1
|
Taskinen JH, Holopainen M, Ruhanen H, van der Stoel M, Käkelä R, Ikonen E, Keskitalo S, Varjosalo M, Olkkonen VM. Functional omics of ORP7 in primary endothelial cells. BMC Biol 2024; 22:292. [PMID: 39695567 DOI: 10.1186/s12915-024-02087-6] [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: 03/20/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Many members of the oxysterol-binding protein-related protein (ORP) family have been characterized in detail over the past decades, but the lipid transport and other functions of ORP7 still remain elusive. What is known about ORP7 points toward an endoplasmic reticulum and plasma membrane-localized protein, which also interacts with GABA type A receptor-associated protein like 2 (GABARAPL2) and unlipidated Microtubule-associated proteins 1A/1B light chain 3B (LC3B), suggesting a further autophagosomal/lysosomal association. Functional roles of ORP7 have been suggested in cholesterol efflux, hypercholesterolemia, and macroautophagy. We performed a hypothesis-free multi-omics analysis of chemical ORP7 inhibition utilizing transcriptomics and lipidomics as well as proximity biotinylation interactomics to characterize ORP7 functions in a primary cell type, human umbilical vein endothelial cells (HUVECs). Moreover, assays on angiogenesis, cholesterol efflux, and lipid droplet quantification were conducted. RESULTS Pharmacological inhibition of ORP7 leads to an increase in gene expression related to lipid metabolism and inflammation, while genes associated with cell cycle and cell division were downregulated. Lipidomic analysis revealed increases in ceramides and lysophosphatidylcholines as well as saturated and monounsaturated triacylglycerols. Significant decreases were seen in all cholesteryl ester and in some unsaturated triacylglycerol species, compatible with the detected decrease of mean lipid droplet area. Along with the reduced lipid stores, ATP-binding cassette subfamily G member 1 (ABCG1)-mediated cholesterol efflux and angiogenesis decreased. Interactomics revealed an interaction of ORP7 with AKT1, a central metabolic regulator. CONCLUSIONS The transcriptomics results suggest an increase in prostanoid as well as oxysterol synthesis, which could be related to the observed upregulation of proinflammatory genes. We envision that the defective angiogenesis in HUVECs subjected to ORP7 inhibition could be the result of an unfavorable plasma membrane lipid composition and/or reduced potential for cell division. To conclude, the present study suggests multifaceted functions of ORP7 in lipid homeostasis, angiogenic tube formation, and gene expression of lipid metabolism, inflammation, and cell cycle in primary endothelial cells.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Minna Holopainen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Miesje van der Stoel
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Elina Ikonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Salla Keskitalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
| | - Markku Varjosalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
2
|
Perus M, Courtaut F, Pais de Barros JP, Aires V, Hermetet F, Delmas D. VEGF-R2/CAV-1 Interaction Induced by Resveratrol/Eicosapentaenoic Acid/Docosahexaenoic Acid-Enriched Formulation through Functional Detergent-Resistant Membranes Is Associated with Decreased VEGF-A Release in ARPE-19 Cells. Mol Nutr Food Res 2024; 68:e2300893. [PMID: 38763919 DOI: 10.1002/mnfr.202300893] [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: 12/14/2023] [Revised: 02/15/2024] [Indexed: 05/21/2024]
Abstract
SCOPE Omega-3 fatty acids (O3FAs) and resveratrol (RSV) are known to be beneficial for certain eye diseases, such as age-related macular degeneration (AMD). Neovascular AMD is characterized by abnormal blood vessel formation due to the excessive synthesis of vascular endothelial growth factor (VEGF) by retinal pigment epithelium (RPE) cells. The study investigates whether a formulation based on eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and RSV is capable of counteracting VEGF-A secretion, and elucidates the molecular mechanism. METHODS AND RESULTS The study finds, using ELISA, that O3FAs/RSV reduces VEGF-A secretion in human RPE cells. This phenomenon is related to the increased interaction between VEGF-receptor 2 (VEGF-R2) and caveolin-1 (CAV-1), a protein of detergent-resistant membranes (DRMs), as determined by co-immunoprecipitation and proximity ligation assay. Using microscale thermophoresis, the study confirms that O3FAs/RSV causes a high-affinity interaction. Isolation and analysis of DRMs reveal that this interaction is concomitant with VEGF-R2 relocalization in DRMs. The depletion of DRMs by a cholesterol-chelating agent blocks the VEGF-R2/CAV-1 interaction and EPA/DHA/RSV-mediated impairment of VEGF production. CONCLUSION This specific interaction can provide a new strategy for countering VEGF-A production in human RPE cells and, consequently, reducing neovascularization in AMD. Further preclinical studies involving O3FAs and polyphenols are warranted.
Collapse
Affiliation(s)
- Maude Perus
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
| | - Flavie Courtaut
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
| | - Jean-Paul Pais de Barros
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
- INSERM UMS58 BioSanD - Diviomic Platform, Dijon, 21000, France
| | - Virginie Aires
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
| | - François Hermetet
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
| | - Dominique Delmas
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, 21000, France
- INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, Dijon, 21000, France
- INSERM UMS58 BioSanD - Diviomic Platform, Dijon, 21000, France
- Centre de Lutte Contre le Cancer Georges François Leclerc, Dijon, 21000, France
| |
Collapse
|
3
|
Fu Y, Zhang Z, Webster KA, Paulus YM. Treatment Strategies for Anti-VEGF Resistance in Neovascular Age-Related Macular Degeneration by Targeting Arteriolar Choroidal Neovascularization. Biomolecules 2024; 14:252. [PMID: 38540673 PMCID: PMC10968528 DOI: 10.3390/biom14030252] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 05/04/2024] Open
Abstract
Despite extensive use of intravitreal anti-vascular endothelial growth factor (anti-VEGF) biologics for over a decade, neovascular age-related macular degeneration (nAMD) or choroidal neovascularization (CNV) continues to be a major cause of irreversible vision loss in developed countries. Many nAMD patients demonstrate persistent disease activity or experience declining responses over time despite anti-VEGF treatment. The underlying mechanisms of anti-VEGF resistance are poorly understood, and no effective treatment strategies are available to date. Here we review evidence from animal models and clinical studies that supports the roles of neovascular remodeling and arteriolar CNV formation in anti-VEGF resistance. Cholesterol dysregulation, inflammation, and ensuing macrophage activation are critically involved in arteriolar CNV formation and anti-VEGF resistance. Combination therapy by neutralizing VEGF and enhancing cholesterol removal from macrophages is a promising strategy to combat anti-VEGF resistance in CNV.
Collapse
Affiliation(s)
- Yingbin Fu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhao Zhang
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
| | - Keith A. Webster
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yannis M. Paulus
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
4
|
Abstract
The formation of membrane vesicles is a common feature in all eukaryotes. Lipid rafts are the best-studied example of membrane domains for both eukaryotes and prokaryotes, and their existence also is suggested in Archaea membranes. Lipid rafts are involved in the formation of transport vesicles, endocytic vesicles, exocytic vesicles, synaptic vesicles and extracellular vesicles, as well as enveloped viruses. Two mechanisms of how rafts are involved in vesicle formation have been proposed: first, that raft proteins and/or lipids located in lipid rafts associate with coat proteins that form a budding vesicle, and second, vesicle budding is triggered by enzymatic generation of cone-shaped ceramides and inverted cone-shaped lyso-phospholipids. In both cases, induction of curvature is also facilitated by the relaxation of tension in the raft domain. In this Review, we discuss the role of raft-derived vesicles in several intracellular trafficking pathways. We also highlight their role in different pathways of endocytosis, and in the formation of intraluminal vesicles (ILVs) through budding inwards from the multivesicular body (MVB) membrane, because rafts inside MVB membranes are likely to be involved in loading RNA into ILVs. Finally, we discuss the association of glycoproteins with rafts via the glycocalyx.
Collapse
Affiliation(s)
- Karolina Sapoń
- Institute of Biology, University of Opole, Kominka 6, 45-032 Opole, Poland
| | - Rafał Mańka
- Institute of Biology, University of Opole, Kominka 6, 45-032 Opole, Poland
| | - Teresa Janas
- Institute of Biology, University of Opole, Kominka 6, 45-032 Opole, Poland
| | - Tadeusz Janas
- Institute of Biology, University of Opole, Kominka 6, 45-032 Opole, Poland
| |
Collapse
|
5
|
Chillà A, Anceschi C, Frediani E, Scavone F, Del Rosso T, Pelagio G, Tufaro A, De Palma G, Del Rosso M, Fibbi G, Chiarugi P, Laurenzana A, Margheri F. Inhibition of MMPs supports amoeboid angiogenesis hampering VEGF-targeted therapies via MLC and ERK 1/2 signaling. J Transl Med 2023; 21:102. [PMID: 36759828 PMCID: PMC9912547 DOI: 10.1186/s12967-023-03954-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND In the past decades studies on anti-tumoral drugs inhibiting matrix metalloproteinase (MMPs) were disappointing. Recently, we demonstrated that mature endothelial cells (ECs) and endothelial colony forming cells (ECFCs) can switch between invasion modes to cope with challenging environments, performing the "amoeboid angiogenesis" in the absence of proteases activity. METHODS We first set out to investigate by ELISA if the inhibitors of the main protease family involved in angiogenesis were differently expressed during breast cancer progression. We used Marimastat, a broad-spectrum MMP inhibitor, as a means of inducing amoeboid characteristics and studied VEGF role in amoeboid angiogenesis. Thus, we performed invasion and capillary morphogenesis assay, morphological, cell signaling and in vivo mouse studies. RESULTS Our data showed that TIMP1, TIMP2, alpha2-antiplasmin, PAI-1 and cystatin increase in breast cancer serum of patients with primary cancer and lymph node positive compared to healthy women. In vitro results revealed that the most high-powered protease inhibitors able to induce amoeboid invasion of ECFCs were TIMP1, 2 and 3. Surprisingly, Marimastat promotes ECFC invasion and tubular formation in vitro and in vivo, inducing amoeboid characteristics. We observed that the combination of Marimastat plus VEGF doesn't boost neither cell invasion nor vessel formation capacity. Moreover, inhibition of VEGF activity with Bevacizumab in the presence of Marimastat confirmed that amoeboid angiogenesis is independent from the stimulus of the main vascular growth factor, VEGF. CONCLUSIONS We underline the importance to consider the amoeboid mechanism of endothelial and cancer cell invasion, probably responsible for the failure of synthetic metalloproteinase inhibitors as cancer therapy and tumor resistance to VEGF-targeted therapies, to set-up new drugs to be used in cancer therapy.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy.
| | - Cecilia Anceschi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Elena Frediani
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Francesca Scavone
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Tommaso Del Rosso
- grid.4839.60000 0001 2323 852XDepartment of Physics, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, RJ 22451-900 Brazil
| | - Giuseppe Pelagio
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Antonio Tufaro
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Giuseppe De Palma
- IRCCS Istituto Tumori Giovanni Paolo II Bari, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Mario Del Rosso
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Gabriella Fibbi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Paola Chiarugi
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Anna Laurenzana
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| | - Francesca Margheri
- grid.8404.80000 0004 1757 2304Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni, 50, 50134 Florence, Italy
| |
Collapse
|
6
|
Fang J, Liu H, Qiao W, Xu T, Yang Y, Xie H, Lam CH, Yeung KWK, Zhao X. Biomimicking Leaf-Vein Engraved Soft and Elastic Membrane Promotes Vascular Reconstruction. Adv Healthc Mater 2023; 12:e2201220. [PMID: 36330558 DOI: 10.1002/adhm.202201220] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Hierarchical vasculature reconstruction is fundamental for tissue regeneration. The regeneration of functional vascular network requires a proper directional guidance, especially in case of large-size defects. To provide the "running track" for vasculature, a leaf-vein mimetic membrane using soft and elastic poly(lactide-co-propylene glycol-co-lactide) dimethacrylate is developed. Engraved with an interconnected and perfusable leaf-vein micropattern, the membrane can guide human umbilical vein endothelial cells (HUVECs) to form vasculature in vitro. In particular, the "running track" upregulates the angiogenesis-related gene expression and promotes the HUVECs to differentiate into tip cells and stalk cells via tuning vascular endothelial growth factor receptor 2 signaling transduction. As a proof of concept, its revascularization capability using a rat calvarial defect model in vivo is evaluated. The in vivo results demonstrate that the leaf-vein engraved membrane accelerates the formation and maturation of vasculature, leading to a hierarchical blood vessel network. With the superior pro-vasculature property, it is believed that the leaf-vein engraved membrane is not only an ideal candidate for the reconstruction of calvarial vasculature but also a promising solution for more complicated vasculature reconstruction, such as muscle, skin, and heart.
Collapse
Affiliation(s)
- Jinghan Fang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Huaqian Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Wei Qiao
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Tianpeng Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Yuhe Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Huizhi Xie
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chun-Hei Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Kelvin W K Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| |
Collapse
|
7
|
Zhang Y, Zhong Y, Liu W, Zheng F, Zhao Y, Zou L, Liu X. PFKFB3-mediated glycometabolism reprogramming modulates endothelial differentiation and angiogenic capacity of placenta-derived mesenchymal stem cells. Stem Cell Res Ther 2022; 13:391. [PMID: 35918720 PMCID: PMC9344722 DOI: 10.1186/s13287-022-03089-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have a great potential ability for endothelial differentiation, contributing to an effective means of therapeutic angiogenesis. Placenta-derived mesenchymal stem cells (PMSCs) have gradually attracted attention, while the endothelial differentiation has not been fully evaluated in PMSCs. Metabolism homeostasis plays an important role in stem cell differentiation, but less is known about the glycometabolic reprogramming during the PMSCs endothelial differentiation. Hence, it is critical to investigate the potential role of glycometabolism reprogramming in mediating PMSCs endothelial differentiation. METHODS Dil-Ac-LDL uptake assay, flow cytometry, and immunofluorescence were all to verify the endothelial differentiation in PMSCs. Seahorse XF Extracellular Flux Analyzers, Mito-tracker red staining, Mitochondrial membrane potential (MMP), lactate secretion assay, and transcriptome approach were to assess the variation of mitochondrial respiration and glycolysis during the PMSCs endothelial differentiation. Glycolysis enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) was considered a potential modulator for endothelial differentiation in PMSCs by small interfering RNA. Furthermore, transwell, in vitro Matrigel tube formation, and in vivo Matrigel plug assays were performed to evaluate the effect of PFKFB3-induced glycolysis on angiogenic capacities in this process. RESULTS PMSCs possessed the superior potential of endothelial differentiation, in which the glycometabolic preference for glycolysis was confirmed. Moreover, PFKFB3-induced glycometabolism reprogramming could modulate the endothelial differentiation and angiogenic abilities of PMSCs. CONCLUSIONS Our results revealed that PFKFB3-mediated glycolysis is important for endothelial differentiation and angiogenesis in PMSCs. Our understanding of cellular glycometabolism and its regulatory effects on endothelial differentiation may propose and improve PMSCs as a putative strategy for clinical therapeutic angiogenesis.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Weifang Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Fanghui Zheng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yin Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
8
|
Enterohemorrhagic Escherichia coli and a Fresh View on Shiga Toxin-Binding Glycosphingolipids of Primary Human Kidney and Colon Epithelial Cells and Their Toxin Susceptibility. Int J Mol Sci 2022; 23:ijms23136884. [PMID: 35805890 PMCID: PMC9266556 DOI: 10.3390/ijms23136884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are the human pathogenic subset of Shiga toxin (Stx)-producing E. coli (STEC). EHEC are responsible for severe colon infections associated with life-threatening extraintestinal complications such as the hemolytic-uremic syndrome (HUS) and neurological disturbances. Endothelial cells in various human organs are renowned targets of Stx, whereas the role of epithelial cells of colon and kidneys in the infection process has been and is still a matter of debate. This review shortly addresses the clinical impact of EHEC infections, novel aspects of vesicular package of Stx in the intestine and the blood stream as well as Stx-mediated extraintestinal complications and therapeutic options. Here follows a compilation of the Stx-binding glycosphingolipids (GSLs), globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) and their various lipoforms present in primary human kidney and colon epithelial cells and their distribution in lipid raft-analog membrane preparations. The last issues are the high and extremely low susceptibility of primary renal and colonic epithelial cells, respectively, suggesting a large resilience of the intestinal epithelium against the human-pathogenic Stx1a- and Stx2a-subtypes due to the low content of the high-affinity Stx-receptor Gb3Cer in colon epithelial cells. The review closes with a brief outlook on future challenges of Stx research.
Collapse
|
9
|
Kuo A, Checa A, Niaudet C, Jung B, Fu Z, Wheelock CE, Singh SA, Aikawa M, Smith LE, Proia RL, Hla T. Murine endothelial serine palmitoyltransferase 1 (SPTLC1) is required for vascular development and systemic sphingolipid homeostasis. eLife 2022; 11:78861. [PMID: 36197001 PMCID: PMC9578713 DOI: 10.7554/elife.78861] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/04/2022] [Indexed: 02/04/2023] Open
Abstract
Serine palmitoyl transferase (SPT), the rate-limiting enzyme in the de novo synthesis of sphingolipids (SL), is needed for embryonic development, physiological homeostasis, and response to stress. The functions of de novo SL synthesis in vascular endothelial cells (EC), which line the entire circulatory system, are not well understood. Here, we show that the de novo SL synthesis in EC not only regulates vascular development but also maintains circulatory and peripheral organ SL levels. Mice with an endothelial-specific gene knockout of SPTLC1 (Sptlc1 ECKO), an essential subunit of the SPT complex, exhibited reduced EC proliferation and tip/stalk cell differentiation, resulting in delayed retinal vascular development. In addition, Sptlc1 ECKO mice had reduced retinal neovascularization in the oxygen-induced retinopathy model. Mechanistic studies suggest that EC SL produced from the de novo pathway are needed for lipid raft formation and efficient VEGF signaling. Post-natal deletion of the EC Sptlc1 also showed rapid reduction of several SL metabolites in plasma, red blood cells, and peripheral organs (lung and liver) but not in the retina, part of the central nervous system (CNS). In the liver, EC de novo SL synthesis was important for acetaminophen-induced rapid ceramide elevation and hepatotoxicity. These results suggest that EC-derived SL metabolites are in constant flux between the vasculature, circulatory elements, and parenchymal cells of non-CNS organs. Taken together, our data point to the central role of the endothelial SL biosynthesis in maintaining vascular development, neovascular proliferation, non-CNS tissue metabolic homeostasis, and hepatocyte response to stress.
Collapse
Affiliation(s)
- Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical SchoolBostonUnited States
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska InstituteStockholmSweden
| | - Colin Niaudet
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical SchoolBostonUnited States
| | - Bongnam Jung
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical SchoolBostonUnited States
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska InstituteStockholmSweden,Department of Respiratory Medicine and Allergy, Karolinska University HospitalStockholmSweden,Gunma University Initiative for Advanced Research, Gunma UniversityMaebashiJapan
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Lois E Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Richard L Proia
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
10
|
Li B, Qin Y, Yu X, Xu X, Yu W. Lipid raft involvement in signal transduction in cancer cell survival, cell death and metastasis. Cell Prolif 2022; 55:e13167. [PMID: 34939255 PMCID: PMC8780926 DOI: 10.1111/cpr.13167] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
Lipid rafts are cholesterol- and sphingolipid-enriched specialized membrane domains within the plasma membrane. Lipid rafts regulate the density and activity of signal receptors by compartmentalizing them, promoting signalling cascades that play important roles in the survival, death and metastasis of cancer cells. In this review, we emphasize the current concept initially postulated by F. Mollinedo and C. Gajate on the importance of lipid rafts in cancer survival, death and metastasis by describing representative signalling pathways, including the IGF system and the PI3K/AKT, Fas/CD95, VEGF/VEGFR2 and CD44 signalling pathways, and we also discuss the concept of CASMER (cluster of apoptotic signalling molecule-enriched rafts), coined, originally introduced and further advanced by F. Mollinedo and C. Gajate in the period 2005-2010. Then, we summarize relevant research progress and suggest that lipid rafts play important roles in the survival, death and metastasis of cancer cells, making them promising targets for cancer therapy.
Collapse
Affiliation(s)
- Borui Li
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Yi Qin
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xiaowu Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Wenyan Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| |
Collapse
|
11
|
Mechanical Aspects of Angiogenesis. Cancers (Basel) 2021; 13:cancers13194987. [PMID: 34638470 PMCID: PMC8508205 DOI: 10.3390/cancers13194987] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The formation of new blood vessels from already existing ones is a process of high clinical relevance, since it is of great importance for both physiological and pathological processes. In regard to tumors, the process is crucial, since it ensures the supply with nutrients and the growth of the tumor. The influence of mechanical factors on this biological process is an emerging field. Until now, the shear force of the blood flow has been considered the main mechanical parameter during angiogenesis. This review article provides an overview of further mechanical cues, with particular focus on the surrounding extracellular matrix impacting the cell behavior and, thus, regulating angiogenesis. This underlines the enormous importance of the mechanical properties of the extracellular matrix on cell biological processes and shows how changing the mechanics of the extracellular matrix could be used as a possible therapeutic approach in cancer therapy. Abstract Angiogenesis is of high clinical relevance as it plays a crucial role in physiological (e.g., tissue regeneration) and pathological processes (e.g., tumor growth). Besides chemical signals, such as VEGF, the relationship between cells and the extracellular matrix (ECM) can influence endothelial cell behavior during angiogenesis. Previously, in terms of the connection between angiogenesis and mechanical factors, researchers have focused on shear forces due to blood flow. However, it is becoming increasingly important to include the direct influence of the ECM on biological processes, such as angiogenesis. In this context, we focus on the stiffness of the surrounding ECM and the adhesion of cells to the ECM. Furthermore, we highlight the mechanical cues during the main stages of angiogenesis: cell migration, tip and stalk cells, and vessel stabilization. It becomes clear that the different stages of angiogenesis require various chemical and mechanical cues to be modulated by/modulate the stiffness of the ECM. Thus, changes of the ECM during tumor growth represent additional potential dysregulations of angiogenesis in addition to erroneous biochemical signals. This awareness could be the basis of therapeutic approaches to counteract specific processes in tumor angiogenesis.
Collapse
|
12
|
Zabroski IO, Nugent MA. Lipid Raft Association Stabilizes VEGF Receptor 2 in Endothelial Cells. Int J Mol Sci 2021; 22:ijms22020798. [PMID: 33466887 PMCID: PMC7830256 DOI: 10.3390/ijms22020798] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The binding of vascular endothelial growth factor A (VEGF) to VEGF receptor-2 (VEGFR-2) stimulates angiogenic signaling. Lipid rafts are cholesterol-dense regions of the plasma membrane that serve as an organizational platform for biomolecules. Although VEGFR2 has been shown to colocalize with lipid rafts to regulate its activation, the effect of lipid rafts on non-activated VEGFR2 has not been explored. Here, we characterized the involvement of lipid rafts in modulating the stability of non-activated VEGFR2 in endothelial cells using raft disrupting agents: methyl-β-cyclodextrin, sphingomyelinase and simvastatin. Disrupting lipid rafts selectively decreased the levels of non-activated VEGFR2 as a result of increased lysosomal degradation. The decreased expression of VEGFR2 translated to reduced VEGF-activation of the extracellular signal-regulated protein kinases (ERK). Overall, our results indicate that lipid rafts stabilize VEGFR2 and its associated signal transduction activities required for angiogenesis. Thus, modulation of lipid rafts may provide a means to regulate the sensitivity of endothelial cells to VEGF stimulation. Indeed, the ability of simvastatin to down regulate VEGFR2 and inhibit VEGF activity suggest a potential mechanism underlying the observation that this drug improves outcomes in the treatment of certain cancers.
Collapse
|
13
|
Mathew SA, Naik C, Cahill PA, Bhonde RR. Placental mesenchymal stromal cells as an alternative tool for therapeutic angiogenesis. Cell Mol Life Sci 2020; 77:253-265. [PMID: 31468060 PMCID: PMC11104823 DOI: 10.1007/s00018-019-03268-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/24/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Dysregulation of angiogenesis is a phenomenon observed in several disorders such as diabetic foot, critical limb ischemia and myocardial infarction. Mesenchymal stromal cells (MSCs) possess angiogenic potential and have recently emerged as a powerful tool for cell therapy to promote angiogenesis. Although bone marrow-derived MSCs are the primary cell of choice, obtaining them has become a challenge. The placenta has become a popular alternative as it is a highly vascular organ, easily available and ethically more favorable with a rich supply of MSCs. Comparatively, placenta-derived MSCs (PMSCs) are clinically promising due to their proliferative, migratory, clonogenic and immunomodulatory properties. PMSCs release a plethora of cytokines and chemokines key to angiogenic signaling and facilitate the possibility of delivering PMSC-derived exosomes as a targeted therapy to promote angiogenesis. However, there still remains the challenge of heterogeneity in the isolated populations, questions on the maternal or fetal origin of these cells and the diversity in previously reported isolation and culture conditions. Nonetheless, the growing rate of clinical trials using PMSCs clearly indicates a shift in favor of PMSCs. The overall aim of the review is to highlight the importance of this rather poorly understood cell type and emphasize the need for further investigations into their angiogenic potential as an alternative source for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Suja Ann Mathew
- School of Regenerative Medicine, Manipal Academy of Higher Education, MAHE, Allalasandra, Near Royal Orchid, Yellahanka, Bangalore, 560 065, India.
| | - Charuta Naik
- School of Regenerative Medicine, Manipal Academy of Higher Education, MAHE, Allalasandra, Near Royal Orchid, Yellahanka, Bangalore, 560 065, India
| | - Paul A Cahill
- School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin Dublin 9, Ireland
| | - Ramesh R Bhonde
- Dr. D.Y. Patil Vidyapeeth (DPU), Pimpri, Pune, 411018, India.
| |
Collapse
|
14
|
Li R, Muraoka T, Kinbara K. Thermo-driven self-assembly of a PEG-containing amphiphile in a bilayer membrane. RSC Adv 2020; 10:25758-25762. [PMID: 35518572 PMCID: PMC9055338 DOI: 10.1039/d0ra03920a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/01/2020] [Indexed: 11/23/2022] Open
Abstract
Self-assembly of lipid molecules in a plasma membrane, namely lipid raft formation, is involved in various dynamic functions of cells. Inspired by the raft formation observed in the cells, here we studied thermally induced self-assembly of a synthetic amphiphile, bola-AkDPA, in a bilayer membrane. The synthetic amphiphile consists of a hydrophobic unit including fluorescent aromatic and aliphatic components and hydrophilic tetraethylene glycol chains attached at both ends of the hydrophobic unit. In a polar solvent, bola-AkDPA formed aggregates to show excimer emission. In a lipid bilayer membrane, bola-AkDPA showed intensified excimer emission upon increase of its concentration or elevation of the temperature; bola-type amphiphiles containing oligoethylene glycol chains likely tend to form self-assemblies in a bilayer membrane triggered by thermal stimuli. A synthetic multi-block amphiphile containing oligoethylene glycol chains formed a self-assembly in a bilayer membrane triggered by thermal stimuli.![]()
Collapse
Affiliation(s)
- Rui Li
- Institute of Multidisciplinary Research for Advanced Materials
- Tohoku University
- Sendai
- Japan
| | - Takahiro Muraoka
- Department of Life Science and Technology
- Tokyo Institute of Technology
- Yokohama
- Japan
| | - Kazushi Kinbara
- Institute of Multidisciplinary Research for Advanced Materials
- Tohoku University
- Sendai
- Japan
- Department of Life Science and Technology
| |
Collapse
|
15
|
Egawa J, Zemljic-Harpf A, Mandyam CD, Niesman IR, Lysenko LV, Kleschevnikov AM, Roth DM, Patel HH, Patel PM, Head BP. Neuron-Targeted Caveolin-1 Promotes Ultrastructural and Functional Hippocampal Synaptic Plasticity. Cereb Cortex 2019; 28:3255-3266. [PMID: 28981594 DOI: 10.1093/cercor/bhx196] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Indexed: 12/15/2022] Open
Abstract
A delicate interneuronal communication between pre- and postsynaptic membranes is critical for synaptic plasticity and the formation of memory. Evidence shows that membrane/lipid rafts (MLRs), plasma membrane microdomains enriched in cholesterol and sphingolipids, organize presynaptic proteins and postsynaptic receptors necessary for synaptic formation and signaling. MLRs establish a cell polarity that facilitates transduction of extracellular cues to the intracellular environment. Here we show that neuron-targeted overexpression of an MLR protein, caveolin-1 (SynCav1), in the adult mouse hippocampus increased the number of presynaptic vesicles per bouton, total excitatory type I glutamatergic synapses, number of same-dendrite multiple-synapse boutons, increased myelination, increased long-term potentiation, and increased MLR-localized N-methyl-d-aspartate receptor subunits (GluN1, GluN2A, and GluN2B). Immunogold electron microscopy revealed that Cav-1 localizes to both the pre- and postsynaptic membrane regions as well as in the synaptic cleft. These findings, which are consistent with a significant increase in ultrastructural and functional synaptic plasticity, provide a fundamental framework that underlies previously demonstrated improvements in learning and memory in adult and aged mice by SynCav1. Such observations suggest that Cav-1 and MLRs alter basic aspects of synapse biology that could serve as potential therapeutic targets to promote neuroplasticity and combat neurodegeneration in a number of neurological disorders.
Collapse
Affiliation(s)
- Junji Egawa
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alice Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chitra D Mandyam
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Larisa V Lysenko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Piyush M Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Sawada A, Wang S, Jian M, Leem J, Wackerbarth J, Egawa J, Schilling JM, Platoshyn O, Zemljic-Harpf A, Roth DM, Patel HH, Patel PM, Marsala M, Head BP. Neuron-targeted caveolin-1 improves neuromuscular function and extends survival in SOD1 G93A mice. FASEB J 2019; 33:7545-7554. [PMID: 30894019 DOI: 10.1096/fj.201802652rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interventions that preserve motor neurons or restore functional motor neuroplasticity may extend longevity in amyotrophic lateral sclerosis (ALS). Delivery of neurotrophins may potentially revive degenerating motor neurons, yet this approach is dependent on the proper subcellular localization of neurotrophin receptor (NTR) to plasmalemmal signaling microdomains, termed membrane/lipid rafts (MLRs). We previously showed that overexpression of synapsin-driven caveolin-1 (Cav-1) (SynCav1) increases MLR localization of NTR [e.g., receptor tyrosine kinase B (TrkB)], promotes hippocampal synaptic and neuroplasticity, and significantly improves learning and memory in aged mice. The present study crossed a SynCav1 transgene-positive (SynCav1+) mouse with the mutant human superoxide dismutase glycine to alanine point mutation at amino acid 93 (hSOD1G93A) mouse model of ALS. When compared with hSOD1G93A, hSOD1G93A/SynCav1+ mice exhibited greater body weight and longer survival as well as better motor function. Microscopic analyses of hSOD1G93A/SynCav1+ spinal cords revealed preserved spinal cord α-motor neurons and preserved mitochondrial morphology. Moreover, hSOD1G93A/SynCav1+ spinal cords contained more MLRs (cholera toxin subunit B positive) and MLR-associated TrkB and Cav-1 protein expression. These findings demonstrate that SynCav1 delays disease progression in a mouse model of ALS, potentially by preserving or restoring NTR expression and localization to MLRs.-Sawada, A., Wang, S., Jian, M., Leem, J., Wackerbarth, J., Egawa, J., Schilling, J. M., Platoshyn, O., Zemljic-Harpf, A., Roth, D. M., Patel, H. H., Patel, P. M., Marsala, M., Head, B. P. Neuron-targeted caveolin-1 improves neuromuscular function and extends survival in SOD1G93A mice.
Collapse
Affiliation(s)
- Atsushi Sawada
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA.,Department of Anesthesiology, Sapporo Medical University, Sapporo, Japan
| | - Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Minyu Jian
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA.,Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Joseph Leem
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Jesse Wackerbarth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Junji Egawa
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA.,Department of Anesthesiology, Nara Medical University, Kashihara, Japan; and
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Oleksandr Platoshyn
- Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Alice Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Piyush M Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Martin Marsala
- Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, School of Medicine, University of California-San Diego, La Jolla, California, USA
| |
Collapse
|
17
|
Limits of the Effective Medium Theory in Particle Amplified Surface Plasmon Resonance Spectroscopy Biosensors. SENSORS 2019; 19:s19030584. [PMID: 30704098 PMCID: PMC6387329 DOI: 10.3390/s19030584] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
Abstract
The resonant wave modes in monomodal and multimodal planar Surface Plasmon Resonance (SPR) sensors and their response to a bidimensional array of gold nanoparticles (AuNPs) are analyzed both theoretically and experimentally, to investigate the parameters that rule the correct nanoparticle counting in the emerging metal nanoparticle-amplified surface plasmon resonance (PA-SPR) spectroscopy. With numerical simulations based on the Finite Element Method (FEM), we evaluate the error performed in the determination of the surface density of nanoparticles σ when the Maxwell-Garnett effective medium theory is used for fast data processing of the SPR reflectivity curves upon nanoparticle detection. The deviation increases directly with the manifestations of non-negligible scattering cross-section of the single nanoparticle, dipole-dipole interactions between adjacent AuNPs and dipolar interactions with the metal substrate. Near field simulations show clearly the set-up of dipolar interactions when the dielectric thickness is smaller than 10 nm and confirm that the anomalous dispersion usually observed experimentally is due to the failure of the effective medium theories. Using citrate stabilized AuNPs with a nominal diameter of about 15 nm, we demonstrate experimentally that Dielectric Loaded Waveguides (DLWGs) can be used as accurate nanocounters in the range of surface density between 20 and 200 NP/µm2, opening the way to the use of PA-SPR spectroscopy on systems mimicking the physiological cell membranes on SiO2 supports.
Collapse
|
18
|
Molecular Targets of Epigallocatechin-Gallate (EGCG): A Special Focus on Signal Transduction and Cancer. Nutrients 2018; 10:nu10121936. [PMID: 30563268 PMCID: PMC6315581 DOI: 10.3390/nu10121936] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022] Open
Abstract
Green tea is a beverage that is widely consumed worldwide and is believed to exert effects on different diseases, including cancer. The major components of green tea are catechins, a family of polyphenols. Among them, epigallocatechin-gallate (EGCG) is the most abundant and biologically active. EGCG is widely studied for its anti-cancer properties. However, the cellular and molecular mechanisms explaining its action have not been completely understood, yet. EGCG is effective in vivo at micromolar concentrations, suggesting that its action is mediated by interaction with specific targets that are involved in the regulation of crucial steps of cell proliferation, survival, and metastatic spread. Recently, several proteins have been identified as EGCG direct interactors. Among them, the trans-membrane receptor 67LR has been identified as a high affinity EGCG receptor. 67LR is a master regulator of many pathways affecting cell proliferation or apoptosis, also regulating cancer stem cells (CSCs) activity. EGCG was also found to be interacting directly with Pin1, TGFR-II, and metalloproteinases (MMPs) (mainly MMP2 and MMP9), which respectively regulate EGCG-dependent inhibition of NF-kB, epithelial-mesenchimal transaction (EMT) and cellular invasion. EGCG interacts with DNA methyltransferases (DNMTs) and histone deacetylases (HDACs), which modulates epigenetic changes. The bulk of this novel knowledge provides information about the mechanisms of action of EGCG and may explain its onco-suppressive function. The identification of crucial signalling pathways that are related to cancer onset and progression whose master regulators interacts with EGCG may disclose intriguing pharmacological targets, and eventually lead to novel combined treatments in which EGCG acts synergistically with known drugs.
Collapse
|
19
|
Mathew SA, Bhonde RR. Omega-3 polyunsaturated fatty acids promote angiogenesis in placenta derived mesenchymal stromal cells. Pharmacol Res 2018; 132:90-98. [PMID: 29665425 DOI: 10.1016/j.phrs.2018.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
Abstract
Enhancement of angiogenesis is solicited in wound repair and regeneration. Mesenchymal stromal cells derived from the placenta (P-MSCs) have an inherent angiogenic potential. Polyunsaturated fatty acids (PUFAs) in turn, specifically the omega-3 (N-3) are essential for growth and development. They are also recommended as dietary supplements during pregnancy. We therefore hypothesized that addition of N-3 PUFAs in P-MSC culture media may enhance their angiogenic potential. Hence, we treated P-MSCs with omega-3 (N-3) fatty acids -Docosahexaenoic acid (DHA) and Eicosapentaenoic acid (EPA) at different concentrations and tested their angiogenic potential. We saw an upregulation of both bFGF and VEGFA. We also found enhanced in vitro tube formation ability of P-MSCs treated with DHA: EPA. We then looked at the influence of the conditioned medium (CM) collected from P-MSCs exposed to DHA: EPA on the key effector cells -HUVECs (Human Umbilical Vein derived endothelial cells and their functionality was further confirmed on chick yolk sac membrane. We found that the CM of P-MSCs exposed to DHA: EPA could enhance angiogenesis in both cases. These result were finally validated in an in vivo matrigel plug assay which revealed enhanced migration and vessel formation in CM treated with DHA: EPA. Our data thus reveals for the first time that supplementation with lower concentration of PUFA enhances the angiogenic potential of P-MSCs making them suitable for chronic wound healing applications.
Collapse
Affiliation(s)
- Suja Ann Mathew
- School of Regenerative Medicine, Manipal University, MAHE, GKVK Post, Bellary Road, Allalasandra, Near Royal Orchid, Yelahanka, Bangalore, 560 065, India.
| | - Ramesh R Bhonde
- Dr. D.Y. Patil Vidyapeeth - (DPU), Pimpri, Pune, 411018, India.
| |
Collapse
|
20
|
Filippini A, Sica G, D'Alessio A. The caveolar membrane system in endothelium: From cell signaling to vascular pathology. J Cell Biochem 2018; 119:5060-5071. [PMID: 29637636 DOI: 10.1002/jcb.26793] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
Caveolae are 50- to 100-nm cholesterol and glycosphingolipid-rich flask-shaped invaginations commonly observed in many terminally differentiated cells. These organelles have been described in many cell types and are particularly abundant in endothelial cells, where they have been involved in the regulation of certain signaling pathways. Specific scaffolding proteins termed caveolins, along with the more recently discovered members of the cavin family, represent the major protein components during caveolae biogenesis. In addition, multiple studies aimed to investigate the expression and the regulation of these proteins significantly contributed to elucidate the role of caveolae and caveolins in endothelial cell physiology and disease. The aim of this review is to survey recent evidence of the involvement of the caveolar network in endothelial cell biology and endothelial cell dysfunction.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gigliola Sica
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessio D'Alessio
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
21
|
Chillà A, Margheri F, Biagioni A, Del Rosso M, Fibbi G, Laurenzana A. Mature and progenitor endothelial cells perform angiogenesis also under protease inhibition: the amoeboid angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:74. [PMID: 29615071 PMCID: PMC5883600 DOI: 10.1186/s13046-018-0742-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023]
Abstract
Background Controlling vascular growth is a challenging aim for the inhibition of tumor growth and metastasis. The amoeboid and mesenchymal types of invasiveness are two modes of migration interchangeable in cancer cells: the Rac-dependent mesenchymal migration requires the activity of proteases; the Rho-ROCK-dependent amoeboid motility is protease-independent and has never been described in endothelial cells. Methods A cocktail of physiologic inhibitors (Ph-C) of serine-proteases, metallo-proteases and cysteine-proteases, mimicking the physiological environment that cells encounter during their migration within the angiogenesis sites was used to induce amoeboid style migration of Endothelial colony forming cells (ECFCs) and mature endothelial cells (ECs). To evaluate the mesenchymal-ameboid transition RhoA and Rac1 activation assays were performed along with immunofluorescence analysis of proteins involved in cytoskeleton organization. Cell invasion was studied in Boyden chambers and Matrigel plug assay for the in vivo angiogenesis. Results In the present study we showed in both ECFCs and ECs, a decrease of activated Rac1 and an increase of activated RhoA upon shifting of cells to the amoeboid conditions. In presence of Ph-C inhibitors both cell lines acquired a round morphology and Matrigel invasion was greatly enhanced with respect to that observed in the absence of protease inhibition. We also observed that the urokinase-plasminogen-activator (uPAR) receptor silencing and uPAR-integrin uncoupling with the M25 peptide abolished both mesenchymal and amoeboid angiogenesis of ECFCs and ECs in vitro and in vivo, indicating a role of the uPAR-integrin-actin axis in the regulation of amoeboid angiogenesis. Furthermore, under amoeboid conditions endothelial cells seem to be indifferent to VEGF stimulation, which induces an amoeboid signaling pattern also in mesenchymal conditions. Conclusion Here we first provide a data set disclosing that endothelial cells can move and differentiate into vascular structures in vitro and in vivo also in the absence of proteases activity, performing a new type of neovascularization: the “amoeboid angiogenesis”. uPAR is indispensable for ECs and ECFCs to perform an efficient amoeboid angiogenesis. Therefore, uPAR silencing or the block of its integrin-interaction, together with standard treatment against VEGF, could be a possible solution for angiogenesis inhibition. Electronic supplementary material The online version of this article (10.1186/s13046-018-0742-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy.
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Viale G.B. Morgagni, 50-50134, Florence, Italy
| |
Collapse
|
22
|
N-3 vs. n-6 fatty acids differentially influence calcium signalling and adhesion of inflammatory activated monocytes: impact of lipid rafts. Inflamm Res 2016; 65:881-894. [DOI: 10.1007/s00011-016-0971-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 04/01/2016] [Accepted: 06/28/2016] [Indexed: 11/30/2022] Open
|
23
|
Carquin M, D'Auria L, Pollet H, Bongarzone ER, Tyteca D. Recent progress on lipid lateral heterogeneity in plasma membranes: From rafts to submicrometric domains. Prog Lipid Res 2015; 62:1-24. [PMID: 26738447 DOI: 10.1016/j.plipres.2015.12.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/29/2022]
Abstract
The concept of transient nanometric domains known as lipid rafts has brought interest to reassess the validity of the Singer-Nicolson model of a fluid bilayer for cell membranes. However, this new view is still insufficient to explain the cellular control of surface lipid diversity or membrane deformability. During the past decades, the hypothesis that some lipids form large (submicrometric/mesoscale vs nanometric rafts) and stable (>min vs s) membrane domains has emerged, largely based on indirect methods. Morphological evidence for stable submicrometric lipid domains, well-accepted for artificial and highly specialized biological membranes, was further reported for a variety of living cells from prokaryot es to yeast and mammalian cells. However, results remained questioned based on limitations of available fluorescent tools, use of poor lipid fixatives, and imaging artifacts due to non-resolved membrane projections. In this review, we will discuss recent evidence generated using powerful and innovative approaches such as lipid-specific toxin fragments that support the existence of submicrometric domains. We will integrate documented mechanisms involved in the formation and maintenance of these domains, and provide a perspective on their relevance on membrane deformability and regulation of membrane protein distribution.
Collapse
Affiliation(s)
- Mélanie Carquin
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ludovic D'Auria
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Hélène Pollet
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ernesto R Bongarzone
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| |
Collapse
|
24
|
Carquin M, Conrard L, Pollet H, Van Der Smissen P, Cominelli A, Veiga-da-Cunha M, Courtoy PJ, Tyteca D. Cholesterol segregates into submicrometric domains at the living erythrocyte membrane: evidence and regulation. Cell Mol Life Sci 2015; 72:4633-51. [PMID: 26077601 PMCID: PMC11113096 DOI: 10.1007/s00018-015-1951-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/07/2015] [Accepted: 06/04/2015] [Indexed: 01/01/2023]
Abstract
Although cholesterol is essential for membrane fluidity and deformability, the level of its lateral heterogeneity at the plasma membrane of living cells is poorly understood due to lack of appropriate probe. We here report on the usefulness of the D4 fragment of Clostridium perfringens toxin fused to mCherry (theta*), as specific, non-toxic, sensitive and quantitative cholesterol-labeling tool, using erythrocyte flat membrane. By confocal microscopy, theta* labels cholesterol-enriched submicrometric domains in coverslip-spread but also gel-suspended (non-stretched) fresh erythrocytes, suggesting in vivo relevance. Cholesterol domains on spread erythrocytes are stable in time and space, restricted by membrane:spectrin anchorage via 4.1R complexes, and depend on temperature and sphingomyelin, indicating combined regulation by extrinsic membrane:cytoskeleton interaction and by intrinsic lipid packing. Cholesterol domains partially co-localize with BODIPY-sphingomyelin-enriched domains. In conclusion, we show that theta* is a useful vital probe to study cholesterol organization and demonstrate that cholesterol forms submicrometric domains in living cells.
Collapse
Affiliation(s)
- Mélanie Carquin
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Louise Conrard
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Hélène Pollet
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Patrick Van Der Smissen
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Antoine Cominelli
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Maria Veiga-da-Cunha
- Laboratory of Physiological Chemistry, de Duve Institute and Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Pierre J Courtoy
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium.
| |
Collapse
|
25
|
Suica VI, Uyy E, Boteanu RM, Ivan L, Antohe F. Alteration of actin dependent signaling pathways associated with membrane microdomains in hyperlipidemia. Proteome Sci 2015; 13:30. [PMID: 26628893 PMCID: PMC4666118 DOI: 10.1186/s12953-015-0087-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/24/2015] [Indexed: 01/05/2023] Open
Abstract
Background Membrane microdomains represent dynamic membrane nano-assemblies enriched in signaling molecules suggesting their active involvement in not only physiological but also pathological molecular processes. The hyperlipidemic stress is a major risk factor of atherosclerosis, but its exact mechanisms of action at the membrane microdomains level remain elusive. The aim of the present study was to determine whether membrane-cytoskeleton proteome in the pulmonary tissue could be modulated by the hyperlipidemic stress, a major risk factor of atherosclerosis. Results High resolution mass spectrometry based proteomics analysis was performed for detergent resistant membrane microdomains isolated from lung homogenates of control, ApoE deficient and statin treated ApoE deficient mice. The findings of the study allowed the identification with high confidence of 1925 proteins, 291 of which were found significantly altered by the modified genetic background, by the statin treatment or both conditions. Principal component analysis revealed a proximal partitioning of the biological replicates, but also a distinct spatial scattering of the sample groups, highlighting different quantitative profiles. The statistical significant over-representation of Regulation of actin cytoskeleton, Focal adhesion and Adherens junction Kyoto Encyclopedia of Genes and Genomes signaling pathways was demonstrated through bioinformatics analysis. The three inter-relation maps comprised 29 of regulated proteins, proving membrane-cytoskeleton coupling targeting and alteration by hyperlipidemia and/or statin treatment. Conclusions The findings of the study allowed the identification with high confidence of the main proteins modulated by the hyperlipidemic stress involved in the actin-dependent pathways. Our study provides the basis for future work probing how the protein activities at the membrane-cytoskeleton interface are dependent upon genetic induced hyperlipidemia. Electronic supplementary material The online version of this article (doi:10.1186/s12953-015-0087-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Viorel-Iulian Suica
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 BP Hasdeu Street, PO Box 35-14, 050568 Bucharest, Romania
| | - Elena Uyy
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 BP Hasdeu Street, PO Box 35-14, 050568 Bucharest, Romania
| | - Raluca Maria Boteanu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 BP Hasdeu Street, PO Box 35-14, 050568 Bucharest, Romania
| | - Luminita Ivan
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 BP Hasdeu Street, PO Box 35-14, 050568 Bucharest, Romania
| | - Felicia Antohe
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 BP Hasdeu Street, PO Box 35-14, 050568 Bucharest, Romania
| |
Collapse
|