1
|
Ma Y, Dong T, Luan F, Yang J, Miao F, Wei P. Interaction of major facilitator superfamily domain containing 2A with the blood-brain barrier. Neural Regen Res 2025; 20:2133-2152. [PMID: 39248155 PMCID: PMC11759009 DOI: 10.4103/nrr.nrr-d-24-00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/02/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
The functional and structural integrity of the blood-brain barrier is crucial in maintaining homeostasis in the brain microenvironment; however, the molecular mechanisms underlying the formation and function of the blood-brain barrier remain poorly understood. The major facilitator superfamily domain containing 2A has been identified as a key regulator of blood-brain barrier function. It plays a critical role in promoting and maintaining the formation and functional stability of the blood-brain barrier, in addition to the transport of lipids, such as docosahexaenoic acid, across the blood-brain barrier. Furthermore, an increasing number of studies have suggested that major facilitator superfamily domain containing 2A is involved in the molecular mechanisms of blood-brain barrier dysfunction in a variety of neurological diseases; however, little is known regarding the mechanisms by which major facilitator superfamily domain containing 2A affects the blood-brain barrier. This paper provides a comprehensive and systematic review of the close relationship between major facilitator superfamily domain containing 2A proteins and the blood-brain barrier, including their basic structures and functions, cross-linking between major facilitator superfamily domain containing 2A and the blood-brain barrier, and the in-depth studies on lipid transport and the regulation of blood-brain barrier permeability. This comprehensive systematic review contributes to an in-depth understanding of the important role of major facilitator superfamily domain containing 2A proteins in maintaining the structure and function of the blood-brain barrier and the research progress to date. This will not only help to elucidate the pathogenesis of neurological diseases, improve the accuracy of laboratory diagnosis, and optimize clinical treatment strategies, but it may also play an important role in prognostic monitoring. In addition, the effects of major facilitator superfamily domain containing 2A on blood-brain barrier leakage in various diseases and the research progress on cross-blood-brain barrier drug delivery are summarized. This review may contribute to the development of new approaches for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yilun Ma
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taiwei Dong
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Fei Luan
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Juanjuan Yang
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| | - Feng Miao
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| |
Collapse
|
2
|
Luo J, Peng S, Jiang Z, Wang Q, Zhang M, Zeng Y, Yuan Y, Xia M, Hong Z, Yan Y, Tan Y, Tang J, Xie C, Gong Y. Roles and therapeutic opportunities of ω-3 long-chain polyunsaturated fatty acids in lung cancer. iScience 2025; 28:111601. [PMID: 39834867 PMCID: PMC11742864 DOI: 10.1016/j.isci.2024.111601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Over the past decades, researchers have continuously investigated the potential functions of long-chain polyunsaturated fatty acids (LCPUFAs) in cancers, including lung cancer. The ω-3 LCPUFAs, primarily consisting of eicosapentaenoic acid and docosahexaenoic acid, were found to modify inflammatory tumor microenvironment, induce cancer cell apoptosis and autophagy, and suppress tumor development when administered alone or with other therapeutical strategies. Although the precise anti-tumor mechanism has not been elucidated yet, ω-3 LCPUFAs are often used in the nutritional treatment of patients with cancer due to their ability to significantly improve patient's nutritional status, increase the sensitivity of tumor cells to treatments, and alleviate cancer-related complications. Here we present the key roles of ω-3 LCPUFAs as dietary supplementations in lung cancer, comprehensively review the recent progress on the underlying mechanisms of cancer cell regulation by ω-3 LCPUFAs, and introduce the application of ω-3 LCPUFAs in the clinical management of lung cancer and its malignant complications.
Collapse
Affiliation(s)
- Jiang Luo
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shu Peng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyu Jiang
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qingwei Wang
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mini Zhang
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuxin Zeng
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Yuan
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Xia
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zixi Hong
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yufei Yan
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yushuang Tan
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiawen Tang
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Pulmonary Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Hammoud MK, Meena C, Dietze R, Hoffmann N, Szymanski W, Finkernagel F, Nist A, Stiewe T, Graumann J, von Strandmann EP, Müller R. Arachidonic acid impairs natural killer cell functions by disrupting signaling pathways driven by activating receptors and reactive oxygen species. Cell Commun Signal 2024; 22:555. [PMID: 39563446 PMCID: PMC11575453 DOI: 10.1186/s12964-024-01940-z] [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: 05/27/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND High levels of the polyunsaturated fatty acid arachidonic acid (AA) within the ovarian carcinoma (OC) microenvironment correlate with reduced relapse-free survival. Furthermore, OC progression is tied to compromised immunosurveillance, partially attributed to the impairment of natural killer (NK) cells. However, potential connections between AA and NK cell dysfunction in OC have not been studied. METHODS We employed a combination of phosphoproteomics, transcriptional profiling and biological assays to investigate AA's impact on NK cell functions. RESULTS AA (i) disrupts interleukin-2/15-mediated expression of pro-inflammatory genes by inhibiting STAT1-dependent signaling, (ii) hampers signaling by cytotoxicity receptors through disruption of their surface expression, (iii) diminishes phosphorylation of NKG2D-induced protein kinases, including ERK1/2, LYN, MSK1/2 and STAT1, and (iv) alters reactive oxygen species production by transcriptionally upregulating detoxification. These modifications lead to a cessation of NK cell proliferation and a reduction in cytotoxicity. CONCLUSION Our findings highlight significant AA-induced alterations in the signaling network that regulates NK cell activity. As low expression of several NK cell receptors correlates with shorter OC patient survival, these findings suggest a functional linkage between AA, NK cell dysfunction and OC progression.
Collapse
Affiliation(s)
- Mohamad K Hammoud
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Institute of Physiological Chemistry, Philipps University, Marburg, Germany
| | - Celina Meena
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Raimund Dietze
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Nathalie Hoffmann
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Witold Szymanski
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Hans-Meerwein-Strasse 3, 35043, Marburg, Germany.
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Hans-Meerwein-Strasse 3, 35043, Marburg, Germany.
| |
Collapse
|
4
|
Singh V, Rochakim N, Ferraresso F, Choudhury A, Kastrup CJ, Ahn HJ. Caveolin-1 and Aquaporin-4 as Mediators of Fibrinogen-Driven Cerebrovascular Pathology in Hereditary Cerebral Amyloid Angiopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623066. [PMID: 39605467 PMCID: PMC11601418 DOI: 10.1101/2024.11.11.623066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hereditary Cerebral Amyloid Angiopathy (HCAA) is a rare inherited form of CAA, characterized by increased vascular deposits of amyloid peptides. HCAA provides a unique opportunity to study the pathogenic mechanisms linked to CAA, as it is associated with severe cerebrovascular pathology. Some of HCAA-associated amyloid-β (Aβ) mutations significantly enhance the interaction between fibrinogen and Aβ, resulting in altered fibrin structure and co-deposition with Aβ in the perivascular space. However, the mechanisms underlying perivascular fibrinogen deposition and the associated cerebrovascular pathology in HCAA remain unclear. To investigate this, we analyzed TgSwDI transgenic mice carrying HCAA-associated mutations and observed a significant age-dependent increase in fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization in the perivascular space. Moreover, Caveolin-1, a protein involved in non-specific transcytosis across the endothelium, significantly increased with age in TgSwDI mice and correlated with fibrin(ogen) extravasation. Additionally, we noted significant aquaporin-4 (AQP4) depolarization in the CAA-laden blood vessels of TgSwDI mice, which also correlated with fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization. Given that AQP4 plays a crucial role in Aβ clearance via the glymphatic pathway, its depolarization may disrupt this critical clearance mechanism, thereby exacerbating CAA pathology. To further explore the relationship between fibrin(ogen) and these factors, we depleted fibrinogen in TgSwDI mice using siRNA against fibrinogen. This intervention resulted in decreased CAA, reduced caveolin-1 levels, attenuated microglial activation, restored polarized expression of AQP4, and improved spatial memory in fibrinogen-depleted TgSwDI mice. These findings suggest that targeting fibrinogen could be a promising strategy for mitigating CAA pathology and its associated cerebrovascular pathology. Significance Statement Our study reveals the mechanism by which fibrin(ogen)-Aβ colocalization could exacerbates CAA pathology. Our findings highlight that the age-dependent increase of endothelial caveolin-1 could facilitate fibrin(ogen) extravasation, which binds with Aβ in the perivascular space inducing microglial neuroinflammation and AQP4 depolarization, thus exacerbating CAA pathology. Furthermore, fibrinogen depletion could mitigate CAA severity, reduce microglial activation, restore AQP4 polarization and memory impairment. These results suggest that targeting fibrinogen and caveolin-1-mediated transcytosis may offer new strategies to address CAA-associated cerebrovascular pathology.
Collapse
|
5
|
Buchanan CDC, Ashraf R, Hillyer LM, Tu W, Kang JX, Subedi S, Ma DWL. RNA-Seq Analysis of Pubertal Mammary Epithelial Cells Reveals Novel n-3 Polyunsaturated Fatty Acid Transcriptomic Changes in the fat-1 Mouse Model. Nutrients 2024; 16:3925. [PMID: 39599711 PMCID: PMC11597760 DOI: 10.3390/nu16223925] [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: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The early exposure of nutrients during pubertal mammary gland development may reduce the risk of developing breast cancer later in life. Anticancer n-3 polyunsaturated fatty acids (n-3 PUFA) are shown to modulate pubertal mammary gland development; however, the mechanisms of action remain unclear. Prior work focused on effects at the whole tissue level, and little is known at the cellular level, such as at the level of mammary epithelial cells (MECs), which are implicated in cancer development. METHODS This pilot study examined the effects of lifelong n-3 PUFA exposure on the transcriptome by RNA-Seq in the isolated MECs of pubertal (6-8-week-old) female fat-1 transgenic mice capable of de novo n-3 PUFA synthesis. edgeR and DESeq2 were used separately for the differential expression analysis of RNA sequencing data followed by the Benjamani-Hochberg procedure for multiple testing correction. RESULTS Nine genes were found concordant and significantly different (p ≤ 0.05) by both the DESeq2 and edgeR methods. These genes were associated with multiple pathways, suggesting that n-3 PUFA stimulates estrogen-related signaling (Mlltl0, Galr3, and Nrip1) and a glycolytic profile (Soga1, Pdpr, and Uso1) while offering protective effects for immune and DNA damage responses (Glpd1, Garre1, and Rpa1) in MECs during puberty. CONCLUSIONS This pilot study highlights the utility of RNA-Seq to better understanding the mechanistic effects of specific nutrients such as n-3 PUFA in a cell-specific manner. Thus, further studies are warranted to investigate the cell-specific mechanisms by which n-3 PUFA influences pubertal mammary gland development and breast cancer risk later in life.
Collapse
Affiliation(s)
- Connor D. C. Buchanan
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (C.D.C.B.); (R.A.); (L.M.H.)
| | - Rahbika Ashraf
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (C.D.C.B.); (R.A.); (L.M.H.)
| | - Lyn M. Hillyer
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (C.D.C.B.); (R.A.); (L.M.H.)
| | - Wangshu Tu
- School of Mathematics and Statistics, Carleton University, Ottawa, ON K1S 5B6, Canada; (W.T.); (S.S.)
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA;
- Omega-3 and Global Health Institute, Boston, MA 02129, USA
| | - Sanjeena Subedi
- School of Mathematics and Statistics, Carleton University, Ottawa, ON K1S 5B6, Canada; (W.T.); (S.S.)
| | - David W. L. Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (C.D.C.B.); (R.A.); (L.M.H.)
| |
Collapse
|
6
|
Lyudinina AY, Parshukova OI, Bojko ER. n-3 Polyunsaturated Fatty Acids Are Associated with Stable Nitric Oxide Metabolites in Highly Trained Athletes. Cells 2024; 13:1110. [PMID: 38994963 PMCID: PMC11240318 DOI: 10.3390/cells13131110] [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: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the relationships between levels of n-3 essential polyunsaturated fatty acids (n-3 PUFAs) and stable nitric oxide (NO) metabolites in the plasma of athletes. METHODS Highly trained cross-country skiers (males, n = 39) were examined. The fatty acid profile of the total plasma lipids was determined by gas chromatography. The plasma NO level was studied by a colorimetric method via reaction with Griess reagent. RESULTS A widespread deficiency of essential n-3 PUFAs in the plasma of athletes (more than 80% of the subjects) was demonstrated in association with an imbalance in the levels of nitrates (NO3) and nitrites (NO2). A lower value of n-3 linolenic acid in the plasma (0.21 mol/%) was associated with a NO3 level below the normal range (n-3 C18:3 and NO3 Rs = 0.461; p = 0.003). Higher levels of n-3 eicosapentaenoic acid (0.8 mol/%) were associated with a concentration of NO2 above the normal value (n-3 C20:5 and NO2 Rs = 0.449; p = 0.004). CONCLUSION For the first time, the participation of essential n-3 PUFAs in the nitrite-nitrate pathway of NO synthesis in highly trained skiers was demonstrated.
Collapse
Affiliation(s)
| | | | - Evgeny R. Bojko
- Institute of Physiology, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya Str., 167982 Syktyvkar, Russia; (A.Y.L.); (O.I.P.)
| |
Collapse
|
7
|
Xiong W, Jiang X, He J, Zhong Y, Ge X, Liu B, Zeng F. Isolation and identification of active components from Grifola frondosa and its anti-EV71 virus effect. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4453-4464. [PMID: 38323723 DOI: 10.1002/jsfa.13332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND It is reported that anti-enterovirus 71 (EV71) drugs have some side effects on human health. Notably, fungi plays a crucial role in promoting human health and anti-virus. Grifola frondosa is a type of large medicinal and edible fungi, rich in active substances. The present study aimed to investigate the anti-EV71 effect of G. frondosa and the potential active substances. RESULTS In the present study, the water extract of G. frondosa was subjected to ethanol precipitation to obtain the water-extracted supernatant of G. frondosa (GFWS) and water-extracted precipitation of G. frondosa. Their inhibitory effects on EV71 virus were studied based on a cell model. The results showed that GFWS had stronger security and anti-EV71 effects. In addition, the chemical constituents of GFWS were identified by ultra-high performance liquid chromatography-tandem mass spectrometry, which were selected for further separation and purification. Three compounds, N-butylaniline, succinic acid and l-tryptophan, were isolated from GFWS by NMR spectroscopy. It is noteworthy that N-butylaniline and l-tryptophan were isolated and identified from the G. frondosa fruiting bodies for the first time. Our study found that l-tryptophan has anti-EV71 virus activity, which reduced EV71-induced apoptosis and significantly inhibited the replication process after virus adsorption. Furthermore, it could also bind to capsid protein VP1 to prevent the virus from attaching to the cells. CONCLUSION l-tryptophan was an inhibitor of the EV71 virus, which could be used in infant nutrition and possibly provide a new drug to treat hand, foot and mouth disease. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenyu Xiong
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoqin Jiang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Junqiang He
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yue Zhong
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaodong Ge
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
- National Engineering Research Center of JUNCAO Technology, Fuzhou, China
| | - Feng Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing, Fujian Agriculture and Forestry University, Fuzhou, China
- National Engineering Research Center of JUNCAO Technology, Fuzhou, China
| |
Collapse
|
8
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [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: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
9
|
Luan M, Wang J, Liang K, Li B, Liu K. Association between the intake of dietary n3 and n6 fatty acids and stroke in US adults: A cross-sectional study of NHANES 2007-2018. PLoS One 2023; 18:e0293893. [PMID: 37971997 PMCID: PMC10653462 DOI: 10.1371/journal.pone.0293893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/21/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The association between the intake of dietary n3 and n6 fatty acids and the risk of stroke is subject to debate. The primary objective of the present research was to establish the correlation in a large sample of American adults. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) between 2007 and 2018, the association of the intake of dietary n3 and n6 fatty acids with stroke events was analyzed in a sample of 29,459 adults. The intake of n3 and n6 fatty acids intake was assessed though two 24-h dietary recalls. Stroke outcomes were identified based on the responses provided in self-reported questionnaire. Logistic regression was fitted to evaluate the correlation of dietary n3, n6 fatty acids intake with stroke events. RESULTS Subjects in the highest tertile (T3) of dietary n3 (OR: 0.67, 95% CI: 0.49-0.93), n6 (OR: 0.65, 95% CI: 0.45-0.95) fatty acids intake were found to have obviously lower risk of stroke compared to those in the lowest tertile (T1), but the n6:n3 ratio was not found to be associated with a stroke event. Results from stratified analysis demonstrated that dietary n3 fatty acids had an inverse correlation of stroke in both male and female, but dietary n6 fatty acids only had this correlation in male. Moreover, findings were made that the interaction was significant in terms of age in the subgroup analysis, and the negative relationship between the intake of dietary n3 and n6 fatty acids and stroke event were particularly pronounced among participants aged ≥60. CONCLUSIONS The present results suggested that increased dietary n3, n6 fatty acids intake correlated with a lower risk of stroke.
Collapse
Affiliation(s)
- Mingya Luan
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| | - Jia Wang
- Public Health Department, Weihai Maternal and Child Health Hospital, the Affiliated Weihai Second Municipal Hospital of Qingdao University, Weihai, China
| | - Kun Liang
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| | - Bo Li
- Zhangcun Town Health Center in Huancui District, Weihai, China
| | - Kewei Liu
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| |
Collapse
|
10
|
Bhowmick S, Biswas T, Ahmed M, Roy D, Mondal S. Caveolin-1 and lipids: Association and their dualism in oncogenic regulation. Biochim Biophys Acta Rev Cancer 2023; 1878:189002. [PMID: 37848094 DOI: 10.1016/j.bbcan.2023.189002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
Caveolin-1 (Cav-1) is a structural protein of caveolae that functions as a molecular organizer for different cellular functions including endocytosis and cellular signaling. Cancer cells take advantage of the physical position of Cav-1, as it can communicate with extracellular matrix, help to organize growth factor receptors, redistribute cholesterol and glycosphingolipids, and finally transduce signals within the cells for oncogenesis. Recent studies emphasize the exceeding involvement of Cav-1 with different lipid bodies and in altering the metabolism, especially lipid metabolism. However, the association of Cav-1 with different lipid bodies like lipid rafts, lipid droplets, cholesterols, sphingolipids, and fatty acids is remarkably dynamic. The lipid-Cav-1 alliance plays a dual role in carcinogenesis. Both cancer progression and regression are modified and affected by the type of lipid molecule's association with Cav-1. Accordingly, this Cav-1-lipid cooperation exemplifies a cancer-type-specific treatment strategy for a better prognosis of the disease. In this review, we first present Cav-1 as an oncogenic molecule and its communication via lipid raft. We discussed the involvement of Cav-1 with lipid droplets, Cholesterol, sphingolipids, gangliosides, and ceramides. Further, we describe the Cav-1-mediated altered Fatty acid metabolism in cancer and the strategic therapeutic approaches toward Cav-1 targeting.
Collapse
Affiliation(s)
- Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Tannishtha Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, USA
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
11
|
Reche-García C, Hernández Morante JJ, Morillas-Ruiz JM. Dietary Assessment for Weight Management and Health Maintenance. Nutrients 2023; 15:4610. [PMID: 37960263 PMCID: PMC10649168 DOI: 10.3390/nu15214610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023] Open
Abstract
An adequate dietary assessment is essential for improving the eating habits of the population and preventing health problems such as obesity and cardiovascular diseases [...].
Collapse
Affiliation(s)
- Cristina Reche-García
- Eating Disorders Research Unit, Campus de Los Jerónimos, Universidad Católica de Murcia, Guadalupe, 30107 Murcia, Spain;
| | - Juan José Hernández Morante
- Eating Disorders Research Unit, Campus de Los Jerónimos, Universidad Católica de Murcia, Guadalupe, 30107 Murcia, Spain;
| | - Juana M. Morillas-Ruiz
- Faculty of Pharmacy and Nutrition, Campus de Los Jerónimos, Universidad Católica de Murcia, Guadalupe, 30107 Murcia, Spain
| |
Collapse
|
12
|
Kenworthy AK, Han B, Ariotti N, Parton RG. The Role of Membrane Lipids in the Formation and Function of Caveolae. Cold Spring Harb Perspect Biol 2023; 15:a041413. [PMID: 37277189 PMCID: PMC10513159 DOI: 10.1101/cshperspect.a041413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Caveolae are plasma membrane invaginations with a distinct lipid composition. Membrane lipids cooperate with the structural components of caveolae to generate a metastable surface domain. Recent studies have provided insights into the structure of essential caveolar components and how lipids are crucial for the formation, dynamics, and disassembly of caveolae. They also suggest new models for how caveolins, major structural components of caveolae, insert into membranes and interact with lipids.
Collapse
Affiliation(s)
- Anne K Kenworthy
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| | - Bing Han
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| | - Nicholas Ariotti
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, 4072 Brisbane, Australia
| |
Collapse
|
13
|
Delgado-Alarcón JM, Hernández Morante JJ, Morillas-Ruiz JM. Modification of Breakfast Fat Composition Can Modulate Cytokine and Other Inflammatory Mediators in Women: A Randomized Crossover Trial. Nutrients 2023; 15:3711. [PMID: 37686743 PMCID: PMC10489665 DOI: 10.3390/nu15173711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Previous trials have demonstrated that modifying dietary fat composition can influence the production of inflammation-related factors. Additionally, it has been suggested that not only the type of fat, but also the timing of fat intake can impact these factors. Therefore, the objective of the present study was to evaluate the effect of altering breakfast fat composition on inflammatory parameters. A 3-month crossover randomized trial was designed, involving 60 institutionalized women who alternately consumed a breakfast rich in polyunsaturated fatty acids (PUFA) (margarine), monounsaturated fatty acids (MUFA) (virgin olive oil), or saturated fatty acids (SFA) (butter), based on randomization. The following inflammatory markers were evaluated: epidermal growth factor (EGF), interferon (IFN)-α, interleukin (IL)-1α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, C-reactive protein (CRP), and vascular/endothelial growth factor (VEGF). The results showed that the most significant effects were observed with the high-MUFA breakfast, as there was a statistically significant decrease in plasma IL-6 (p = 0.016) and VEGF values (p = 0.035). Other factors, such as IL-1α and CRP, also decreased substantially, but did not reach the statistically significant level. On the other hand, the high-PUFA breakfast induced a significant decrease in EGF levels (p < 0.001), whereas the high-SFA breakfast had no apparent effect on these factors. In conclusion, modifying breakfast fat, particularly by increasing MUFA or PUFA intake, appears to be sufficient for promoting a lower inflammatory marker synthesis profile and may be beneficial in improving cardiovascular complications.
Collapse
Affiliation(s)
- Jessica M. Delgado-Alarcón
- Department of Food Technology and Nutrition, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain;
| | - Juan José Hernández Morante
- Eating Disorders Research Unit, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain
| | - Juana M. Morillas-Ruiz
- Department of Food Technology and Nutrition, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain;
| |
Collapse
|
14
|
Zhu S, Liu Q, Xiang X, Cui K, Zhao F, Mai K, Ai Q. Docosahexaenoic Acid Ameliorates the Toll-Like Receptor 22-Triggered Inflammation in Fish by Disrupting Lipid Raft Formation. J Nutr 2022; 152:1991-2002. [PMID: 35679100 DOI: 10.1093/jn/nxac125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Although dietary DHA alleviates Toll-like receptor (TLR)-associated chronic inflammation in fish, the underlying mechanism is not well understood. OBJECTIVES This study aimed to explore the role of Tlr22 in the innate immunity of large yellow croaker and investigate the anti-inflammatory effects of DHA on Tlr22-triggered inflammation. METHODS Head kidney-derived macrophages of croaker and HEK293T cells were or were not pretreated with 100 μM DHA for 10 h prior to polyinosinic-polycytidylic acid (poly I:C) stimulation. We executed qRT-PCR, immunoblotting, and lipidomic analysis to examine the impact of DHA on Tlr22-triggered inflammation and membrane lipid composition. In vivo, croakers (12.03 ± 0.05 g) were fed diets containing 0.2% [control (Ctrl)], 0.8%, and 1.6% DHA for 8 wk before injection with poly I:C. Inflammatory genes expression and rafts-related lipids and protein expression were measured in the head kidney. Data were analyzed by ANOVA or Student t test. RESULTS The activation of Tlr22 by poly I:C induced inflammation, and DHA diminished Tlr22-targeted inflammatory gene expression by 56-73% (P ≤ 0.05). DHA reduced membrane sphingomyelin (SM) and SFA-containing phosphatidylcholine (SFA-PC) contents, as well as lipid raft marker caveolin 1 amounts. Furthermore, lipid raft disruption suppressed Tlr22-induced Nf-κb and interferon h activation and p65 nuclear translocation. In vivo, expression of Tlr22 target inflammatory genes was 32-64% lower in the 1.6% DHA group than in the Ctrl group upon poly I:C injection (P ≤ 0.05). Also, the 1.6% DHA group showed a reduction in membrane SM and SFA-PC contents, accompanied by a decrease in caveolin 1 amounts, compared with the Ctrl group. CONCLUSIONS The activation of Tlr22 signaling depends on lipid rafts, and DHA ameliorates the Tlr22-triggered inflammation in both head kidney and head kidney-derived macrophages of croaker partially by altering membrane SMs and SFA-PCs that are required for lipid raft organization.
Collapse
Affiliation(s)
- Si Zhu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Kun Cui
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| |
Collapse
|
15
|
Abstract
Several studies have reported a significant association between the metabolic syndrome (MetS) and mortality around the world. Caveolin-1 (CAV-1) has been widely studied in dyslipidaemia, and several studies have indicated that CAV-1 genetic variations may correlate with dietary intake of fatty acids. This study aimed to investigate the interaction of CAV-1 rs3807992 with types of dietary fatty acid in the MetS risk. This cross-sectional study was carried out on 404 overweight and obese females. Dietary intake was obtained from a 147-item FFQ. The CAV-1 genotype was measured using the PCR-restriction fragment length polymorphism method. Anthropometric values and serum levels (TC, LDL, HDL, TAG and FBS) were measured by standard methods. It was observed that the (AA + AG) group had significantly higher BMI, waist circumference and DBP (P = 0·02, P = 0·02, and P = 0·01, respectively) and lower serum LDL, HDL and TC (P < 0·05) than the GG group. It was found that A allele carriers were at higher odds of the MetS (P = 0·01), abdominal obesity (P = 0·06), increased TAG concentration (P = 0·01), elevated blood pressure (BP) (P = 0·01), increased glucose concentration (P = 0·45) and decreased HDL-cholesterol concentration (P = 0·03). Moreover, the interaction of CAV-1 and SFA intake was significant in terms of the MetS (P = 0·03), LDL (P = 0·03) and BP (P = 0·01). Additionally, the (AA + AG) group was significantly related to PUFA intake in terms of the MetS (P = 0·04), TAG (P = 0·02), glucose (P = 0·02) and homoeostasis model assessment insulin resistance (P = 0·01). Higher PUFA consumption might attenuate the CAV-1 rs3807992 associations with the MetS, and individuals with greater genetic predisposition appeared to have a higher risk of the MetS, associated with higher SFA consumption.
Collapse
|
16
|
Parnova RG. Critical Role of Endothelial Lysophosphatidylcholine Transporter Mfsd2a in Maintaining Blood–Brain Barrier Integrity and Delivering Omega 3 PUFA to the Brain. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022030103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Hammoud MK, Dietze R, Pesek J, Finkernagel F, Unger A, Bieringer T, Nist A, Stiewe T, Bhagwat AM, Nockher WA, Reinartz S, Müller-Brüsselbach S, Graumann J, Müller R. Arachidonic acid, a clinically adverse mediator in the ovarian cancer microenvironment, impairs JAK-STAT signaling in macrophages by perturbing lipid raft structures. Mol Oncol 2022; 16:3146-3166. [PMID: 35451191 PMCID: PMC9441005 DOI: 10.1002/1878-0261.13221] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/29/2022] [Accepted: 04/20/2022] [Indexed: 11/08/2022] Open
Abstract
Survival of ovarian carcinoma is associated with the abundance of immunosuppressed CD163highCD206high tumor‐associated macrophages (TAMs) and high levels of arachidonic acid (AA) in the tumor microenvironment. Here, we show that both associations are functionally linked. Transcriptional profiling revealed that high CD163 and CD206/MRC1 expression in TAMs is strongly associated with an inhibition of cytokine‐triggered signaling, mirrored by an impaired transcriptional response to interferons and IL‐6 in monocyte‐derived macrophages by AA. This inhibition of pro‐inflammatory signaling is caused by dysfunctions of the cognate receptors, indicated by the inhibition of JAK1, JAK2, STAT1, and STAT3 phosphorylation, and by the displacement of the interferon receptor IFNAR1, STAT1 and other immune‐regulatory proteins from lipid rafts. AA exposure led to a dramatic accumulation of free AA in lipid rafts, which appears to be mechanistically crucial, as the inhibition of its incorporation into phospholipids did not affect the AA‐mediated interference with STAT1 phosphorylation. Inhibition of interferon‐triggered STAT1 phosphorylation by AA was reversed by water‐soluble cholesterol, known to prevent the perturbation of lipid raft structure by AA. These findings suggest that the pharmacologic restoration of lipid raft functions in TAMs may contribute to the development new therapeutic approaches.
Collapse
Affiliation(s)
- Mohamad K Hammoud
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Raimund Dietze
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Jelena Pesek
- Medical Mass Spectrometry Core Facility, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Annika Unger
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Tim Bieringer
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.,Hochschule Landshut, 84036, Landshut, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Aditya M Bhagwat
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - W Andreas Nockher
- Medical Mass Spectrometry Core Facility, Philipps University, Marburg, Germany
| | - Silke Reinartz
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | | | - Johannes Graumann
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Institute for Translational Proteomics, Philipps University, Marburg, Germany
| | - Rolf Müller
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
18
|
Fuentes NR, Mlih M, Wang X, Webster G, Cortes-Acosta S, Salinas ML, Corbin IR, Karpac J, Chapkin RS. Membrane therapy using DHA suppresses epidermal growth factor receptor signaling by disrupting nanocluster formation. J Lipid Res 2021; 62:100026. [PMID: 33515553 PMCID: PMC7933808 DOI: 10.1016/j.jlr.2021.100026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) signaling drives the formation of many types of cancer, including colon cancer. Docosahexaenoic acid (DHA, 22∶6Δ4,7,10,13,16,19), a chemoprotective long-chain n-3 polyunsaturated fatty acid suppresses EGFR signaling. However, the mechanism underlying this phenotype remains unclear. Therefore, we used super-resolution microscopy techniques to investigate the mechanistic link between EGFR function and DHA-induced alterations to plasma membrane nanodomains. Using isogenic in vitro (YAMC and IMCE mouse colonic cell lines) and in vivo (Drosophila, wild type and Fat-1 mice) models, cellular DHA enrichment via therapeutic nanoparticle delivery, endogenous synthesis, or dietary supplementation reduced EGFR-mediated cell proliferation and downstream Ras/ERK signaling. Phospholipid incorporation of DHA reduced membrane rigidity and the size of EGFR nanoclusters. Similarly, pharmacological reduction of plasma membrane phosphatidic acid (PA), phosphatidylinositol-4,5-bisphosphate (PIP2) or cholesterol was associated with a decrease in EGFR nanocluster size. Furthermore, in DHA-treated cells only the addition of cholesterol, unlike PA or PIP2, restored EGFR nanoscale clustering. These findings reveal that DHA reduces EGFR signaling in part by reshaping EGFR proteolipid nanodomains, supporting the feasibility of using membrane therapy, i.e., dietary/drug-related strategies to target plasma membrane organization, to reduce EGFR signaling and cancer risk.
Collapse
Affiliation(s)
- Natividad R Fuentes
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA; Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA
| | - Mohamed Mlih
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Gabriella Webster
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Sergio Cortes-Acosta
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Michael L Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Ian R Corbin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA; Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
20
|
Asico LD, Rozyyev S, Crusan AM, Jose PA, Villar VAM. Elucidating the Role of Lipid Rafts on G Protein-Coupled Receptor Function in the Mouse Kidney: An In Vivo Approach. Methods Mol Biol 2021; 2187:187-206. [PMID: 32770507 DOI: 10.1007/978-1-0716-0814-2_10] [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] [Indexed: 12/11/2022]
Abstract
Numerous G protein-coupled receptors (GPCRs) and GPCR-signaling molecules reside in lipid rafts and thus, are inherently regulated in these microdomains. However, the limitations of current methods to investigate lipid raft biology and GPCR activity in situ have hindered the complete understanding of the molecular underpinnings of GPCR trafficking and signaling, especially in the whole organism. This book chapter details an innovative in vivo approach to study the crucial role of lipid rafts on the workings of GPCRs in the mouse kidney. This protocol involves the use of a modified mini osmotic pump to deliver an agent that selectively disrupts the lipid raft in the kidney.
Collapse
Affiliation(s)
- Laureano D Asico
- Division of Renal Diseases and Hypertension, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Selim Rozyyev
- Division of Renal Diseases and Hypertension, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, DC, USA
| | - Annabelle M Crusan
- Research Animal Facility, The Children's Research Institute, Children's National Health System, Washington, DC, USA
| | - Pedro A Jose
- Division of Renal Diseases and Hypertension, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Van Anthony M Villar
- Division of Renal Diseases and Hypertension, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
21
|
Parton RG, Kozlov MM, Ariotti N. Caveolae and lipid sorting: Shaping the cellular response to stress. J Cell Biol 2020; 219:133844. [PMID: 32328645 PMCID: PMC7147102 DOI: 10.1083/jcb.201905071] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Caveolae are an abundant and characteristic surface feature of many vertebrate cells. The uniform shape of caveolae is characterized by a bulb with consistent curvature connected to the plasma membrane (PM) by a neck region with opposing curvature. Caveolae act in mechanoprotection by flattening in response to increased membrane tension, and their disassembly influences the lipid organization of the PM. Here, we review evidence for caveolae as a specialized lipid domain and speculate on mechanisms that link changes in caveolar shape and/or protein composition to alterations in specific lipid species. We propose that high membrane curvature in specific regions of caveolae can enrich specific lipid species, with consequent changes in their localization upon caveolar flattening. In addition, we suggest how changes in the association of lipid-binding caveolar proteins upon flattening of caveolae could allow release of specific lipids into the bulk PM. We speculate that the caveolae-lipid system has evolved to function as a general stress-sensing and stress-protective membrane domain.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicholas Ariotti
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Kensington, Australia.,Department of Pathology, School of Medical Sciences, The University of New South Wales, Kensington, Australia
| |
Collapse
|
22
|
Taga H, Dallaire MP, Gervais R, Richard FJ, Ma L, Corl BA, Chouinard PY. Characterization of raft microdomains in bovine mammary tissue during lactation: How they are modulated by fatty acid treatments. J Dairy Sci 2020; 104:2384-2395. [PMID: 33246605 DOI: 10.3168/jds.2020-19267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/03/2020] [Indexed: 11/19/2022]
Abstract
The objective of the current study was first to characterize lipid raft microdomains isolated as detergent-resistant membranes (DRM) from mammary gland tissue, and second to determine how dietary fatty acids (FA) such as conjugated linoleic acid (CLA), 19:1 cyclo, and long-chain n-3 polyunsaturated FA affect lipid raft markers of mammary cells, and to finally establish relationships between these markers and lactation performance in dairy cows. Eight Holstein cows were used in a replicated 4 × 4 Latin square design with periods of 28 d. For the first 14 d, cows received daily an abomasal infusion of (1) 406 g of a saturated FA supplement (112 g of 16:0 + 230 g of 18:0) used as a control; (2) 36 g of a CLA supplement (13.9 g of trans-10,cis-12 18:2) + 370 g of saturated FA; (3) 7 g of Sterculia fetida oil (3.1 g of 19:1 cyclo, STO) + 399 g of saturated FA; or (4) 406 g of fish oil (55.2 g of cis-5,cis-8,cis-11,cis-14,cis-17 20:5 + 59.3 g of cis-4,cis-7,cis-10,cis-13,cis-16,cis-19 22:6, FO). Mammary biopsies were harvested on d 14 of each infusion period and were followed by a 14-d washout interval. Cholera toxin subunit B, which specifically binds to ganglioside M-1 (GM-1), a lipid raft marker, was used to assess its distribution in DRM. Infusions of CLA, STO, and FO were individually compared with the control, and significance was declared at P ≤ 0.05. Milk fat yield was decreased with CLA and FO, but was not affected by STO. Milk lactose yield was decreased with CLA and STO, but was not affected by FO. Mammary tissue shows a strong GM-1-signal enrichment in isolated DRM from mammary gland tissue. Caveolin (CAV) and flotillin (FLOT) are 2 proteins considered as lipid raft markers and they are present in DRM from mammary gland tissue. Distributions of GM-1, CAV-1, and FLOT-1 showed an effect of treatments determined by their subcellular distributions in sucrose gradient fractions. Regardless of treatments, data showed positive relationships between the yield of milk fat, protein, and lactose, and the abundance GM-1 in DRM fraction. Milk protein yield was positively correlated with relative proportion of FLOT-1 in the soluble fraction, whereas lactose yield was positively correlated with relative proportion of CAV-1 in the DRM fractions. Infusion of CLA decreased mRNA abundance of CAV-1, FLOT-1, and FLOT-2. Regardless of treatments, a positive relationship was observed between fat yield and mRNA abundance of FLOT-2. In conclusion, although limited to a few markers, results of the current experiment raised potential links between variation in specific biologically active component of raft microdomains in bovine mammary gland and lactation performances in dairy cows.
Collapse
Affiliation(s)
- H Taga
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - M P Dallaire
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - R Gervais
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - F J Richard
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada
| | - L Ma
- Department of Dairy Science, Virginia Tech, Blacksburg 24061
| | - B A Corl
- Department of Dairy Science, Virginia Tech, Blacksburg 24061
| | - P Y Chouinard
- Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada.
| |
Collapse
|
23
|
Del Pozo MA, Lolo FN, Echarri A. Caveolae: Mechanosensing and mechanotransduction devices linking membrane trafficking to mechanoadaptation. Curr Opin Cell Biol 2020; 68:113-123. [PMID: 33188985 DOI: 10.1016/j.ceb.2020.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Mechanical forces (extracellular matrix stiffness, vascular shear stress, and muscle stretching) reaching the plasma membrane (PM) determine cell behavior. Caveolae are PM-invaginated nanodomains with specific lipid and protein composition. Being highly abundant in mechanically challenged tissues (muscles, lungs, vessels, and adipose tissues), they protect cells from mechanical stress damage. Caveolae flatten upon increased PM tension, enabling both force sensing and accommodation, critical for cell mechanoprotection and homeostasis. Thus, caveolae are highly plastic, ranging in complexity from flattened membranes to vacuolar invaginations surrounded by caveolae-rosettes-which also contribute to mechanoprotection. Caveolar components crosstalk with mechanotransduction pathways and recent studies show that they translocate from the PM to the nucleus to convey stress information. Furthermore, caveolae components can regulate membrane traffic from/to the PM to adapt to environmental mechanical forces. The interdependence between lipids and caveolae starts to be understood, and the relevance of caveolae-dependent membrane trafficking linked to mechanoadaption to different physiopathological processes is emerging.
Collapse
Affiliation(s)
- Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
24
|
Newell M, Patel D, Goruk S, Field CJ. Docosahexaenoic Acid Incorporation Is Not Affected by Doxorubicin Chemotherapy in either Whole Cell or Lipid Raft Phospholipids of Breast Cancer Cells in vitro and Tumor Phospholipids in vivo. Lipids 2020; 55:549-565. [PMID: 32588470 DOI: 10.1002/lipd.12252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/07/2022]
Abstract
To better understand how docosahexaenoic acid (DHA) improves the effects of doxorubicin (DOX), we examined DHA ± DOX on changes in whole cell and lipid raft phospholipids (PL) of MDA-MB-231 and MCF-7 breast cancer cells. We sought to confirm whether the relative changes in PL DHA content of MDA-MB-231 cells could be extended to PL from MDA-MB-231 tumors grown in mice fed a DHA supplemented diet ±DOX. Treatment with DHA did not change PL composition yet DOX increased the proportion of phosphatidylserine in MCF-7 cell lipid rafts by two-fold (p < 0.001). Regardless of DOX, the relative percent incorporation of DHA was higher in MDA-MB-231 cells compared to MCF-7 cells in phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine (whole cell and lipid rafts); and higher in phosphatidylethanolamine vs. phosphatidylcholine (4.4-fold in MCF-7 and 6-fold in MDA-MB-231 cells respectively). DHA treatment increased eicosapentaenoic acid and docosapentaenoic acid in MDA-MB-231 cells but not MCF-7 cells. Increased DHA content in MDA-MB-231 cells, MCF-7 cells, and MDA-MB-231 tumors in all PL moieties (except sphingomyelin) corresponded with reduced arachidonic acid (p < 0.05). Feeding mice 2.8% (w/w of fat) DHA ± DOX increased tumor necrotic regions (p < 0.05). This study established differential incorporation of DHA into whole cell and lipid rafts between human breast cancer cell lines. However, within each cell line, this incorporation was not altered by DOX confirming that DOX does not change membrane lipid composition. Furthermore, our findings indicate that membrane changes observed in vitro are translatable to in vivo changes and that DHA + DOX could contribute to the anticancer effects through increased necrosis.
Collapse
Affiliation(s)
- Marnie Newell
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Dhruvesh Patel
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
25
|
Salinas ML, Fuentes NR, Choate R, Wright RC, McMurray DN, Chapkin RS. AdipoRon Attenuates Wnt Signaling by Reducing Cholesterol-Dependent Plasma Membrane Rigidity. Biophys J 2020; 118:885-897. [PMID: 31630812 PMCID: PMC7036725 DOI: 10.1016/j.bpj.2019.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
The increasing prevalence of adult and adolescent obesity and its associated risk of colorectal cancer reinforces the urgent need to elucidate the underlying mechanisms contributing to the promotion of colon cancer in obese individuals. Adiponectin is an adipose tissue-derived adipokine, whose levels are reduced during obesity. Both epidemiological and preclinical data indicate that adiponectin suppresses colon tumorigenesis. We have previously demonstrated that both adiponectin and AdipoRon, a small-molecule adiponectin receptor agonist, suppress colon cancer risk in part by reducing the number of Lgr5+ stem cells in mouse colonic organoids. However, the mechanism by which the adiponectin signaling pathway attenuates colon cancer risk remains to be addressed. Here, we have hypothesized that adiponectin signaling supports colonic stem cell maintenance through modulation of the biophysical properties of the plasma membrane (PM). Specifically, we investigated the effects of adiponectin receptor activation by AdipoRon on the biophysical perturbations linked to the attenuation of Wnt-driven signaling and cell proliferation as determined by LEF luciferase reporter assay and colonic organoid proliferation, respectively. Using physicochemical sensitive dyes, Di-4-ANEPPDHQ and C-laurdan, we demonstrated that AdipoRon decreased the rigidity of the colonic cell PM. The decrease in membrane rigidity was associated with a reduction in PM free cholesterol levels and the intracellular accumulation of free cholesterol in lysosomes. These results suggest that adiponectin signaling plays a role in modulating cellular cholesterol homeostasis, PM biophysical properties, and Wnt-driven signaling. These findings are noteworthy because they may in part explain how obesity drives colon cancer progression.
Collapse
Affiliation(s)
- Michael L Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Natividad R Fuentes
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas; Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, Texas
| | - Rachel Choate
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - David N McMurray
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas; Department of Nutrition and Food Science, Texas A&M University, College Station, Texas; Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, Texas; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas; Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas; Center for Environmental Health Research, Texas A&M University, College Station, Texas.
| |
Collapse
|
26
|
Zhao C, Ma J, Wang Z, Li H, Shen H, Li X, Chen G. Mfsd2a Attenuates Blood-Brain Barrier Disruption After Sub-arachnoid Hemorrhage by Inhibiting Caveolae-Mediated Transcellular Transport in Rats. Transl Stroke Res 2020; 11:1012-1027. [PMID: 31907728 DOI: 10.1007/s12975-019-00775-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/09/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) disruption is one of the critical mechanisms of brain injury induced by subarachnoid hemorrhage (SAH). Past studies have often focused on the tight junctions of endothelial cells. However, low transcellular transport levels also play an important role in the normal functioning of the BBB. Major facilitator superfamily domain-containing 2a (Mfsd2a) has been demonstrated to be essential for the maintenance of the normal BBB. Our present study aimed to explore the roles and mechanisms of Mfsd2a in BBB disruption after SAH. In this study, a prechiasmatic cistern single-injection model was used to produce experimental SAH in Sprague-Dawley rats. Specific small-interfering RNA and plasmids were used to downregulate and upregulate the expression of Mfsd2a prior to assessments in our SAH model. Omega-3 fatty acid deficiency diet was used to reduce DHA in rat brain. The expression level of Mfsd2a decreased significantly after SAH and reached its lowest level at 72 h post-SAH, which then gradually recovered. At 72 h after SAH, BBB function was disrupted; upregulation of Mfsd2a reversed this damage, whereas downregulation of Mfsd2a exacerbated this damage. These effects were primarily mediated through transcellular transport, especially for changes in caveolae compared to those of tight junctions. After stopping the supply of omega-3 fatty acids, the effect of Mfsd2a on inhibition of caveolae and protection of the blood-brain barrier was eliminated. Taken together, Mfsd2a inhibits caveolae-based transcellular transport by transporting omega-3 fatty acids to protect the BBB after SAH.
Collapse
Affiliation(s)
- Chongshun Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Junwei Ma
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
27
|
Abstract
The prevalence of food allergy is raising in industrialized countries, but the mechanisms behind this increased incidence are not fully understood. Environmental factors are believed to play a role in allergic diseases, including lifestyle influences, such as diet. There is a close relationship between allergens and lipids, with many allergenic proteins having the ability to bind lipids. Dietary lipids exert pro-inflammatory or anti-inflammatory functions on cells of the innate immunity and influence antigen presentation to cells of the adaptive immunity. In addition to modifying the immunostimulating properties of proteins, lipids also alter their digestibility and intestinal absorption, changing allergen bioavailability. This study provides an overview of the role of dietary lipids in food allergy, taking into account epidemiological information, as well as results of mechanistic investigations using in vivo, ex vivo and in vitro models. The emerging link among high-fat diets, obesity, and allergy is also discussed.
Collapse
Affiliation(s)
- Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| |
Collapse
|
28
|
Functional link between plasma membrane spatiotemporal dynamics, cancer biology, and dietary membrane-altering agents. Cancer Metastasis Rev 2019; 37:519-544. [PMID: 29860560 DOI: 10.1007/s10555-018-9733-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cell plasma membrane serves as a nexus integrating extra- and intracellular components, which together enable many of the fundamental cellular signaling processes that sustain life. In order to perform this key function, plasma membrane components assemble into well-defined domains exhibiting distinct biochemical and biophysical properties that modulate various signaling events. Dysregulation of these highly dynamic membrane domains can promote oncogenic signaling. Recently, it has been demonstrated that select membrane-targeted dietary bioactives (MTDBs) have the ability to remodel plasma membrane domains and subsequently reduce cancer risk. In this review, we focus on the importance of plasma membrane domain structural and signaling functionalities as well as how loss of membrane homeostasis can drive aberrant signaling. Additionally, we discuss the intricacies associated with the investigation of these membrane domain features and their associations with cancer biology. Lastly, we describe the current literature focusing on MTDBs, including mechanisms of chemoprevention and therapeutics in order to establish a functional link between these membrane-altering biomolecules, tuning of plasma membrane hierarchal organization, and their implications in cancer prevention.
Collapse
|
29
|
Mentoor I, Engelbrecht AM, Nell T. Fatty acids: Adiposity and breast cancer chemotherapy, a bad synergy? Prostaglandins Leukot Essent Fatty Acids 2019; 140:18-33. [PMID: 30553399 DOI: 10.1016/j.plefa.2018.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Globally, breast cancer continues to be a major concern in women's health. Lifestyle related risk factors, specifically excess adipose tissue (adiposity) has reached epidemic proportions and has been identified as a major risk factor in the development of breast cancer. Dysfunctional adipose tissue has evoked research focusing on its association with metabolic-related conditions, breast cancer risk and progression. Adipose dysfunction in coordination with immune cells and inflammation, are responsible for accelerated cell growth and survival of cancer cells. Recently, evidence also implicates adiposity as a potential risk factor for chemotherapy resistance. Chemotherapeutic agents have been shown to negatively impact adipose tissue. Since adipose tissue is a major storage site for fatty acids, it is not unlikely that these negative effects may disrupt adipose tissue homeostasis. It is therefore argued that fatty acid composition may be altered due to the chemotherapeutic pharmacokinetics, which in turn could have severe health related outcomes. The underlying molecular mechanisms elucidating the effects of fatty acid composition in adiposity-linked drug resistance are still unclear and under explored. This review focuses on the potential role of adiposity in breast cancer and specifically emphasizes the role of fatty acids in cancer progression and treatment resistance.
Collapse
Affiliation(s)
- Ilze Mentoor
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - A-M Engelbrecht
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - Theo Nell
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa.
| |
Collapse
|
30
|
Ajith TA, Jayakumar TG. Omega-3 fatty acids in coronary heart disease: Recent updates and future perspectives. Clin Exp Pharmacol Physiol 2019; 46:11-18. [PMID: 30230571 DOI: 10.1111/1440-1681.13034] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 12/31/2022]
Abstract
Incidence of coronary heart disease (CHD) increases worldwide with varying etiological factors. In addition to the control of risk factors, dietary modification has been recommended to reduce the prevalence. Omega-3 (ω-3) fatty acids (FAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), of fish oil are beneficial for the prevention of CHD. The effect can be ascribed to anti-inflammatory, vasodilating, antiarrhythmic, antihypertensive activities and lowering of triacyl glycerol level. The American Heart Association advises two fish meals per week in subjects without CHD or supplementation of 1 g of EPA plus DHA per day in subjects with CHD. Despite the beneficial effects of EPA/DHA reported in some of the clinical trials, results of many others were inconsistent that can be ascribed to short duration of studies, low doses of ω-3 FAs, variations in the EPA:DHA ratio, selection of patients with different risk factors or interaction of ω-3 FAs with drugs used in the therapy. Therefore, well designed clinical trials in various populations are warranted. This article discusses the current situation and future prospective of ω-3 FAs in CHD.
Collapse
|
31
|
Eser Ocak P, Ocak U, Sherchan P, Zhang JH, Tang J. Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res 2018; 98:29-41. [PMID: 30345547 DOI: 10.1002/jnr.24327] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023]
Abstract
Major facilitator superfamily domain-containing protein-2a (Mfsd2a) which was considered as an orphan transporter has recently gained attention for its regulatory role in the maintenance of proper functioning of the blood-brain barrier. Besides the major role of Mfsd2a in maintaining the barrier function, increasing evidence has emerged with regard to the contributions of Mfsd2a to various biological processes such as transport, cell fusion, cell cycle, inflammation and regeneration, managing tumor growth, functioning of other organs with barrier functions or responses to injury. The purpose of this article is to review the different roles of Mfsd2a and its involvement in the physiological and pathophysiological processes primarily in the central nervous system and throughout the mammalian body under the lights of the current literature.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Umut Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
32
|
Okada LSDRR, Oliveira CP, Stefano JT, Nogueira MA, Silva IDCGD, Cordeiro FB, Alves VAF, Torrinhas RS, Carrilho FJ, Puri P, Waitzberg DL. Omega-3 PUFA modulate lipogenesis, ER stress, and mitochondrial dysfunction markers in NASH - Proteomic and lipidomic insight. Clin Nutr 2018; 37:1474-1484. [PMID: 29249532 DOI: 10.1016/j.clnu.2017.08.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Currently there is no FDA-approved therapy for nonalcoholic steatohepatitis (NASH). Increased n-6/n-3 polyunsaturated fatty acids (PUFA) ratio can induce endoplasmic reticulum (ER) stress and mitochondrial dysfunction that characterize NASH. Our recent study with n-3 PUFA showed improvement in individual histologic parameters like steatosis, ballooning and lobular inflammation. We hypothesized that n-3 PUFA therapy mediated improvement in histologic parameters is modulated by lipidomic and proteomic changes. METHODS We therefore evaluated hepatic proteomic and plasma lipidomic profiles before and after n-3 PUFA therapy in subjects with NASH. In a double-blind, randomized, placebo-controlled trial, patients with NASH received 6-month treatment with n-3 PUFA (0.945 g/day [64% alpha-linolenic (ALA), 21% eicosapentaenoic (EPA), and 16% docosahexaenoic (DHA) acids]). Paired liver biopsy and plasma collected before and after-n-3 PUFA therapy were assessed using mass spectrometry and gas chromatography for hepatic proteomics and plasma lipidomics. Data were matched to UniProt and LIPID MAPS database, respectively. Cytoscape software was used to analyze functional pathways. Twenty-seven NASH patients with paired liver histology and plasma before and after n-3 PUFA treatment were studied. RESULTS Treatment with n-3 PUFA significantly increased ALA, EPA, and glycerophospholipids, and decreased arachidonic acid (p < 0.05 for all). Further, proteomic markers of cell matrix, lipid metabolism, ER stress and cellular respiratory pathways were also modulated. Interestingly, these alterations reflected functional changes highly suggestive of decreased cellular lipotoxicity potential; reduced ER proteasome degradation of proteins and induction of chaperones; and a shift in cell energy homeostasis towards mitochondrial beta-oxidation. CONCLUSION Six-month treatment with omega-3 PUFAs significantly improved hepatic proteomic and plasma lipidomic markers of lipogenesis, endoplasmic reticulum stress and mitochondrial functions in patients with NASH.
Collapse
Affiliation(s)
| | - Claudia P Oliveira
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| | - José Tadeu Stefano
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Monize Aydar Nogueira
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | | | - Fernanda Bertucce Cordeiro
- Human Reproduction Section, Division of Urology, Department of Surgery, Sao Paulo Federal University, São Paulo, Brazil
| | | | - Raquel Susana Torrinhas
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Flair José Carrilho
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Puneet Puri
- Virginia Commonwealth University-VCU, Richmond, VA, USA
| | - Dan L Waitzberg
- Department of Gastroenterology (LIM-07/LIM-35), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| |
Collapse
|
33
|
Daray FM, Mann JJ, Sublette ME. How lipids may affect risk for suicidal behavior. J Psychiatr Res 2018; 104:16-23. [PMID: 29920417 PMCID: PMC6102068 DOI: 10.1016/j.jpsychires.2018.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/30/2018] [Accepted: 06/08/2018] [Indexed: 01/06/2023]
Abstract
Suicide and nonfatal suicidal behaviors are major causes of mortality and morbidity worldwide. Variability in rates of suicide and suicidal behaviors within and between countries has been attributed to population and individual risk factors, including economic status and cultural differences, both of which can have suicide risk effects mediated through a variety of factors, of which perhaps the least understood is the role of diet. We therefore review the scientific literature concerning two major dietary lipid classes, cholesterol and polyunsaturated fatty acids (PUFAs), that have been associated with higher risk of suicide attempts and suicide. We consider potential mechanistic intermediates including serotonin transporters and receptors, toll-like receptors (TLRs), nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and peroxisome proliferator activated receptors (PPARs). Based on this review, we describe a theoretical model linking cholesterol and PUFA status to suicide risk, taking into account the effects of cholesterol-lowering interventions on PUFA balance, membrane lipid microdomains (rafts) as a nexus of interaction between cholesterol and omega-3 PUFAs, and downstream effects on serotonergic neurotransmission and specific inflammatory pathways.
Collapse
Affiliation(s)
- Federico M. Daray
- Institute of Pharmacology. School of Medicine. University of Buenos Aires, Paraguay 2155, piso 9, Ciudad Autónoma de Buenos Aires, Argentina, C1121ABG
| | - J. John Mann
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032,Division of Molecular Imaging & Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 42, New York, NY 10032,Department of Radiology, Columbia University, 622 West 168th St, New York, NY 10032
| | - M. Elizabeth Sublette
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032,Division of Molecular Imaging & Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 42, New York, NY 10032,To whom correspondence should be addressed: New York State Psychiatric Institute, 1051 Riverside Drive, Unit 42, New York, NY 10032, Tel: 646 774-7514, Fax: 646 774-7589,
| |
Collapse
|
34
|
Fuentes NR, Mlih M, Barhoumi R, Fan YY, Hardin P, Steele TJ, Behmer S, Prior IA, Karpac J, Chapkin RS. Long-Chain n-3 Fatty Acids Attenuate Oncogenic KRas-Driven Proliferation by Altering Plasma Membrane Nanoscale Proteolipid Composition. Cancer Res 2018; 78:3899-3912. [PMID: 29769200 DOI: 10.1158/0008-5472.can-18-0324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/26/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
Ras signaling originates from transient nanoscale compartmentalized regions of the plasma membrane composed of specific proteins and lipids. The highly specific lipid composition of these nanodomains, termed nanoclusters, facilitates effector recruitment and therefore influences signal transduction. This suggests that Ras nanocluster proteolipid composition could represent a novel target for future chemoprevention interventions. There is evidence that consumption of fish oil containing long-chain n-3 polyunsaturated fatty acids (n-3 PUFA) such as eicosapentaenoic acid (EPA, 20:5Δ5,8,11,14,17) and docosahexaenoic acid (DHA, 22:6Δ4,7,10,13,16,19) may reduce colon cancer risk in humans, yet the mechanism underlying this effect is unknown. Here, we demonstrate that dietary n-3 PUFA reduce the lateral segregation of cholesterol-dependent and -independent nanoclusters, suppressing phosphatidic acid-dependent oncogenic KRas effector interactions, via their physical incorporation into plasma membrane phospholipids. This results in attenuation of oncogenic Ras-driven colonic hyperproliferation in both Drosophila and murine models. These findings demonstrate the unique properties of dietary n-3 PUFA in the shaping of Ras nanoscale proteolipid complexes and support the emerging role of plasma membrane-targeted therapies.Significance: The influence of dietary long chain n-3 polyunsaturated fatty acids on plasma membrane protein nanoscale organization and KRas signaling supports development of plasma membrane-targeted therapies in colon cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3899/F1.large.jpg Cancer Res; 78(14); 3899-912. ©2018 AACR.
Collapse
Affiliation(s)
- Natividad R Fuentes
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas.,Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Mohamed Mlih
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Sciences Center, College Station, Texas
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Yang-Yi Fan
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas
| | - Paul Hardin
- Department of Biology, Texas A&M University, College Station, Texas
| | - Trevor J Steele
- Department of Entomology, Texas A&M University, College Station, Texas
| | - Spencer Behmer
- Department of Entomology, Texas A&M University, College Station, Texas
| | - Ian A Prior
- Division of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Sciences Center, College Station, Texas
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas. .,Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| |
Collapse
|
35
|
Influence of omega-3 polyunsaturated fatty acids from fish oil or meal on the structure of lipid microdomains in bovine luteal cells. Anim Reprod Sci 2018; 193:40-57. [PMID: 29673917 DOI: 10.1016/j.anireprosci.2018.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 03/08/2018] [Accepted: 03/28/2018] [Indexed: 11/23/2022]
Abstract
Biological membranes are composed of a lipid bilayer and proteins that form lipid microdomains. This study examined the effects of fish byproducts on lipid-protein interactions within lipid microdomains of bovine luteal cells. In Exp. 1 and 2, luteal cells were prepared from corpora lutea (CL; n = 4 to 8) collected at an abattoir. Exp. 1 was conducted to optimize ultrasonication in a detergent-free protocol for isolation of lipid microdomains. A power setting of 10 to 20% was effective in isolating lipid microdomains from bulk lipid. In Exp. 2, cells were cultured in control medium or fish oil to determine influence of fish oil on distribution of lipid microdomain markers and prostaglandin F2α (FP) receptors. Cells treated with fish oil had a smaller percentage of microdomain markers and FP receptor in microdomains (P < 0.05). In Exp. 3 and 4, cells were prepared from mid-cycle CL obtained from cows supplemented with corn gluten meal (n = 4) or fish meal (n = 4). Exp. 3 examined effects of dietary supplementation on distribution of lipid microdomain markers and FP receptor and Exp. 4 on fatty acid composition within lipid microdomains. A smaller percentage of lipid microdomain markers and FP receptor was detected in microdomains of cells collected from fish meal supplemented animals (P < 0.05). In Exp. 4, a greater percentage of omega-3 polyunsaturated fatty acids was detected in bulk lipid from fish meal supplemented cows (P < 0.05). Results show that fish byproducts influence lipid-protein interactions in lipid microdomains in bovine luteal cells.
Collapse
|
36
|
Fuentes NR, Kim E, Fan YY, Chapkin RS. Omega-3 fatty acids, membrane remodeling and cancer prevention. Mol Aspects Med 2018; 64:79-91. [PMID: 29627343 DOI: 10.1016/j.mam.2018.04.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 03/27/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022]
Abstract
Proteins are often credited as the macromolecule responsible for performing critical cellular functions, however lipids have recently garnered more attention as our understanding of their role in cell function and human health becomes more apparent. Although cellular membranes are the lipid environment in which many proteins function, it is now apparent that protein and lipid assemblies can be organized to form distinct micro- or nanodomains that facilitate signaling events. Indeed, it is now appreciated that cellular function is partly regulated by the specific spatiotemporal lipid composition of the membrane, down to the nanosecond and nanometer scale. Furthermore, membrane composition is altered during human disease processes such as cancer and obesity. For example, an increased rate of lipid/cholesterol synthesis in cancerous tissues has long been recognized as an important aspect of the rewired metabolism of transformed cells. However, the contribution of lipids/cholesterol to cellular function in disease models is not yet fully understood. Furthermore, an important consideration in regard to human health is that diet is a major modulator of cell membrane composition. This can occur directly through incorporation of membrane substrates, such as fatty acids, e.g., n-3 polyunsaturated fatty acids (n-3 PUFA) and cholesterol. In this review, we describe scenarios in which changes in membrane composition impact human health. Particular focus is placed on the importance of intrinsic lipid/cholesterol biosynthesis and metabolism and extrinsic dietary modification in cancer and its effect on plasma membrane properties.
Collapse
Affiliation(s)
- Natividad R Fuentes
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Faculty of Toxicology, Texas A&M University, USA
| | - Eunjoo Kim
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Department of Molecular and Cellular Medicine, Texas A&M University, USA
| | - Yang-Yi Fan
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Department of Nutrition & Food Science, Texas A&M University, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Faculty of Toxicology, Texas A&M University, USA; Department of Nutrition & Food Science, Texas A&M University, USA; Center for Translational Environmental Health Research, Texas A&M University, USA.
| |
Collapse
|
37
|
Singh DP, Kaur G, Bagam P, Pinkston R, Batra S. Membrane microdomains regulate NLRP10- and NLRP12-dependent signalling in A549 cells challenged with cigarette smoke extract. Arch Toxicol 2018; 92:1767-1783. [PMID: 29623357 DOI: 10.1007/s00204-018-2185-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 03/13/2018] [Indexed: 12/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is predicted to become the third leading cause of death and disability worldwide by 2030; with cigarette smoking (active or passive) being one of the chief cause of its occurrence. Cigarette smoke exposure has been found to result in excessive inflammation and tissue injury, which might lead to COPD, although the exact pathophysiology of the disease remains elusive. While previous studies have demonstrated the role of membrane-bound Toll-like receptors (TLRs) in cigarette smoke (CS)-induced inflammation, scant information is available about the role of cytosolic NOD-like receptors (NLRs) in regulating CS-mediated inflammatory responses. Thus, we investigated the role of NLRP10 and NLRP12 in regulating inflammatory responses in human alveolar type II epithelial cells (A549) and human monocytic cells (THP-1) in response to a challenge with cigarette smoke extract (CSE). We observed CSE-mediated increase in caspase-1 activity; production of IL-1β and IL-18; and expression of NLRP10 and NLRP12 in A549 and THP-1 cells. Interestingly, immunofluorescence imaging results demonstrated an increase in the membrane recruitment of NLRP10 and NLRP12 proteins in CSE-challenged A549 cells. We also observed an increase in the expression of lipid raft proteins (caveolin-1, caveolin-2, and flotillin-1) and an induction of lipid raft assembly following CSE-exposure in A549 cells. Lipid rafts are cholesterol-rich membrane microdomains well known to act as harbours for signalling molecules. Here we demonstrate the recruitment of NLRP10 and NLRP12 in lipid raft entities as well as the interaction of NLRP12 with the lipid raft protein caveolin-1 in CSE-challenged A549 cells. Furthermore, enrichment of lipid raft entities with poly-unsaturated fatty acids (PUFA) rescued A549 cells from CSE-mediated membrane recruitment of NLRP10 and NLRP12, and also from inflammatory responses and inflammasome activation. Enrichment of membrane microdomains with PUFA was able to reverse filipin (chemical agent used for disrupting lipid rafts)-mediated enhanced inflammation in CSE-challenged A549 cells. Overall, our findings unveil a novel mechanism by identifying an important role of membrane microdomains (lipid rafts) in regulating CSE-induced inflammation and NLRP10/NLRP12-dependent signalling in A549 cells.
Collapse
Affiliation(s)
- Dhirendra P Singh
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Gagandeep Kaur
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Rakeysha Pinkston
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA, 70813, USA. .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
38
|
Protective Effects of ω-3 PUFA in Anthracycline-Induced Cardiotoxicity: A Critical Review. Int J Mol Sci 2017; 18:ijms18122689. [PMID: 29231904 PMCID: PMC5751291 DOI: 10.3390/ijms18122689] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/29/2017] [Accepted: 12/08/2017] [Indexed: 12/31/2022] Open
Abstract
It has been demonstrated that ω-3 polyunsaturated fatty acids (ω-3 PUFA) may exert a beneficial role as adjuvants in the prevention and treatment of many disorders, including cardiovascular diseases and cancer. Particularly, several in vitro and in vivo preclinical studies have shown the antitumor activity of ω-3 PUFA in different kinds of cancers, and several human studies have shown that ω-3 PUFA are able to decrease the risk of a series of cardiovascular diseases. Several mechanisms have been proposed to explain their pleiotropic beneficial effects. ω-3 PUFA have also been shown to prevent harmful side-effects (including cardiotoxicity and heart failure) induced by conventional and innovative anti-cancer drugs in both animals and patients. The available literature regarding the possible protective effects of ω-3 PUFA against anthracycline-induced cardiotoxicity, as well as the mechanisms involved, will be critically discussed herein. The study will analyze the critical role of different levels of ω-3 PUFA intake in determining the results of the combinatory studies with anthracyclines. Suggestions for future research will also be considered.
Collapse
|
39
|
Bagam P, Singh DP, Inda ME, Batra S. Unraveling the role of membrane microdomains during microbial infections. Cell Biol Toxicol 2017; 33:429-455. [PMID: 28275881 PMCID: PMC7088210 DOI: 10.1007/s10565-017-9386-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023]
Abstract
Infectious diseases pose major socioeconomic and health-related threats to millions of people across the globe. Strategies to combat infectious diseases derive from our understanding of the complex interactions between the host and specific bacterial, viral, and fungal pathogens. Lipid rafts are membrane microdomains that play important role in life cycle of microbes. Interaction of microbial pathogens with host membrane rafts influences not only their initial colonization but also their spread and the induction of inflammation. Therefore, intervention strategies aimed at modulating the assembly of membrane rafts and/or regulating raft-directed signaling pathways are attractive approaches for the. management of infectious diseases. The current review discusses the latest advances in terms of techniques used to study the role of membrane microdomains in various pathological conditions and provides updated information regarding the role of membrane rafts during bacterial, viral and fungal infections.
Collapse
Affiliation(s)
- Prathyusha Bagam
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha, Rosario, Argentina
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
40
|
Plewes MR, Burns PD, Hyslop RM, George Barisas B. Influence of omega-3 fatty acids on bovine luteal cell plasma membrane dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2413-2419. [PMID: 28912100 DOI: 10.1016/j.bbamem.2017.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/09/2017] [Accepted: 09/10/2017] [Indexed: 01/07/2023]
Abstract
Fish oil is a rich source of omega-3 fatty acids which disrupt lipid microdomain structure and affect mobility of the prostaglandin F2α (FP) receptor in bovine luteal cells. The objectives of this study were to determine the effects of individual omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on 1) membrane fatty acid composition, 2) lipid microdomain structure, and 3) lateral mobility of the FP receptor in bovine luteal cells. Ovaries were collected from a local abattoir (n=5/experiment). The corpus luteum was resected and enzymatically digested using collagenase to generate a mixed luteal cell population. In all experiments, luteal cells were treated with 0, 1, 10 or 100μM EPA or DHA for 72h to allow incorporation of fatty acids into membrane lipids. Results from experiment 1 show that culturing luteal cells in the presence of EPA or DHA increased these luteal fatty acids. In experiment 2, both EPA and DHA increased spatial distribution of lipid microdomains in a dose-dependent manner. Single particle tracking results from experiment 3 show that increasing both EPA and DHA concentrations increased micro- and macro-diffusion coefficients, increased domain size, and decreased residence time of FP receptors. Collectively, results from this study demonstrate similar effects of EPA and DHA on lipid microdomain structure and lateral mobility of FP receptors in cultured bovine luteal cells. Moreover, only 10μM of either fatty acid was needed to mimic the effects of fish oil.
Collapse
Affiliation(s)
- Michele R Plewes
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, United States
| | - Patrick D Burns
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, United States.
| | - Richard M Hyslop
- Department of Chemistry and Biochemistry, University of Northern Colorado, Greeley, CO 80639, United States
| | - B George Barisas
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
41
|
Fuentes NR, Salinas ML, Kim E, Chapkin RS. Emerging role of chemoprotective agents in the dynamic shaping of plasma membrane organization. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2017; 1859:1668-1678. [PMID: 28342710 PMCID: PMC5501766 DOI: 10.1016/j.bbamem.2017.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/15/2017] [Accepted: 03/19/2017] [Indexed: 12/22/2022]
Abstract
In the context of an organism, epithelial cells by nature are designed to be the defining barrier between self and the outside world. This is especially true for the epithelial cells that form the lining of the digestive tract, which absorb nutrients and serve as a barrier against harmful substances. These cells are constantly bathed by a complex mixture of endogenous (bile acids, mucus, microbial metabolites) and exogenous (food, nutrients, drugs) bioactive compounds. From a cell biology perspective, this type of exposure would directly impact the plasma membrane, which consists of a myriad of complex lipids and proteins. The plasma membrane not only functions as a barrier but also as the medium in which cellular signaling complexes form and function. This property is mediated by the organization of the plasma membrane, which is exquisitely temporally (nanoseconds to minutes) and spatially (nanometers to micrometers) regulated. Since numerous bioactive compounds found in the intestinal lumen can directly interact with lipid membranes, we hypothesize that the dynamic reshaping of plasma membrane organization underlies the chemoprotective effect of select membrane targeted dietary bioactives (MTDBs). This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Natividad R Fuentes
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Faculty of Toxicology, Texas A&M University, USA
| | - Michael L Salinas
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Department of Nutrition & Food Science, Texas A&M University, USA
| | - Eunjoo Kim
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Department of Molecular and Cellular Medicine, Texas A&M University, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, USA; Faculty of Toxicology, Texas A&M University, USA; Department of Nutrition & Food Science, Texas A&M University, USA; Center for Translational Environmental Health Research, Texas A&M University, USA.
| |
Collapse
|
42
|
Yang B, Ren XL, Huang H, Guo XJ, Ma AG, Li D. Circulating long-chain n-3 polyunsaturated fatty acid and incidence of stroke: a meta-analysis of prospective cohort studies. Oncotarget 2017; 8:83781-83791. [PMID: 29137382 PMCID: PMC5663554 DOI: 10.18632/oncotarget.19530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/12/2017] [Indexed: 01/29/2023] Open
Abstract
Background Circulating long-chain (LC) n-3 polyunsaturated fatty acid (PUFA) can provide objective measures that reflect both dietary consumption and relevant biological processes. Nevertheless, prospective cohort studies on circulating LC n-3 PUFA in relation to incidence of stroke have yielded inconsistent results. We therefore conducted a meta-analysis to quantitatively evaluate the association. Results A total of 2,836 stroke events occurred among 20,460 individuals aged 35–79 yr from 10 prospective cohort studies. Circulating LC n-3 PUFA was significantly associated with reduced risk of stroke (RR: 0.86; 95% CI: 0.76, 0.98; I2 = 0.00%), especially 22:5n-3 (RR: 0.74; 95% CI: 0.60, 0.92) and 22:6n-3 (RR: 0.78; 95% CI: 0.65, 0.94). The associations were more pronounced with ischemic stroke (RR: 0.81; 95% CI: 0.68, 0.96), but not with hemorrhagic stroke (RR: 0.95; 95% CI: 0.60, 1.49). A 1% increment of 22:5n-3 and 22:6n-3 proportions in circulating blood was associated with 25% (RR: 0.75; 95% CI: 0.64, 0.87) and 11% (RR: 0.89; 95% CI: 0.83, 0.95) reduced risk of stroke, respectively. Materials and Methods Pertinent studies were identified from Cochrane Library, PubMed and EMBASE database through June 2017. Multivariate-adjusted risk ratios (RRs) with 95% confidence interval (CI) for incident stroke when comparing the top with the bottom tertiles of baseline LC n-3 PUFA proportions in blood were pooled using a random-effect model. Conclusions Circulating LC n-3 PUFAs were linearly associated with reduced risk of stroke, especially 22:5n-3 and 22:6n-3. Such findings highlight the importance of circulating LC n-3 PUFA in the development of ischemic stroke.
Collapse
Affiliation(s)
- Bo Yang
- School of Public Health, Qingdao University, Qingdao, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Xiao-Li Ren
- The Laboratory of Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Hong Huang
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Xiao-Juan Guo
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Ai-Guo Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Duo Li
- School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Díaz-Coránguez M, Ramos C, Antonetti DA. The inner blood-retinal barrier: Cellular basis and development. Vision Res 2017; 139:123-137. [PMID: 28619516 DOI: 10.1016/j.visres.2017.05.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 01/01/2023]
Abstract
The blood-retinal barrier (BRB) regulates transport across retinal capillaries maintaining proper neural homeostasis and protecting the neural tissue from potential blood borne toxicity. Loss of the BRB contributes to the pathophysiology of a number of blinding retinal diseases including diabetic retinopathy. In this review, we address the basis of the BRB, including the molecular mechanisms that regulate flux across the retinal vascular bed. The routes of transcellular and paracellular flux are described as well as alterations in these pathways in response to permeabilizing agents in diabetes. Finally, we provide information on exciting new studies that help to elucidate the process of BRB development or barriergenesis and how understanding this process may lead to new opportunities for barrier restoration in diabetic retinopathy.
Collapse
Affiliation(s)
- Mónica Díaz-Coránguez
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - Carla Ramos
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
44
|
Blood-Brain Barrier Permeability Is Regulated by Lipid Transport-Dependent Suppression of Caveolae-Mediated Transcytosis. Neuron 2017; 94:581-594.e5. [PMID: 28416077 DOI: 10.1016/j.neuron.2017.03.043] [Citation(s) in RCA: 395] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 03/02/2017] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) provides a constant homeostatic brain environment that is essential for proper neural function. An unusually low rate of vesicular transport (transcytosis) has been identified as one of the two unique properties of CNS endothelial cells, relative to peripheral endothelial cells, that maintain the restrictive quality of the BBB. However, it is not known how this low rate of transcytosis is achieved. Here we provide a mechanism whereby the regulation of CNS endothelial cell lipid composition specifically inhibits the caveolae-mediated transcytotic route readily used in the periphery. An unbiased lipidomic analysis reveals significant differences in endothelial cell lipid signatures from the CNS and periphery, which underlie a suppression of caveolae vesicle formation and trafficking in brain endothelial cells. Furthermore, lipids transported by Mfsd2a establish a unique lipid environment that inhibits caveolae vesicle formation in CNS endothelial cells to suppress transcytosis and ensure BBB integrity.
Collapse
|
45
|
Plewes MR, Burns PD, Graham PE, Hyslop RM, Barisas BG. Effect of fish oil on lateral mobility of prostaglandin F 2α (FP) receptors and spatial distribution of lipid microdomains in bovine luteal cell plasma membrane in vitro. Domest Anim Endocrinol 2017; 58:39-52. [PMID: 27643975 PMCID: PMC5135567 DOI: 10.1016/j.domaniend.2016.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/29/2016] [Accepted: 08/03/2016] [Indexed: 11/16/2022]
Abstract
Lipid microdomains are ordered regions on the plasma membrane of cells, rich in cholesterol and sphingolipids, ranging in size from 10 to 200 nm in diameter. These lipid-ordered domains may serve as platforms to facilitate colocalization of intracellular signaling proteins during agonist-induced signal transduction. It is hypothesized that fish oil will disrupt the lipid microdomains, increasing spatial distribution of these lipid-ordered domains and lateral mobility of the prostaglandin (PG) F2α (FP) receptors in bovine luteal cells. The objectives of this study were to examine the effects of fish oil on (1) the spatial distribution of lipid microdomains, (2) lateral mobility of FP receptors, and (3) lateral mobility of FP receptors in the presence of PGF2α on the plasma membrane of bovine luteal cells in vitro. Bovine ovaries were obtained from a local abattoir and corpora lutea were digested using collagenase. In experiment 1, lipid microdomains were labeled using cholera toxin subunit B Alexa Fluor 555. Domains were detected as distinct patches on the plasma membrane of mixed luteal cells. Fish oil treatment decreased fluorescent intensity in a dose-dependent manner (P < 0.01). In experiment 2, single particle tracking was used to examine the effects of fish oil treatment on lateral mobility of FP receptors. Fish oil treatment increased microdiffusion and macrodiffusion coefficients of FP receptors as compared to control cells (P < 0.05). In addition, compartment diameters of domains were larger, and residence times were reduced for receptors in fish oil-treated cells (P < 0.05). In experiment 3, single particle tracking was used to determine the effects of PGF2α on lateral mobility of FP receptors and influence of fish oil treatment. Lateral mobility of receptors was decreased within 5 min following the addition of ligand for control cells (P < 0.05). However, lateral mobility of receptors was unaffected by addition of ligand for fish oil-treated cells (P > 0.10). The data presented provide strong evidence that fish oil causes a disruption in lipid microdomains and affects lateral mobility of FP receptors in the absence and presence of PGF2α.
Collapse
Affiliation(s)
- M R Plewes
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, 80639
| | - P D Burns
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, 80639.
| | - P E Graham
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, 80639
| | - R M Hyslop
- Department of Chemistry and Biochemistry, University of Northern Colorado, Greeley, Colorado, 80639
| | - B G Barisas
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
46
|
Alaarg A, Jordan NY, Verhoef JJ, Metselaar JM, Storm G, Kok RJ. Docosahexaenoic acid liposomes for targeting chronic inflammatory diseases and cancer: an in vitro assessment. Int J Nanomedicine 2016; 11:5027-5040. [PMID: 27785012 PMCID: PMC5063558 DOI: 10.2147/ijn.s115995] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammation, oxidative stress, and uncontrolled cell proliferation are common key features of chronic inflammatory diseases, such as atherosclerosis and cancer. ω3 polyunsaturated fatty acids (PUFAs; also known as omega3 fatty acids or fish oil) have beneficial effects against inflammation upon dietary consumption. However, these effects cannot be fully exploited unless diets are enriched with high concentrations of fish oil supplements over long periods of time. Here, a nanomedicine-based approach is presented for delivering effective levels of PUFAs to inflammatory cells. Nanoparticles are internalized by immune cells, and hence can adequately deliver bioactive lipids into these target cells. The ω3 FA docosahexaenoic acid was formulated into liposomes (ω-liposomes), and evaluated for anti-inflammatory effects in different types of immune cells. ω-Liposomes strongly inhibited the release of reactive oxygen species and reactive nitrogen species from human neutrophils and murine macrophages, and also inhibited the production of the proinflammatory cytokines TNFα and MCP1. Moreover, ω-liposomes inhibited tumor-cell proliferation when evaluated in FaDu head and neck squamous carcinoma and 4T1 breast cancer cells in in vitro cultures. We propose that ω-liposomes are a promising nanonutraceutical formulation for intravenous delivery of fish oil FAs, which may be beneficial in the treatment of inflammatory disorders and cancer.
Collapse
Affiliation(s)
- Amr Alaarg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht; Department of Biomaterials Science and Technology, Institute for Biomedical Technology and Technical Medicine (MIRA), University of Twente, Enschede, the Netherlands
| | - Nan Yeun Jordan
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| | - Johan Jf Verhoef
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, Institute for Biomedical Technology and Technical Medicine (MIRA), University of Twente, Enschede, the Netherlands; Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht; Department of Biomaterials Science and Technology, Institute for Biomedical Technology and Technical Medicine (MIRA), University of Twente, Enschede, the Netherlands
| | - Robbert J Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| |
Collapse
|
47
|
See Hoe LE, May LT, Headrick JP, Peart JN. Sarcolemmal dependence of cardiac protection and stress-resistance: roles in aged or diseased hearts. Br J Pharmacol 2016; 173:2966-91. [PMID: 27439627 DOI: 10.1111/bph.13552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022] Open
Abstract
Disruption of the sarcolemmal membrane is a defining feature of oncotic death in cardiac ischaemia-reperfusion (I-R), and its molecular makeup not only fundamentally governs this process but also affects multiple determinants of both myocardial I-R injury and responsiveness to cardioprotective stimuli. Beyond the influences of membrane lipids on the cytoprotective (and death) receptors intimately embedded within this bilayer, myocardial ionic homeostasis, substrate metabolism, intercellular communication and electrical conduction are all sensitive to sarcolemmal makeup, and critical to outcomes from I-R. As will be outlined in this review, these crucial sarcolemmal dependencies may underlie not only the negative effects of age and common co-morbidities on myocardial ischaemic tolerance but also the on-going challenge of implementing efficacious cardioprotection in patients suffering accidental or surgically induced I-R. We review evidence for the involvement of sarcolemmal makeup changes in the impairment of stress-resistance and cardioprotection observed with ageing and highly prevalent co-morbid conditions including diabetes and hypercholesterolaemia. A greater understanding of membrane changes with age/disease, and the inter-dependences of ischaemic tolerance and cardioprotection on sarcolemmal makeup, can facilitate the development of strategies to preserve membrane integrity and cell viability, and advance the challenging goal of implementing efficacious 'cardioprotection' in clinically relevant patient cohorts. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Queensland, Australia
| | - Lauren T May
- Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, VIC, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|
48
|
Long chain n-3 polyunsaturated fatty acids increase the efficacy of docetaxel in mammary cancer cells by downregulating Akt and PKCε/δ-induced ERK pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:380-90. [DOI: 10.1016/j.bbalip.2016.01.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/15/2016] [Accepted: 01/22/2016] [Indexed: 12/21/2022]
|
49
|
Ciesielska A, Kwiatkowska K. Modification of pro-inflammatory signaling by dietary components: The plasma membrane as a target. Bioessays 2015; 37:789-801. [PMID: 25966354 DOI: 10.1002/bies.201500017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
You are what you eat - this well-known phrase properly describes the phenomenon of the effects of diet on acute and chronic inflammation. Several lipids and lipophilic compounds that are delivered with food or are produced in situ in pathological conditions exert immunomodulatory activity due to their interactions with the plasma membrane. This group of compounds includes cholesterol and its oxidized derivatives, fatty acids, α-tocopherol, and polyphenols. Despite their structural heterogeneity, all these compounds ultimately induce changes in plasma membrane architecture and fluidity. By doing this, they modulate the dynamics of plasma membrane receptors, such as TLR4. This receptor is activated by lipopolysaccharide, triggering acute inflammation during bacterial infection, which often leads to sepsis and is linked with diverse chronic inflammatory diseases. In this review, we discuss how the impact on plasma membrane properties contributes to the immunomodulatory activity of dietary compounds, pointing to the therapeutic potential of some of them. Also watch the Video Abstract.
Collapse
Affiliation(s)
- Anna Ciesielska
- Nencki Institute of Experimental Biology, Laboratory of Molecular Membrane Biology, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Nencki Institute of Experimental Biology, Laboratory of Molecular Membrane Biology, Warsaw, Poland
| |
Collapse
|
50
|
Omega-3 polyunsaturated fatty acids as an angelus custos to rescue patients from NSAID-induced gastroduodenal damage. J Gastroenterol 2015; 50:614-25. [PMID: 25578017 DOI: 10.1007/s00535-014-1034-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/17/2014] [Indexed: 02/04/2023]
Abstract
Nonsteroidal anti-inflammat ory drugs (NSAIDs) are one of the drug types frequently prescribed for their analgesic, anti-inflammatory, and antithrombotic actions, but carry a risk of major gastroduodenal damage from mild erosive changes to serious ulceration leading to fatal outcomes. From the long history of willow tree bark and its extracts being applied for the relief of pain and fever, the synthesis of acetylsalicylic acid, the development of selective cyclooxygenase 2 inhibitors (coxibs), and the identification of a G-protein-coupled receptor for prostaglandin, the popular combination regimen of an NSAID and a proton pump inhibitor was invented, but development was continued for further improvement. With regard to major NSAID adverse effects, gastrointestinal (GI) and cardiovascular (CV) risks still remained as problems to be solved. In this review, it is shown that n-3 polyunsaturated fatty acid (PUFA) based NSAIDs can be an angelus custos, supported with facts that an intake of essential n-3 PUFAs orchestrates concerted protective actions against two notorious side effects of NSAIDs, the aforementioned GI risk and CV risk of NSAIDs. Since pills containing n-3 PUFAs, omega-3-acid ethyl ester capsules (Lovaza, Omarcor), have already been safely prescribed to prevent atherosclerosis through lessening lipid burdening, the introduction of a drug delivery system such as a gastroretentive form of n-3 PUFA based NSAIDs will highlight newer hope for GI safety under the guarantee of reduced CV risk. Because n-3 PUFAs have been proven to attenuate cytotoxicity, inhibit lipid-raft-associated harmful signaling, and relieve oxidative stress relevant to NSAIDs, n-3 PUFA based NSAIDs will be next-generation GI-safe NSAIDs.
Collapse
|