulates p53 protein levels under basal and DNA-damage conditions. Counter-intuitively, however, DGKζ deletion suppresses p53 transcriptional activity despite increased p53 levels. In contrast, DGKζ knockdown engenders enhancement of NF-κB pathway in response to cytokines such as TNF-α and IL-1β. In response to these cytokines, DGKζ downregulation accelerates phosphorylation of the p65 subunit and its nuclear translocation, thereby enhancing NF-κB transcriptional activity. Furthermore, DGKζ deficiency is shown to promote increased association of p65 subunit with the transcriptional cofactor CBP. It is particularly interesting that this association is observed even under basal conditions in the absence of stimulation. These findings suggest that DGKζ plays a role in sequestration of the limiting pool of CBP/p300 between the NF-κB p65 subunit and p53, and that DGKζ downregulation shifts CBP/p300 toward the NF-κB subunit to regulate reciprocally antagonistic phenotypes of these transcription factors.
Collapse
Affiliation(s)
- Toshiaki Tanaka
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan.
| | - Rieko Tsuchiya
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Yasukazu Hozumi
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Tomoyuki Nakano
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Masashi Okada
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan.
| |
Collapse
|
25
|
Zamani A, Decker C, Cremasco V, Hughes L, Novack DV, Faccio R. Diacylglycerol Kinase ζ (DGKζ) Is a Critical Regulator of Bone Homeostasis Via Modulation of c-Fos Levels in Osteoclasts. J Bone Miner Res 2015; 30:1852-63. [PMID: 25891971 PMCID: PMC4580562 DOI: 10.1002/jbmr.2533] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Increased diacylglycerol (DAG) levels are observed in numerous pathologies, including conditions associated with bone loss. However, the effects of DAG accumulation on the skeleton have never been directly examined. Because DAG is strictly controlled by tissue-specific diacylglycerol kinases (DGKs), we sought to examine the biological consequences of DAG accumulation on bone homeostasis by genetic deletion of DGKζ, a highly expressed DGK isoform in osteoclasts (OCs). Strikingly, DGKζ(-/-) mice are osteoporotic because of a marked increase in OC numbers. In vitro, DGKζ(-/-) bone marrow macrophages (BMMs) form more numerous, larger, and highly resorptive OCs. Surprisingly, although increased DAG levels do not alter receptor activator of NF-κB (RANK)/RANK ligand (RANKL) osteoclastogenic pathway, DGKζ deficiency increases responsiveness to the proliferative and pro-survival cytokine macrophage colony-stimulating factor (M-CSF). We find that M-CSF is responsible for increased DGKζ(-/-) OC differentiation by promoting higher expression of the transcription factor c-Fos, and c-Fos knockdown in DGKζ(-/-) cultures dose-dependently reduces OC differentiation. Using a c-Fos luciferase reporter assay lacking the TRE responsive element, we also demonstrate that M-CSF induces optimal c-Fos expression through DAG production. Finally, to demonstrate the importance of the M-CSF/DGKζ/DAG axis on regulation of c-Fos during osteoclastogenesis, we turned to PLCγ2(+/-) BMMs, which have reduced DAG levels and form fewer OCs because of impaired expression of the master regulator of osteoclastogenesis NFATc1 and c-Fos. Strikingly, genetic deletion of DGKζ in PLCγ2(+/-) mice rescues OC formation and normalizes c-Fos levels without altering NFATc1 expression. To our knowledge, this is the first report implicating M-CSF/DGKζ/DAG axis as a critical regulator of bone homeostasis via its actions on OC differentiation and c-Fos expression.
Collapse
Affiliation(s)
- Ali Zamani
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Corinne Decker
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Viviana Cremasco
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Lindsey Hughes
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Deborah V. Novack
- Department of Pathology and Immunology; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Roberta Faccio
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
- Corresponding Author Roberta Faccio, Box 8233, 660 S. Euclid, St. Louis, MO 63110, USA, Phone: 314-747-4602, Fax: 314-362-0334,
| |
Collapse
|
26
|
Kaneko YK, Ishikawa T. Diacylglycerol Signaling Pathway in Pancreatic β-Cells: An Essential Role of Diacylglycerol Kinase in the Regulation of Insulin Secretion. Biol Pharm Bull 2015; 38:669-73. [DOI: 10.1248/bpb.b15-00060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yukiko K. Kaneko
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, University of Shizuoka
| | - Tomohisa Ishikawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
27
|
Li J, Ziemba BP, Falke J, Voth GA. Interactions of protein kinase C-α C1A and C1B domains with membranes: a combined computational and experimental study. J Am Chem Soc 2014; 136:11757-66. [PMID: 25075641 PMCID: PMC4140453 DOI: 10.1021/ja505369r] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Indexed: 02/01/2023]
Abstract
Protein kinase C-α (PKCα) has been studied widely as a paradigm for conventional PKCs, with two C1 domains (C1A and C1B) being important for the regulation and function of the kinase. However, it is challenging to explore these domains in membrane-bound environments with either simulations or experiments alone. In this work, we have combined modeling, simulations, and experiments to understand the molecular basis of the PKCα C1A and C1B domain interactions with membranes. Our atomistic simulations of the PKCα C1 domains reveal the dynamic interactions of the proteins with anionic lipids, as well as the conserved hydrogen bonds and the distinct nonpolar contacts formed with lipid activators. Corroborating evidence is obtained from additional simulations and experiments in terms of lipid binding and protein diffusion. Overall, our study, for the first time, explains with atomistic detail how the PKCα C1A and C1B domains interact differently with various lipids. On the molecular level, the information provided by our study helps to shed light on PKCα regulation and activation mechanism. The combined computational/experimental approach demonstrated in this work is anticipated to enable further studies to explore the roles of C1 domains in many signaling proteins and to better understand their molecular mechanisms in normal cellular function and disease development.
Collapse
Affiliation(s)
- Jianing Li
- Department
of Chemistry, Institute for Biophysical Dynamics, James Franck Institute
and Computation Institute, The University
of Chicago, 5735 South
Ellis Avenue, Chicago, Illinois 60637, United States
| | - Brian P. Ziemba
- Department
of Chemistry and Biochemistry and the Molecular Biophysics Program, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Joseph
J. Falke
- Department
of Chemistry and Biochemistry and the Molecular Biophysics Program, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Gregory A. Voth
- Department
of Chemistry, Institute for Biophysical Dynamics, James Franck Institute
and Computation Institute, The University
of Chicago, 5735 South
Ellis Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|