1
|
Soteriou C, Xu M, Connell SD, Tyler AII, Kalli AC, Thorne JL. Two cooperative lipid binding sites within the pleckstrin homology domain are necessary for AKT binding and stabilization to the plasma membrane. Structure 2024:S0969-2126(24)00455-6. [PMID: 39504965 DOI: 10.1016/j.str.2024.10.020] [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: 10/19/2023] [Revised: 09/02/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
Almost four decades after the identification of the AKT protein and understanding of its role in cancer, barriers remain in the translation of AKT inhibitors for clinical applications. Here, we provide new molecular insight into the first step of AKT activation where AKT binds to the plasma membrane and its orientation is stabilized in a bilayer with lateral heterogeneity (Lo-Ld phase coexistence). We have applied molecular dynamic simulations and molecular and cell biology approaches, and demonstrate that AKT recruitment to the membrane requires a second binding site in the AKT pleckstrin homology (PH) domain that acts cooperatively with the known canonical binding site. Given the precision with which we have identified the protein-lipid interactions, the study offers new directions for AKT-targeted therapy and for testing small molecules to target these specific amino acid-PIP molecular bonds.
Collapse
Affiliation(s)
- Chrysa Soteriou
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK; Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK; Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Mengfan Xu
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Simon D Connell
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK; Astbury Center for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Arwen I I Tyler
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Antreas C Kalli
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK; Astbury Center for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - James L Thorne
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
2
|
Huang YX, Lin YC, Lin CK, Huang HM. ω-9 monounsaturated fatty acids in Sapindus mukorossi seed oil enhance calcium deposition expression of Wharton's jelly mesenchymal stem cells. Tissue Cell 2024; 91:102595. [PMID: 39481221 DOI: 10.1016/j.tice.2024.102595] [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: 06/05/2024] [Revised: 10/12/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Promoting osteogenesis is crucial to improve successful bone regeneration in bone tissue engineering. Several studies on osteogenesis have reported positive effects of ω-9 monounsaturated fatty acids (MUFA) on bone regeneration. This study examined the potential of ω-9 monounsaturated fatty acid abundant seed oil mechanically extracted from Sapindus mukorossi (S. mukorossi) fruit to improve osteogeneses. After showing the presence of ω-9 MUFA in S. mukorossi seed oil (SM oil) through GC-MS spectrum analysis, the proliferation and differentiation of human umbilical cord Wharton's jelly mesenchymal stem cells (WJMSCs) under SM treatment was evaluated. Our results indicate that WJMSC differentiation was induced by adding an osteogenesis-induced medium combined with SM oil. A high level of calcium deposition expression in WJMSCs induced by SM oil appears to be due to the effect of oleic acid and eicosenoic acid, both of which are ω-9 monounsaturated fatty acids. In addition, we found major contributors of SM oil-promoted WJMSC osteogenesis to be extracellular signal-regulated kinase (ERKs), c-Jun N-terminal kinase (JNKs), and the p38 MAPK pathway. The enhancement of WJMSC osteogenesis via ERK/MAPK pathways as demonstrated by qPCR analysis indicates the promise of SM oil for stem cell-based bone tissue engineering applications.
Collapse
Affiliation(s)
- Yu-Xuan Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Chung Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chin-Kai Lin
- Department of Dentistry, En Chu Kong Hospital, New Taipei City 237, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
3
|
Casarrubios L, Cicuéndez M, Polo-Montalvo A, Feito MJ, Martínez-Del-Pozo Á, Arcos D, Duarte IF, Portolés MT. Metabolomic characterization of MC3T3-E1pre-osteoblast differentiation induced by ipriflavone-loaded mesoporous nanospheres. BIOMATERIALS ADVANCES 2024; 166:214085. [PMID: 39490191 DOI: 10.1016/j.bioadv.2024.214085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
This study reports on the metabolic changes accompanying the differentiation of MC3T3-E1 osteoprogenitor cells induced by mesoporous bioactive glass nanospheres (nMBG) loaded with ipriflavone (nMBG-IP). Ipriflavone (IP) is a synthetic isoflavone known for inhibiting bone resorption, maintaining bone density, and preventing osteoporosis. Delivering IP intracellularly is a promising strategy to modulate bone remodeling at significantly lower doses compared to free drug administration. Our results demonstrate that nMBG are efficiently internalized by pre-osteoblasts and, when loaded with IP, induce their differentiation. This differentiation process is accompanied by pronounced metabolic alterations, as monitored by NMR analysis of medium supernatants and cell extracts (exo- and endo-metabolomics, respectively). The main effects include an early-stage intensification of glycolysis and changes in several metabolic pathways, such as nucleobase metabolism, osmoregulatory and antioxidant pathways, and lipid metabolism. Notably, the metabolic impacts of nMBG-IP and free IP were very similar, whereas nMBG alone induced only mild changes in the intracellular metabolic profile without affecting the cells' consumption/secretion patterns or lipid composition. This finding indicates that the observed effects are primarily related to IP-induced differentiation and that nMBG nanospheres serve as convenient carriers with both efficient internalization and minimal metabolic impact. Furthermore, the observed link between pre-osteoblast differentiation and metabolism underscores the potential of utilizing metabolites and metabolic reprogramming as strategies to modulate the osteogenic process, for instance, in the context of osteoporosis and other bone diseases.
Collapse
Affiliation(s)
- Laura Casarrubios
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Mónica Cicuéndez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Alberto Polo-Montalvo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - María José Feito
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Álvaro Martínez-Del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Daniel Arcos
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - María Teresa Portolés
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain.
| |
Collapse
|
4
|
Wang H, Cui G, Meng X, Wang X, Luan Z, Gong J, Dai S, Gao T. Association of serum fatty acids with bone health in rural elderly population in Qingdao, China: A cross-sectional study. Lipids 2024. [PMID: 39394914 DOI: 10.1002/lipd.12421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/11/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
As a type of macronutrient, fatty acids (FA) play significant roles in the bone health of elderly people. However, the specific association between different types of FA and bone health is not fully understood, especially in rural elderly populations. To address this gap, a study was conducted in rural areas of Qingdao, China. Participants aged 65 and older were randomly recruited from 11 rural villages in Licha town, Qingdao City. The levels of serum FA in their serum were measured to investigate the associations between FA and bone mass. The results showed that levels of saturated fatty acids (SFA), n-3 polyunsaturated fatty acids (n-3 PUFA), and n-6 polyunsaturated fatty acids (n-6 PUFA) were all significantly associated with bone mass. Specifically, higher levels of SFA were positively correlated with low bone mass (LBM), while PUFA levels were inversely correlated with LBM. Furthermore, the odds ratio (OR) for LBM exhibited a significant nonlinear dose-response relationship (pnonlinearity = 0.1989) with SFA levels, and a significant nonlinear dose-dependent relationship was also observed with the levels of n-3PUFA and n-6PUFA (pnonlinearity = 0.6183, 0.5808, respectively), indicating that increasing dietary PUFA intake appropriately and controlling SFA intake may benefit the bone health of elderly individuals in rural areas.
Collapse
Affiliation(s)
- Haoyu Wang
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- School of Public Health, Qingdao University, Qingdao, China
| | - Guangwei Cui
- Health Service Center of Licha Community, Qingdao, China
| | - Xiangyuan Meng
- School of Public Health, Jilin University, Changchun, China
| | - Xingxu Wang
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- School of Public Health, Qingdao University, Qingdao, China
| | - Zhaohui Luan
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- School of Public Health, Qingdao University, Qingdao, China
| | - Jianbao Gong
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Shiyou Dai
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Tianlin Gao
- School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Xiao H, Li W, Qin Y, Lin Z, Qian C, Wu M, Xia Y, Bai J, Geng D. Crosstalk between Lipid Metabolism and Bone Homeostasis: Exploring Intricate Signaling Relationships. RESEARCH (WASHINGTON, D.C.) 2024; 7:0447. [PMID: 39165638 PMCID: PMC11334918 DOI: 10.34133/research.0447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Bone is a dynamic tissue reshaped by constant bone formation and bone resorption to maintain its function. The skeletal system accounts for approximately 70% of the total volume of the body, and continuous bone remodeling requires quantities of energy and material consumption. Adipose tissue is the main energy storehouse of the body and has a strong adaptive capacity to participate in the regulation of various physiological processes. Considering that obesity and metabolic syndrome have become major public health challenges, while osteoporosis and osteoporotic fractures have become other major health problems in the aging population, it would be interesting to explore these 2 diseases together. Currently, an increasing number of researchers are focusing on the interactions between multiple tissue systems, i.e., multiple organs and tissues that are functionally coordinated together and pathologically pathologically interact with each other in the body. However, there is lack of detailed reviews summarizing the effects of lipid metabolism on bone homeostasis and the interactions between adipose tissue and bone tissue. This review provides a detailed summary of recent advances in understanding how lipid molecules and adipose-derived hormones affect bone homeostasis, how bone tissue, as a metabolic organ, affects lipid metabolism, and how lipid metabolism is regulated by bone-derived cytokines.
Collapse
Affiliation(s)
- Haixiang Xiao
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230022, China
| | - Wenming Li
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yi Qin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhixiang Lin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Chen Qian
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingzhou Wu
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yu Xia
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, Jingjiang People’s Hospital Affiliated to Yangzhou University, Jingjiang 214500, Jiangsu Province, China
| | - Dechun Geng
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
6
|
Longarzo ML, Vázquez RF, Bellini MJ, Zamora RA, Redondo-Morata L, Giannotti MI, Oliveira Jr ON, Fanani ML, Maté SM. Understanding the effects of omega-3 fatty acid supplementation on the physical properties of brain lipid membranes. iScience 2024; 27:110362. [PMID: 39071883 PMCID: PMC11277689 DOI: 10.1016/j.isci.2024.110362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/24/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
A deficiency in omega-3 fatty acids (ω3 FAs) in the brain has been correlated with cognitive impairment, learning deficiencies, and behavioral changes. In this study, we provided ω3 FAs as a supplement to spontaneously hypertensive rats (SHR+ ω3). Our focus was on examining the impact of dietary supplementation on the physicochemical properties of the brain-cell membranes. Significant increases in ω3 levels in the cerebral cortex of SHR+ ω3 were observed, leading to alterations in brain lipid membranes molecular packing, elasticity, and lipid miscibility, resulting in an augmented phase disparity. Results from synthetic lipid mixtures confirmed the disordering effect introduced by ω3 lipids, showing its consequences on the hydration levels of the monolayers and the organization of the membrane domains. These findings suggest that dietary ω3 FAs influence the organization of brain membranes, providing insight into a potential mechanism for the broad effects of dietary fat on brain health and disease.
Collapse
Affiliation(s)
- María L. Longarzo
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - Romina F. Vázquez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - María J. Bellini
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| | - Ricardo A. Zamora
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Instituto de Investigación Interdisciplinaria (I³), Vicerrectoría Académica, and Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Campus Lircay, Talca 3460000, Chile
| | - Lorena Redondo-Morata
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Marina I. Giannotti
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- CIBER-BBN, ISCIII, 08028 Barcelona, Spain
- Department of Materials Science and Physical Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Osvaldo N. Oliveira Jr
- São Carlos Institute of Physics (IFSC-USP), University of São Paulo, 13566-590 São Carlos, São Paulo, Brazil
| | - María L. Fanani
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Cordoba, Argentina
- Departamento de Química Biológica Raquel Caputto, Facultad de Cs. Químicas, Universidad Nacional de Córdoba. Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Sabina M. Maté
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, (1900), La Plata, Argentina
| |
Collapse
|
7
|
Wang ZH, Combs C, Zhao W, Xu H. A protocol for measuring plasma membrane tension in the Drosophila ovary using fluorescence lifetime imaging microscopy. STAR Protoc 2024; 5:102959. [PMID: 38489272 PMCID: PMC10951584 DOI: 10.1016/j.xpro.2024.102959] [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: 01/05/2024] [Revised: 02/05/2024] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
Mechanosensation of plasma membrane tension by various mechanoresponsive machineries is crucial for regulating stem cell fate, cell adhesion, and tissue morphogenesis. Here, we present a protocol for evaluating plasma membrane stretching during the differentiation of Drosophila ovarian cyst using a fluorescent lipid tension reporter (Flipper-TR). We describe the steps for microphone setup, ovary dissection, Flipper-TR staining, fluorescence lifetime imaging microscopy imaging, and image processing and analysis. This protocol demonstrates the utility of Flipper-TR for investigating the impact of mechanical forces in living tissue. For complete details on the use and execution of this protocol, please refer to Wang et al.1.
Collapse
Affiliation(s)
- Zong-Heng Wang
- Laboratory of Molecular Genetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian Combs
- Light Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenjing Zhao
- Laboratory of Molecular Genetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong Xu
- Laboratory of Molecular Genetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Kuo PJ, Lin YH, Huang YX, Lee SY, Huang HM. Effects of Sapindus mukorossi Seed Oil on Bone Healing Efficiency: An Animal Study. Int J Mol Sci 2024; 25:6749. [PMID: 38928455 PMCID: PMC11204041 DOI: 10.3390/ijms25126749] [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: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Natural products have attracted great interest in the development of tissue engineering. Recent studies have demonstrated that unsaturated fatty acids found in natural plant seed oil may exhibit positive osteogenic effects; however, few in vivo studies have focused on the use of plant seed oil for bone regeneration. The aim of this study is to investigate the effects of seed oil found in Sapindus mukorossi (S. mukorossi) on the osteogenic differentiation of mesenchymal stem cells and bone growth in artificial bone defects in vivo. In this study, Wharton-jelly-derived mesenchymal stem cells (WJMSCs) were co-cultured with S. mukorossi seed oil. Cellular osteogenic capacity was assessed using Alizarin Red S staining. Real-time PCR was carried out to evaluate ALP and OCN gene expression. The potential of S. mukorossi seed oil to enhance bone growth was assessed using an animal model. Four 6 mm circular defects were prepared at the parietal bone of New Zealand white rabbits. The defects were filled with hydrogel and hydrogel-S. mukorossi seed oil, respectively. Quantitative analysis of micro-computed tomography (Micro-CT) and histological images was conducted to compare differences in osteogenesis between oil-treated and untreated samples. Although our results showed no significant differences in viability between WJMSCs treated with and without S. mukorossi seed oil, under osteogenic conditions, S. mukorossi seed oil facilitated an increase in mineralized nodule secretion and upregulated the expression of ALP and OCN genes in the cells (p < 0.05). In the animal study, both micro-CT and histological evaluations revealed that new bone formation in artificial bone defects treated with S. mukorossi seed oil were nearly doubled compared to control defects (p < 0.05) after 4 weeks of healing. Based on these findings, it is reasonable to suggest that S. mukorossi seed oil holds promise as a potential candidate for enhancing bone healing efficiency in bone tissue engineering.
Collapse
Affiliation(s)
- Po-Jan Kuo
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yu-Hsiang Lin
- Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; (Y.-H.L.); (S.-Y.L.)
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Xuan Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Sheng-Yang Lee
- Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; (Y.-H.L.); (S.-Y.L.)
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| |
Collapse
|
9
|
Castello-Serrano I, Heberle FA, Diaz-Rohrer B, Ippolito R, Shurer CR, Lujan P, Campelo F, Levental KR, Levental I. Partitioning to ordered membrane domains regulates the kinetics of secretory traffic. eLife 2024; 12:RP89306. [PMID: 38837189 PMCID: PMC11152573 DOI: 10.7554/elife.89306] [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] [Indexed: 06/06/2024] Open
Abstract
The organelles of eukaryotic cells maintain distinct protein and lipid compositions required for their specific functions. The mechanisms by which many of these components are sorted to their specific locations remain unknown. While some motifs mediating subcellular protein localization have been identified, many membrane proteins and most membrane lipids lack known sorting determinants. A putative mechanism for sorting of membrane components is based on membrane domains known as lipid rafts, which are laterally segregated nanoscopic assemblies of specific lipids and proteins. To assess the role of such domains in the secretory pathway, we applied a robust tool for synchronized secretory protein traffic (RUSH, Retention Using Selective Hooks) to protein constructs with defined affinity for raft phases. These constructs consist solely of single-pass transmembrane domains (TMDs) and, lacking other sorting determinants, constitute probes for membrane domain-mediated trafficking. We find that while raft affinity can be sufficient for steady-state PM localization, it is not sufficient for rapid exit from the endoplasmic reticulum (ER), which is instead mediated by a short cytosolic peptide motif. In contrast, we find that Golgi exit kinetics are highly dependent on raft affinity, with raft preferring probes exiting the Golgi ~2.5-fold faster than probes with minimal raft affinity. We rationalize these observations with a kinetic model of secretory trafficking, wherein Golgi export can be facilitated by protein association with raft domains. These observations support a role for raft-like membrane domains in the secretory pathway and establish an experimental paradigm for dissecting its underlying machinery.
Collapse
Affiliation(s)
- Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | | | | | - Rossana Ippolito
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Carolyn R Shurer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Pablo Lujan
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Felix Campelo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
10
|
Qiu Y, Wang X, Sun Y, Jin T, Tang R, Zhou X, Xu M, Gan Y, Wang R, Luo H, Liu M, Tang X. ACSL4-Mediated Membrane Phospholipid Remodeling Induces Integrin β1 Activation to Facilitate Triple-Negative Breast Cancer Metastasis. Cancer Res 2024; 84:1856-1871. [PMID: 38471082 PMCID: PMC11148537 DOI: 10.1158/0008-5472.can-23-2491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and has a poor prognosis and a high propensity to metastasize. Lipid metabolism has emerged as a critical regulator of tumor progression and metastasis in other cancer types. Characterization of the lipid metabolic features of TNBC could provide important insights into the drivers of TNBC metastasis. Here, we showed that metastatic TNBC tumors harbor more unsaturated phospholipids, especially long-chain polyunsaturated fatty acids, at the sn-2 position of phosphatidylcholine and phosphatidylethanolamine compared with primary tumors. Metastatic TNBC tumors upregulated ACSL4, a long-chain polyunsaturated acyl-CoA synthetase that drives the preferential incorporation of polyunsaturated fatty acids into phospholipids, resulting in the alteration of membrane phospholipid composition and properties. Moreover, ACSL4-mediated phospholipid remodeling of the cell membrane induced lipid-raft localization and activation of integrin β1 in a CD47-dependent manner, which led to downstream focal adhesion kinase phosphorylation that promoted metastasis. Importantly, pharmacologic inhibition of ACSL4 suppressed tumor growth and metastasis and increased chemosensitivity in TNBC models in vivo. These findings indicate that ACSL4-mediated phospholipid remodeling enables TNBC metastasis and can be inhibited as a potential strategy to improve the efficacy of chemotherapy in TNBC. SIGNIFICANCE ACSL4 upregulation in triple-negative breast cancer alters cell membrane phospholipid composition to increase integrin β1 activation and drive metastasis, indicating that targeting ACSL4 could potentially block metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Yuxiang Qiu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xing Wang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Ting Jin
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xinyue Zhou
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ming Xu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yubi Gan
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Rui Wang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xi Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Lin WJ, Chiang AWT, Zhou EH, Liang C, Liu CH, Ma WL, Cheng WC, Lewis NE. iLipidome: enhancing statistical power and interpretability using hidden biosynthetic interdependencies in the lipidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594607. [PMID: 38826229 PMCID: PMC11142111 DOI: 10.1101/2024.05.16.594607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Numerous biological processes and diseases are influenced by lipid composition. Advances in lipidomics are elucidating their roles, but analyzing and interpreting lipidomics data at the systems level remain challenging. To address this, we present iLipidome, a method for analyzing lipidomics data in the context of the lipid biosynthetic network, thus accounting for the interdependence of measured lipids. iLipidome enhances statistical power, enables reliable clustering and lipid enrichment analysis, and links lipidomic changes to their genetic origins. We applied iLipidome to investigate mechanisms driving changes in cellular lipidomes following supplementation of docosahexaenoic acid (DHA) and successfully identified the genetic causes of alterations. We further demonstrated how iLipidome can disclose enzyme-substrate specificity and pinpoint prospective glioblastoma therapeutic targets. Finally, iLipidome enabled us to explore underlying mechanisms of cardiovascular disease and could guide the discovery of early lipid biomarkers. Thus, iLipidome can assist researchers studying the essence of lipidomic data and advance the field of lipid biology.
Collapse
|
12
|
Ponsuksili S, Siengdee P, Li S, Kriangwanich W, Oster M, Reyer H, Wimmers K. Effect of metabolically divergent pig breeds and tissues on mesenchymal stem cell expression patterns during adipogenesis. BMC Genomics 2024; 25:407. [PMID: 38664635 PMCID: PMC11044395 DOI: 10.1186/s12864-024-10308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Unraveling the intricate and tightly regulated process of adipogenesis, involving coordinated activation of transcription factors and signaling pathways, is essential for addressing obesity and related metabolic disorders. The molecular pathways recruited by mesenchymal stem cells (MSCs) during adipogenesis are also dependent on the different sources of the cells and genetic backgrounds of donors, which contribute to the functional heterogeneity of the stem cells and consequently affect the developmental features and fate of the cells. METHODS In this study, the alteration of transcripts during differentiation of synovial mesenchymal stem cells (SMSCs) derived from fibrous synovium (FS) and adipose synovial tissue (FP) of two pig breeds differing in growth performance (German Landrace (DL)) and fat deposition (Angeln Saddleback (AS)) was investigated. SMSCs from both tissues and breeds were stimulated to differentiate into adipocytes in vitro and sampled at four time points (day 1, day 4, day 7 and day 14) to obtain transcriptomic data. RESULTS We observed numerous signaling pathways related to the cell cycle, cell division, cell migration, or cell proliferation during early stages of adipogenesis. As the differentiation process progresses, cells begin to accumulate intracellular lipid droplets and changes in gene expression patterns in particular of adipocyte-specific markers occur. PI3K-Akt signaling and metabolic pathways changed most during adipogenesis, while p53 signaling and ferroptosis were affected late in adipogenesis. When comparing MSCs from FS and FP, only a limited number of differentially expressed genes (DEGs) and enriched signaling pathways were identified. Metabolic pathways, including fat, energy or amino acid metabolism, were highly enriched in the AS breed SMSCs compared to those of the DL breed, especially at day 7 of adipogenesis, suggesting retention of the characteristic metabolic features of their original source, demonstrating donor memory in culture. In contrast, the DL SMSCs were more enriched in immune signaling pathways. CONCLUSIONS Our study has provided important insights into the dynamics of adipogenesis and revealed metabolic shifts in SMSCs associated with different cell sources and genetic backgrounds of donors. This emphasises the critical role of metabolic and genetic factors as important indications and criteria for donor stem cell selection.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Puntita Siengdee
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
- Program in Applied Biological Sciences: Environmental Health, Chulabhorn Graduate Institute, Kamphaeng Phet 6 Road, Laksi, 10210, Bangkok, Thailand
| | - Shuaichen Li
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Wannapimol Kriangwanich
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
- Department of Veterinary Biosciences and Public Health, Faculty of Veterinary Medicine, Chiang Mai University, 50100, Chiang Mai, Thailand
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University Rostock, 18059, Rostock, Germany
| |
Collapse
|
13
|
Levental KR, Henry WS. Lipidomes define immune cell identity. Nat Cell Biol 2024; 26:516-518. [PMID: 38589530 DOI: 10.1038/s41556-024-01398-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Affiliation(s)
- Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA.
| | - Whitney S Henry
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, MIT, Cambridge, MA, USA.
| |
Collapse
|
14
|
Henry WS, Müller S, Yang JS, Innes-Gold S, Das S, Reinhardt F, Sigmund K, Phadnis VV, Wan Z, Eaton E, Sampaio JL, Bell GW, Viravalli A, Hammond PT, Kamm RD, Cohen AE, Boehnke N, Hsu VW, Levental KR, Rodriguez R, Weinberg RA. Ether lipids influence cancer cell fate by modulating iron uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585922. [PMID: 38562716 PMCID: PMC10983928 DOI: 10.1101/2024.03.20.585922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cancer cell fate has been widely ascribed to mutational changes within protein-coding genes associated with tumor suppressors and oncogenes. In contrast, the mechanisms through which the biophysical properties of membrane lipids influence cancer cell survival, dedifferentiation and metastasis have received little scrutiny. Here, we report that cancer cells endowed with a high metastatic ability and cancer stem cell-like traits employ ether lipids to maintain low membrane tension and high membrane fluidity. Using genetic approaches and lipid reconstitution assays, we show that these ether lipid-regulated biophysical properties permit non-clathrin-mediated iron endocytosis via CD44, leading directly to significant increases in intracellular redox-active iron and enhanced ferroptosis susceptibility. Using a combination of in vitro three-dimensional microvascular network systems and in vivo animal models, we show that loss of ether lipids also strongly attenuates extravasation, metastatic burden and cancer stemness. These findings illuminate a mechanism whereby ether lipids in carcinoma cells serve as key regulators of malignant progression while conferring a unique vulnerability that can be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Whitney S Henry
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe Labellisée Ligue Contre le Cancer, Paris 75005, France
| | - Jia-Shu Yang
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, and Dept. of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Innes-Gold
- Dept. of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sunny Das
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Ferenc Reinhardt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Kim Sigmund
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Vaishnavi V Phadnis
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Dept. of Biology, MIT, Cambridge, MA 02139, USA
| | - Zhengpeng Wan
- Dept. of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Elinor Eaton
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Julio L Sampaio
- Institut Curie, INSERM, Mines ParisTech, Paris 75005, France
| | - George W Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Amartya Viravalli
- Dept. of Chemical Engineering and Materials Science, University of Minnesota Minneapolis, MN 55455, USA
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Dept. of Chemical Engineering, MIT, Cambridge, MA 02139, USA
- Senior author
| | - Roger D Kamm
- Dept. of Biological Engineering, MIT, Cambridge, MA 02139, USA
- Dept. of Physics, Harvard University, Cambridge, MA 02138, USA
- Senior author
| | - Adam E Cohen
- Dept. of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Dept. of Physics, Harvard University, Cambridge, MA 02138, USA
- Senior author
| | - Natalie Boehnke
- Dept. of Chemical Engineering and Materials Science, University of Minnesota Minneapolis, MN 55455, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Senior author
| | - Victor W Hsu
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, and Dept. of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Senior author
| | - Kandice R Levental
- Dept. of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
- Senior author
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, Equipe Labellisée Ligue Contre le Cancer, Paris 75005, France
- Senior author
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Dept. of Biology, MIT, Cambridge, MA 02139, USA
- Ludwig Center for Molecular Oncology, Cambridge, MA 02139, USA
- Senior author
| |
Collapse
|
15
|
Müller GA, Müller TD. Transfer of membrane(s) matter(s)-non-genetic inheritance of (metabolic) phenotypes? Front Mol Biosci 2024; 11:1347397. [PMID: 38516184 PMCID: PMC10955475 DOI: 10.3389/fmolb.2024.1347397] [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] [Received: 11/30/2023] [Accepted: 01/26/2024] [Indexed: 03/23/2024] Open
Abstract
Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are anchored at the outer phospholipid layer of eukaryotic plasma membranes exclusively by a glycolipid. GPI-APs are not only released into extracellular compartments by lipolytic cleavage. In addition, certain GPI-APs with the glycosylphosphatidylinositol anchor including their fatty acids remaining coupled to the carboxy-terminus of their protein components are also detectable in body fluids, in response to certain stimuli, such as oxidative stress, radicals or high-fat diet. As a consequence, the fatty acid moieties of GPI-APs must be shielded from access of the aqueous environment by incorporation into membranes of extracellular vesicles or into micelle-like complexes together with (lyso)phospholipids and cholesterol. The GPI-APs released from somatic cells and tissues are transferred via those complexes or EVs to somatic as well as pluripotent stem cells with metabolic consequences, such as upregulation of glycogen and lipid synthesis. From these and additional findings, the following hypotheses are developed: i) Transfer of GPI-APs via EVs or micelle-like complexes leads to the induction of new phenotypes in the daughter cells or zygotes, which are presumably not restricted to metabolism. ii) The membrane topographies transferred by the concerted action of GPI-APs and interacting components are replicated by self-organization and self-templation and remain accessible to structural changes by environmental factors. iii) Transfer from mother cells and gametes to their daughter cells and zygotes, respectively, is not restricted to DNA and genes, but also encompasses non-genetic matter, such as GPI-APs and specific membrane constituents. iv) The intergenerational transfer of membrane matter between mammalian organisms is understood as an epigenetic mechanism for phenotypic plasticity, which does not rely on modifications of DNA and histones, but is regarded as molecular mechanism for the inheritance of acquired traits, such as complex metabolic diseases. v) The missing interest in research of non-genetic matter of inheritance, which may be interpreted in the sense of Darwin's "Gemmules" or Galton's "Stirps", should be addressed in future investigations of the philosophy of science and sociology of media.
Collapse
Affiliation(s)
- Günter A. Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Oberschleissheim, Germany
- Department of Media Studies, Media, Culture and Society, Faculty of Arts and Humanities, University Paderborn, Paderborn, Germany
| | - Timo D. Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Oberschleissheim, Germany
| |
Collapse
|
16
|
Schuchardt JP, Kräter M, Schlögel M, Guck J, van Oirschot-Hermans BA, Bos J, van Wijk R, Tintle NL, Westra J, Kerlikowsky F, Hahn A, Harris WS. Omega-3 supplementation changes the physical properties of leukocytes but not erythrocytes in healthy individuals: An exploratory trial. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102636. [PMID: 39159530 DOI: 10.1016/j.plefa.2024.102636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
n3-PUFA impact health in several ways, including cardiovascular protection and anti-inflammatory effects, but the underlying mechanisms are not fully understood. In this exploratory study involving 31 healthy subjects, we aimed to investigate the effects of 12 weeks of fish-oil supplementation (1500 mg EPA+DHA/day) on the physical properties of multiple blood cell types. We used deformability cytometry (DC) for all cell types and Laser-assisted Optical Rotational Red Cell Analysis (Lorrca) to assess red blood cell (RBC) deformability. We also investigated the correlation between changes in the physical properties of blood cells and changes in the Omega-3 Index (O3I), defined as the relative content of EPA+DHA in RBCs. Following supplementation, the mean±SD O3I increased from 5.3 %±1.5 % to 8.3 %±1.4 % (p < 0.001). No significant changes in RBC properties were found by both techniques. However, by DC we observed a consistent pattern of physical changes in lymphocytes, neutrophils and monocytes. Among these were significant increases in metrics correlated with the cells' deformability resulting in less stiff cells. The results suggest that leukocytes become softer and have an increased ability to deform under induced short-term physical stress such as hydrodynamic force in the circulation. These changes could impact immune function since softer leukocytes can potentially circulate more easily and could facilitate a more rapid response to systemic inflammation or infection. In conclusion, fish-oil supplementation modulates some physical properties of leukocyte-subfractions, potentially enhancing their biological function. Further studies are warranted to explore the impact of n3-PUFA on blood cell biology, particularly in disease states associated with leukocyte dysregulation.
Collapse
Affiliation(s)
- Jan Philipp Schuchardt
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany; The Fatty Acid Research Institute, Sioux Falls, SD, USA.
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Maximilian Schlögel
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Brigitte A van Oirschot-Hermans
- Central Diagnostic Laboratory - Red Blood Cell Research Group, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jennifer Bos
- Central Diagnostic Laboratory - Red Blood Cell Research Group, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Richard van Wijk
- Central Diagnostic Laboratory - Red Blood Cell Research Group, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Nathan L Tintle
- The Fatty Acid Research Institute, Sioux Falls, SD, USA; Department of Population Health Nursing Science, College of Nursing, University of Illinois - Chicago, Chicago, IL, USA
| | - Jason Westra
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
| | - Felix Kerlikowsky
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - Andreas Hahn
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - William S Harris
- The Fatty Acid Research Institute, Sioux Falls, SD, USA; Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| |
Collapse
|
17
|
Piacentino ML, Fasse AJ, Camacho-Avila A, Grabylnikov I, Bronner ME. SMPD3 expression is spatially regulated in the developing embryo by SOXE factors. Dev Biol 2024; 506:31-41. [PMID: 38052296 PMCID: PMC10872304 DOI: 10.1016/j.ydbio.2023.11.011] [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: 08/10/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
During epithelial-to-mesenchymal transition (EMT), significant rearrangements occur in plasma membrane protein and lipid content that are important for membrane function and acquisition of cell motility. To gain insight into how neural crest cells regulate their lipid content at the transcriptional level during EMT, here we identify critical enhancer sequences that regulate the expression of SMPD3, a gene responsible for sphingomyelin hydrolysis to produce ceramide and necessary for neural crest EMT. We uncovered three enhancer regions within the first intron of the SMPD3 locus that drive reporter expression in distinct spatial and temporal domains, together collectively recapitulating the expression domains of endogenous SMPD3 within the ectodermal lineages. We further dissected one enhancer that is specifically active in the migrating neural crest. By mutating putative transcriptional input sites or knocking down upstream regulators, we find that the SOXE-family transcription factors SOX9 and SOX10 regulate the expression of SMPD3 in migrating neural crest cells. Further, ChIP-seq and nascent transcription analysis reveal that SOX10 directly regulates expression of an SMPD3 enhancer specific to migratory neural crest cells. Together these results shed light on how core components of developmental gene regulatory networks interact with metabolic effector genes to control changes in membrane lipid content.
Collapse
Affiliation(s)
- Michael L Piacentino
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Aria J Fasse
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Alexis Camacho-Avila
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Ilya Grabylnikov
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| |
Collapse
|
18
|
Nesbeth PDC, Ziegler TR, Tripathi AK, Dabeer S, Weiss D, Hao L, Smith MR, Jones DP, Maner-Smith KM, Tu CL, Chang W, Weitzmann MN, Alvarez JA. Linoleic acid blunts early osteoblast differentiation and impairs oxidative phosphorylation in vitro. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102617. [PMID: 38788347 PMCID: PMC11293625 DOI: 10.1016/j.plefa.2024.102617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Linoleic acid (LNA), an essential polyunsaturated fatty acid (PUFA), plays a crucial role in cellular functions. However, excessive intake of LNA, characteristic of Western diets, can have detrimental effects on cells and organs. Human observational studies have shown an inverse relationship between plasma LNA concentrations and bone mineral density. The mechanism by which LNA impairs the skeleton is unclear, and there is a paucity of research on the effects of LNA on bone-forming osteoblasts. METHODS The effect of LNA on osteoblast differentiation, cellular bioenergetics, and production of oxidized PUFA metabolites in vitro, was studied using primary mouse bone marrow stromal cells (BMSC) and MC3T3-E1 osteoblast precursors. RESULTS LNA treatment decreased alkaline phosphatase activity, an early marker of osteoblast differentiation, but had no effect on committed osteoblasts or on mineralization by differentiated osteoblasts. LNA suppressed osteoblast commitment by blunting the expression of Runx2 and Osterix, key transcription factors involved in osteoblast differentiation, and other key osteoblast-related factors involved in bone formation. LNA treatment was associated with increased production of oxidized LNA- and arachidonic acid-derived metabolites and blunted oxidative phosphorylation, resulting in decreased ATP production. CONCLUSION Our results show that LNA inhibited early differentiation of osteoblasts and this inhibitory effect was associated with increased production of oxidized PUFA metabolites that likely impaired energy production via oxidative phosphorylation.
Collapse
Affiliation(s)
- Paula-Dene C Nesbeth
- Nutrition and Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA, USA; Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Emory Center for Clinical and Molecular Nutrition, Emory University, Atlanta, GA, USA
| | - Thomas R Ziegler
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Emory Center for Clinical and Molecular Nutrition, Emory University, Atlanta, GA, USA
| | - Ashish Kumar Tripathi
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Sadaf Dabeer
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, USA
| | - Daiana Weiss
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Li Hao
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Matthew R Smith
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Emory Center for Clinical and Molecular Nutrition, Emory University, Atlanta, GA, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | | | - Chia-Ling Tu
- Endocrine Research Unit, San Francisco VA Healthcare System, University of California, San Francisco, CA, USA
| | - Wenhan Chang
- Endocrine Research Unit, San Francisco VA Healthcare System, University of California, San Francisco, CA, USA
| | - M Neale Weitzmann
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, USA
| | - Jessica A Alvarez
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Emory Center for Clinical and Molecular Nutrition, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Castello-Serrano I, Heberle FA, Diaz-Rohrer B, Ippolito R, Shurer CR, Lujan P, Campelo F, Levental KR, Levental I. Partitioning to ordered membrane domains regulates the kinetics of secretory traffic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.18.537395. [PMID: 37131599 PMCID: PMC10153169 DOI: 10.1101/2023.04.18.537395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The organelles of eukaryotic cells maintain distinct protein and lipid compositions required for their specific functions. The mechanisms by which many of these components are sorted to their specific locations remain unknown. While some motifs mediating subcellular protein localization have been identified, many membrane proteins and most membrane lipids lack known sorting determinants. A putative mechanism for sorting of membrane components is based on membrane domains known as lipid rafts, which are laterally segregated nanoscopic assemblies of specific lipids and proteins. To assess the role of such domains in the secretory pathway, we applied a robust tool for synchronized secretory protein traffic (RUSH, Retention Using Selective Hooks) to protein constructs with defined affinity for raft phases. These constructs consist solely of single-pass transmembrane domains (TMDs) and, lacking other sorting determinants, constitute probes for membrane domain-mediated trafficking. We find that while raft affinity can be sufficient for steady-state PM localization, it is not sufficient for rapid exit from the endoplasmic reticulum (ER), which is instead mediated by a short cytosolic peptide motif. In contrast, we find that Golgi exit kinetics are highly dependent on raft affinity, with raft preferring probes exiting Golgi ~2.5-fold faster than probes with minimal raft affinity. We rationalize these observations with a kinetic model of secretory trafficking, wherein Golgi export can be facilitated by protein association with raft domains. These observations support a role for raft-like membrane domains in the secretory pathway and establish an experimental paradigm for dissecting its underlying machinery.
Collapse
|
20
|
Wang HY, Dey S, Levental KR. Applications of phase-separating multi-bilayers in protein-membrane domain interactions. Methods Enzymol 2024; 700:275-294. [PMID: 38971603 DOI: 10.1016/bs.mie.2024.04.024] [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] [Indexed: 07/08/2024]
Abstract
Synthetic model membranes are important tools to elucidate lipid domain and protein interactions due to predefined lipid compositions and characterizable biophysical properties. Here, we introduce a model membrane with multiple lipid bilayers (multi-bilayers) stacked on a mica substrate that is prepared through a spin-coating technique. The spin-coated multi-bilayers are useful in the study of phase separated membranes with a high cholesterol content, mobile lipids, microscopic and reversible phase separation, and easy conjugation with proteins, which make them a good model to study interactions between proteins and membrane domains.
Collapse
Affiliation(s)
- Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, United States
| | - Simli Dey
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris, France
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
21
|
Benova A, Ferencakova M, Bardova K, Funda J, Prochazka J, Spoutil F, Cajka T, Dzubanova M, Balcaen T, Kerckhofs G, Willekens W, van Lenthe GH, Charyyeva A, Alquicer G, Pecinova A, Mracek T, Horakova O, Coupeau R, Hansen MS, Rossmeisl M, Kopecky J, Tencerova M. Omega-3 PUFAs prevent bone impairment and bone marrow adiposity in mouse model of obesity. Commun Biol 2023; 6:1043. [PMID: 37833362 PMCID: PMC10575870 DOI: 10.1038/s42003-023-05407-8] [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: 04/06/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Obesity adversely affects bone and fat metabolism in mice and humans. Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) have been shown to improve glucose metabolism and bone homeostasis in obesity. However, the impact of omega-3 PUFAs on bone marrow adipose tissue (BMAT) and bone marrow stromal cell (BMSC) metabolism has not been intensively studied yet. In the present study we demonstrated that omega-3 PUFA supplementation in high fat diet (HFD + F) improved bone parameters, mechanical properties along with decreased BMAT in obese mice when compared to the HFD group. Primary BMSCs isolated from HFD + F mice showed decreased adipocyte and higher osteoblast differentiation with lower senescent phenotype along with decreased osteoclast formation suggesting improved bone marrow microenvironment promoting bone formation in mice. Thus, our study highlights the beneficial effects of omega-3 PUFA-enriched diet on bone and cellular metabolism and its potential use in the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Michaela Ferencakova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Funda
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Tim Balcaen
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | - Arzuv Charyyeva
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Glenda Alquicer
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Pecinova
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Mracek
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Roman Coupeau
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Morten Svarer Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense, C DK-5000, Denmark
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
22
|
Morsczeck C, De Pellegrin M, Reck A, Reichert TE. Evaluation of Current Studies to Elucidate Processes in Dental Follicle Cells Driving Osteogenic Differentiation. Biomedicines 2023; 11:2787. [PMID: 37893160 PMCID: PMC10604663 DOI: 10.3390/biomedicines11102787] [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: 09/07/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
When research on osteogenic differentiation in dental follicle cells (DFCs) began, projects focused on bone morphogenetic protein (BMP) signaling. The BMP pathway induces the transcription factor DLX3, whichh in turn induces the BMP signaling pathway via a positive feedback mechanism. However, this BMP2/DLX3 signaling pathway only seems to support the early phase of osteogenic differentiation, since simultaneous induction of BMP2 or DLX3 does not further promote differentiation. Recent data showed that inhibition of classical protein kinase C (PKCs) supports the mineralization of DFCs and that osteogenic differentiation is sensitive to changes in signaling pathways, such as protein kinase B (PKB), also known as AKT. Small changes in the lipidome seem to confirm the participation of AKT and PKC in osteogenic differentiation. In addition, metabolic processes, such as fatty acid biosynthesis, oxidative phosphorylation, or glycolysis, are essential for the osteogenic differentiation of DFCs. This review article attempts not only to bring the various factors into a coherent picture of osteogenic differentiation in DFCs, but also to relate them to recent developments in other types of osteogenic progenitor cells.
Collapse
Affiliation(s)
- Christian Morsczeck
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany (A.R.); (T.E.R.)
| | | | | | | |
Collapse
|
23
|
Pinigin KV, Akimov SA. The Membrane-Mediated Interaction of Liquid-Ordered Lipid Domains in the Presence of Amphipathic Peptides. MEMBRANES 2023; 13:816. [PMID: 37887988 PMCID: PMC10608175 DOI: 10.3390/membranes13100816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023]
Abstract
The lipid membranes of living cells are composed of a large number of lipid types and can undergo phase separation with the formation of nanometer-scale liquid-ordered lipid domains, also called rafts. Raft coalescence, i.e., the fusion of lipid domains, is involved in important cell processes, such as signaling and trafficking. In this work, within the framework of the theory of elasticity of lipid membranes, we explore how amphipathic peptides adsorbed on lipid membranes may affect the domain-domain fusion processes. We show that the elastic deformations of lipid membranes drive amphipathic peptides to the boundary of lipid domains, which leads to an increase in the average energy barrier of the domain-domain fusion, even if the surface concentration of amphipathic peptides is low and the domain boundaries are only partially occupied by the peptides. This inhibition of the fusion of lipid domains may lead to negative side effects of using amphipathic peptides as antimicrobial agents.
Collapse
Affiliation(s)
- Konstantin V. Pinigin
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia
| |
Collapse
|
24
|
Wang HY, Chan SH, Dey S, Castello-Serrano I, Rosen MK, Ditlev JA, Levental KR, Levental I. Coupling of protein condensates to ordered lipid domains determines functional membrane organization. SCIENCE ADVANCES 2023; 9:eadf6205. [PMID: 37126554 PMCID: PMC10132753 DOI: 10.1126/sciadv.adf6205] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
During T cell activation, the transmembrane adaptor protein LAT (linker for activation of T cells) forms biomolecular condensates with Grb2 and Sos1, facilitating signaling. LAT has also been associated with cholesterol-rich condensed lipid domains; However, the potential coupling between protein condensation and lipid phase separation and its role in organizing T cell signaling were unknown. Here, we report that LAT/Grb2/Sos1 condensates reconstituted on model membranes can induce and template lipid domains, indicating strong coupling between lipid- and protein-based phase separation. Correspondingly, activation of T cells induces cytoplasmic protein condensates that associate with and stabilize raft-like membrane domains. Inversely, lipid domains nucleate and stabilize LAT protein condensates in both reconstituted and living systems. This coupling of lipid and protein assembly is functionally important, as uncoupling of lipid domains from cytoplasmic protein condensates abrogates T cell activation. Thus, thermodynamic coupling between protein condensates and ordered lipid domains regulates the functional organization of living membranes.
Collapse
Affiliation(s)
- Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Simli Dey
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Michael K Rosen
- Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathon A Ditlev
- Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Program in Molecular Medicine, Program in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
25
|
Diaz-Rohrer B, Castello-Serrano I, Chan SH, Wang HY, Shurer CR, Levental KR, Levental I. Rab3 mediates a pathway for endocytic sorting and plasma membrane recycling of ordered microdomains. Proc Natl Acad Sci U S A 2023; 120:e2207461120. [PMID: 36848577 PMCID: PMC10013782 DOI: 10.1073/pnas.2207461120] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 01/31/2023] [Indexed: 03/01/2023] Open
Abstract
The composition of the plasma membrane (PM) must be tightly controlled despite constant, rapid endocytosis, which requires active, selective recycling of endocytosed membrane components. For many proteins, the mechanisms, pathways, and determinants of this PM recycling remain unknown. We report that association with ordered, lipid-driven membrane microdomains (known as rafts) is sufficient for PM localization of a subset of transmembrane proteins and that abrogation of raft association disrupts their trafficking and leads to degradation in lysosomes. Using orthogonal, genetically encoded probes with tunable raft partitioning, we screened for the trafficking machinery required for efficient recycling of engineered microdomain-associated cargo from endosomes to the PM. Using this screen, we identified the Rab3 family as an important mediator of PM localization of microdomain-associated proteins. Disruption of Rab3 reduced PM localization of raft probes and led to their accumulation in Rab7-positive endosomes, suggesting inefficient recycling. Abrogation of Rab3 function also mislocalized the endogenous raft-associated protein Linker for Activation of T cells (LAT), leading to its intracellular accumulation and reduced T cell activation. These findings reveal a key role for lipid-driven microdomains in endocytic traffic and suggest Rab3 as a mediator of microdomain recycling and PM composition.
Collapse
Affiliation(s)
- Barbara Diaz-Rohrer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Carolyn R. Shurer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| |
Collapse
|
26
|
Jia W, Wu X, Shi L. Naturally forming benzoic acid orientates perilipin to facilitate glyceride-type polyunsaturated fatty acid degradation via fermentation behavior. J Dairy Sci 2023; 106:1650-1671. [PMID: 36710193 DOI: 10.3168/jds.2022-22381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/07/2022] [Indexed: 01/29/2023]
Abstract
Naturally forming benzoic acid in fermented dairy products accumulates in organisms and biomagnifies through collateral transport. The association between benzoic acid agglomeration and susceptible lipid nutrients remains obscure. Horizontal analysis of lipidomic alteration in response to benzoic acid was conducted and the spatially proteomic map was constructed using label-free quantitative proteomics. From synergistic integration of multi-omics in benzoic acid accumulated fermented goat milk model, the biological processes of significant proteins mostly focused on glyceride-type polyunsaturated fatty acids degradation (143.818 ± 0.51 mg/kg to 104.613 ± 0.29 mg/kg). As a physiological barrier shield, perilipin, which is coated on the surface of lipid droplets, protects triacylglycerols from cytosolic lipases, thus preventing triglyceride hydrolysis. The expression of perilipin decreased by 90% compared with the control group, leading to the decrease of triglycerides. Benzoic acid suppressed phosphatidylethanolamines and phosphatidylcholines synthesis by attenuating choline phosphotransferase and ethanolamine phosphotransferase. Less diglyceride generated by the dephosphorylation of phosphatidic acid entered choline phosphotransferase and ethanolamine phosphotransferase-mediated glycerophospholipid metabolisms. Fermentation of goat milk at a low temperature and less incubation time leads to the production of less benzoic acid and mitigation of lipid nutrient loss. The present study delineated the molecular landscape of fermented goat milk containing endogenous benzoic acid and further dissected the trajectory guiding lipid alteration to advance control of benzoic acid residue.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, 710021 China.
| | - Xixuan Wu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China
| | - Lin Shi
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China
| |
Collapse
|
27
|
Vamadeva SG, Patel K, Ravi Mangu S, Ellur G, Sukhdeo SV, Sharan K. Maternal omega-3 LC-PUFA supplementation programs an improved bone mass in the offspring with a more pronounced effect in females than males at adulthood. J Nutr Biochem 2023; 113:109245. [PMID: 36473540 DOI: 10.1016/j.jnutbio.2022.109245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 11/17/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Early balanced nutrition is vital in achieving optimal skeletal mass and its maintenance. Although a lower omega-6 (n-6): omega-3 (n-3) long-chain polyunsaturated fatty acid (LC-PUFA) ratio is strongly linked with bone health, its maternal effect in the programming of the offspring's skeleton remains to be elucidated. Plugged C57BL/6 mice were fed either n-3 LC-PUFA Enriched Diet (LED) or a control diet (C) throughout their gestation and lactation. Offspring born to both the groups were weaned onto C till 6, 12, and 24 weeks of their age. Offspring's skeleton metabolism and serum fatty acid composition was studied. In humans, seventy-five mother-female newborns pairs from term gestation were tested for their maternal LC-PUFA status relationships to venous cord blood bone biomarkers. Offspring of maternal LED supplemented mice exhibited a superior bone phenotype over C, more prominent in females than males. A lower serum n-6/n-3 LC-PUFA in the LED group offspring was strongly associated with blood biomarkers of bone metabolism. Sexual dimorphism evidenced had a strong correlation between offspring's LC-PUFA levels and bone turnover markers in serum. A higher potential for osteoblastic differentiation in both LED offspring genders and reduced osteoclastogenesis in females was cell-autonomous effect. The human cross-sectional study also showed a positive correlation between maternal n-3 PUFA and cord blood markers of bone formation in female newborns at birth. Maternal dietary n-6/ n-3 fat quality determines offspring's bone growth and development. Our data suggest that the skeleton of female offspring is likely to be more sensitive to this early exposure.
Collapse
Affiliation(s)
- Sowmya Giriyapura Vamadeva
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Kalpana Patel
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Svvs Ravi Mangu
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Govindraj Ellur
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Shinde Vijay Sukhdeo
- Department of Meat and Marine Sciences, CSIR- Central Food Technological Research Institute, Mysuru, Karnataka, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
28
|
Migdadi L, Sharar N, Jafar H, Telfah A, Hergenröder R, Wöhler C. Machine Learning in Automated Monitoring of Metabolic Changes Accompanying the Differentiation of Adipose-Tissue-Derived Human Mesenchymal Stem Cells Employing 1H- 1H TOCSY NMR. Metabolites 2023; 13:352. [PMID: 36984792 PMCID: PMC10055867 DOI: 10.3390/metabo13030352] [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: 01/20/2023] [Revised: 02/12/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
The ability to monitor the dynamics of stem cell differentiation is a major goal for understanding biochemical evolution pathways. Automating the process of metabolic profiling using 2D NMR helps us to understand the various differentiation behaviors of stem cells, and therefore sheds light on the cellular pathways of development, and enhances our understanding of best practices for in vitro differentiation to guide cellular therapies. In this work, the dynamic evolution of adipose-tissue-derived human Mesenchymal stem cells (AT-derived hMSCs) after fourteen days of cultivation, adipocyte and osteocyte differentiation, was inspected based on 1H-1H TOCSY using machine learning. Multi-class classification in addition to the novelty detection of metabolites was established based on a control hMSC sample after four days' cultivation and we successively detected the changes of metabolites in differentiated MSCs following a set of 1H-1H TOCSY experiments. The classifiers Kernel Null Foley-Sammon Transform and Kernel Density Estimation achieved a total classification error between 0% and 3.6% and false positive and false negative rates of 0%. This approach was successfully able to automatically reveal metabolic changes that accompanied MSC cellular evolution starting from their undifferentiated status to their prolonged cultivation and differentiation into adipocytes and osteocytes using machine learning supporting the research in the field of metabolic pathways of stem cell differentiation.
Collapse
Affiliation(s)
- Lubaba Migdadi
- Image Analysis Group, TU Dortmund, 44227 Dortmund, Germany
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
| | - Nour Sharar
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
- Cell Therapy Center, University of Jordan, Amman 11942, Jordan
| | - Hanan Jafar
- Cell Therapy Center, University of Jordan, Amman 11942, Jordan
- Department of Anatomy and Histology, College of Medicine, University of Jordan, Amman 11942, Jordan
| | - Ahmad Telfah
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
- Nanotechnology Center, The University of Jordan, Amman 11942, Jordan
| | - Roland Hergenröder
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
| | | |
Collapse
|
29
|
Beltran LC, Cvirkaite-Krupovic V, Miller J, Wang F, Kreutzberger MAB, Patkowski JB, Costa TRD, Schouten S, Levental I, Conticello VP, Egelman EH, Krupovic M. Archaeal DNA-import apparatus is homologous to bacterial conjugation machinery. Nat Commun 2023; 14:666. [PMID: 36750723 PMCID: PMC9905601 DOI: 10.1038/s41467-023-36349-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
Conjugation is a major mechanism of horizontal gene transfer promoting the spread of antibiotic resistance among human pathogens. It involves establishing a junction between a donor and a recipient cell via an extracellular appendage known as the mating pilus. In bacteria, the conjugation machinery is encoded by plasmids or transposons and typically mediates the transfer of cognate mobile genetic elements. Much less is known about conjugation in archaea. Here, we determine atomic structures by cryo-electron microscopy of three conjugative pili, two from hyperthermophilic archaea (Aeropyrum pernix and Pyrobaculum calidifontis) and one encoded by the Ti plasmid of the bacterium Agrobacterium tumefaciens, and show that the archaeal pili are homologous to bacterial mating pili. However, the archaeal conjugation machinery, known as Ced, has been 'domesticated', that is, the genes for the conjugation machinery are encoded on the chromosome rather than on mobile genetic elements, and mediates the transfer of cellular DNA.
Collapse
Affiliation(s)
- Leticia C Beltran
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Jessalyn Miller
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Biochemistry and Molecular Genetics, University of Alabama Birmingham, Birmingham, AL, 35233, USA
| | - Mark A B Kreutzberger
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jonasz B Patkowski
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Tiago R D Costa
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Stefan Schouten
- NIOZ Royal Netherlands Institute for Sea Research, Department of Marine Microbiology and Biogeochemistry, Texel, The Netherlands
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA.
| | - Mart Krupovic
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Archaeal Virology Unit, 75015, Paris, France.
| |
Collapse
|
30
|
Bermudez B, Ishii T, Wu YH, Carpenter RD, Sherk VD. Energy Balance and Bone Health: a Nutrient Availability Perspective. Curr Osteoporos Rep 2023; 21:77-84. [PMID: 36542294 DOI: 10.1007/s11914-022-00765-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Obesity is highly prevalent and is associated with bone fragility and fracture. The changing nutrient availability to bone in obesity is an important facet of bone health. The goal of this article is to summarize current knowledge on the effects of carbohydrate and dietary fat availability on bone, particularly in the context of other tissues. RECENT FINDINGS The skeleton is a primary site for fatty acid and glucose uptake. The trafficking of carbohydrates and fats into tissues changes with weight loss and periods of weight gain. Exercise acutely influences nutrient uptake into bone and may affect nutrient partitioning to bone. Bone cells secrete hormones that signal to the brain and other tissues information about its energetic state, which may alter whole-body nutrient trafficking. There is a critical need for studies to address the changes that metabolic perturbations have on nutrient availability in bone.
Collapse
Affiliation(s)
- Beatriz Bermudez
- Department of Mechanical Engineering, University of Colorado Denver, Denver, CO, USA
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Toru Ishii
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yuan-Haw Wu
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - R Dana Carpenter
- Department of Mechanical Engineering, University of Colorado Denver, Denver, CO, USA
| | - Vanessa D Sherk
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Division of Translational and Clinical Sciences, Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Levental I, Lyman E. Regulation of membrane protein structure and function by their lipid nano-environment. Nat Rev Mol Cell Biol 2023; 24:107-122. [PMID: 36056103 PMCID: PMC9892264 DOI: 10.1038/s41580-022-00524-4] [Citation(s) in RCA: 155] [Impact Index Per Article: 155.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 02/04/2023]
Abstract
Transmembrane proteins comprise ~30% of the mammalian proteome, mediating metabolism, signalling, transport and many other functions required for cellular life. The microenvironment of integral membrane proteins (IMPs) is intrinsically different from that of cytoplasmic proteins, with IMPs solvated by a compositionally and biophysically complex lipid matrix. These solvating lipids affect protein structure and function in a variety of ways, from stereospecific, high-affinity protein-lipid interactions to modulation by bulk membrane properties. Specific examples of functional modulation of IMPs by their solvating membranes have been reported for various transporters, channels and signal receptors; however, generalizable mechanistic principles governing IMP regulation by lipid environments are neither widely appreciated nor completely understood. Here, we review recent insights into the inter-relationships between complex lipidomes of mammalian membranes, the membrane physicochemical properties resulting from such lipid collectives, and the regulation of IMPs by either or both. The recent proliferation of high-resolution methods to study such lipid-protein interactions has led to generalizable insights, which we synthesize into a general framework termed the 'functional paralipidome' to understand the mutual regulation between membrane proteins and their surrounding lipid microenvironments.
Collapse
Affiliation(s)
- Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Molecular and Cell Physiology, University of Virginia, Charlottesville, VA, USA.
| | - Ed Lyman
- Department of Physics and Astronomy, Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA.
| |
Collapse
|
32
|
Danielli M, Perne L, Jarc Jovičić E, Petan T. Lipid droplets and polyunsaturated fatty acid trafficking: Balancing life and death. Front Cell Dev Biol 2023; 11:1104725. [PMID: 36776554 PMCID: PMC9911892 DOI: 10.3389/fcell.2023.1104725] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Lipid droplets are fat storage organelles ubiquitously distributed across the eukaryotic kingdom. They have a central role in regulating lipid metabolism and undergo a dynamic turnover of biogenesis and breakdown to meet cellular requirements for fatty acids, including polyunsaturated fatty acids. Polyunsaturated fatty acids esterified in membrane phospholipids define membrane fluidity and can be released by the activity of phospholipases A2 to act as ligands for nuclear receptors or to be metabolized into a wide spectrum of lipid signaling mediators. Polyunsaturated fatty acids in membrane phospholipids are also highly susceptible to lipid peroxidation, which if left uncontrolled leads to ferroptotic cell death. On the one hand, lipid droplets act as antioxidant organelles that control polyunsaturated fatty acid storage in triglycerides in order to reduce membrane lipid peroxidation, preserve organelle function and prevent cell death, including ferroptosis. On the other hand, lipid droplet breakdown fine-tunes the delivery of polyunsaturated fatty acids into metabolic and signaling pathways, but unrestricted lipid droplet breakdown may also lead to the release of lethal levels of polyunsaturated fatty acids. Precise regulation of lipid droplet turnover is thus essential for polyunsaturated fatty acid distribution and cellular homeostasis. In this review, we focus on emerging aspects of lipid droplet-mediated regulation of polyunsaturated fatty acid trafficking, including the management of membrane lipid peroxidation, ferroptosis and lipid mediator signaling.
Collapse
Affiliation(s)
| | | | | | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
33
|
Rose TD, Köhler N, Falk L, Klischat L, Lazareva OE, Pauling JK. Lipid network and moiety analysis for revealing enzymatic dysregulation and mechanistic alterations from lipidomics data. Brief Bioinform 2023; 24:bbac572. [PMID: 36592059 PMCID: PMC9851308 DOI: 10.1093/bib/bbac572] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 01/03/2023] Open
Abstract
Lipidomics is of growing importance for clinical and biomedical research due to many associations between lipid metabolism and diseases. The discovery of these associations is facilitated by improved lipid identification and quantification. Sophisticated computational methods are advantageous for interpreting such large-scale data for understanding metabolic processes and their underlying (patho)mechanisms. To generate hypothesis about these mechanisms, the combination of metabolic networks and graph algorithms is a powerful option to pinpoint molecular disease drivers and their interactions. Here we present lipid network explorer (LINEX$^2$), a lipid network analysis framework that fuels biological interpretation of alterations in lipid compositions. By integrating lipid-metabolic reactions from public databases, we generate dataset-specific lipid interaction networks. To aid interpretation of these networks, we present an enrichment graph algorithm that infers changes in enzymatic activity in the context of their multispecificity from lipidomics data. Our inference method successfully recovered the MBOAT7 enzyme from knock-out data. Furthermore, we mechanistically interpret lipidomic alterations of adipocytes in obesity by leveraging network enrichment and lipid moieties. We address the general lack of lipidomics data mining options to elucidate potential disease mechanisms and make lipidomics more clinically relevant.
Collapse
Affiliation(s)
- Tim D Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Nikolai Köhler
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Lisa Falk
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Lucie Klischat
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Olga E Lazareva
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Junior Clinical Cooperation Unit Multiparametric methods for early detection of prostate cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Josch K Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
34
|
Argentati C, Dominici F, Morena F, Rallini M, Tortorella I, Ferrandez-Montero A, Pellegrino RM, Ferrari B, Emiliani C, Lieblich M, Torre L, Martino S, Armentano I. Thermal treatment of magnesium particles in polylactic acid polymer films elicits the expression of osteogenic differentiation markers and lipidome profile remodeling in human adipose stem cells. Int J Biol Macromol 2022; 223:684-701. [PMID: 36356880 DOI: 10.1016/j.ijbiomac.2022.11.005] [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: 07/27/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
The efficacy of polylactic acid (PLA)/Magnesium (Mg)-based materials for driving stem cells toward bone tissue engineering applications requires specific Mg surface properties to modulate the interface of stem cells with the film. Here, we have developed novel PLA/Mg-based composites and explored their osteogenic differentiation potential on human adipose stem cells (hASCs). Mg-particles/polymer interface was improved by two treatments: heating in oxidative atmosphere (TT) and surface modification with a compatibilizer (PEI). Different contents of Mg particles were dispersed in PLA and composite surface and bulk properties, protein adsorption, stem cell-PLA/Mg interactions, osteogenic markers expressions, and lipids composition profile were evaluated. Mg particles were uniformly distributed on the surface and in the bulk PLA polymer. Improved and modulated particle-polymer adhesion was observed in Mg particle-treated composites. After 21 days in canonical growth culture conditions, hASCs on PLA/MgTT displayed the highest expression of the general osteogenic markers, RUNX2, SSP1, and BGLAP genes, Alkaline Phosphatase, type I Collagen, Osteopontin, and Calcium deposits. Moreover, by LC/MS QTOF mass-spectrophotometry lipidomic analysis, we found in PLA/MgTT-cells, for the first time, a remodeling of the lipid classes composition associated with the osteogenic differentiation. We ascribed these results to MgTT characteristics, which improve Mg availability and composite osteoinductive performance.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Franco Dominici
- Department of Civil and Environmental Engineering (DICA), University of Perugia, Strada di Pentima 4, 05100 Terni, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Marco Rallini
- Department of Civil and Environmental Engineering (DICA), University of Perugia, Strada di Pentima 4, 05100 Terni, Italy
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Ana Ferrandez-Montero
- Instituto de Cerámica y Vidrio, CSIC, Campus de Cantoblanco, c/ Kelsen 5, 28049 Madrid, Spain.
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Begoña Ferrari
- Instituto de Cerámica y Vidrio, CSIC, Campus de Cantoblanco, c/ Kelsen 5, 28049 Madrid, Spain.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy; CEMIN, University of Perugia, 06122 Perugia, Italy
| | - Marcela Lieblich
- Department Physical Metallurgy, National Centre for Metallurgical Research (CENIM), CSIC, Avenida Gregorio del Amo 8, Madrid 28040, Spain
| | - Luigi Torre
- Department of Civil and Environmental Engineering (DICA), University of Perugia, Strada di Pentima 4, 05100 Terni, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy; CEMIN, University of Perugia, 06122 Perugia, Italy.
| | - Ilaria Armentano
- Department of Economics, Engineering, Society and Business Organization (DEIM), University of Tuscia, Viterbo 01100, Italy.
| |
Collapse
|
35
|
Tao SS, Wang P, Wang XY, Yin KJ, Yang XK, Wang ZX, Wang DG, Pan HF. Causal effect of polyunsaturated fatty acids on bone mineral density and fracture. Front Nutr 2022; 9:1014847. [PMID: 36570136 PMCID: PMC9772990 DOI: 10.3389/fnut.2022.1014847] [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] [Received: 08/09/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Background Polyunsaturated fatty acids (PUFAs) are closely related to osteoporosis. To test their causal relationship, we conducted a Mendelian randomization (MR) analysis. Methods We analyzed the causal relationship between four PUFAs measures, n-3 PUFAs (n-3), n-6 PUFAs (n-6), the ratio of n-3 PUFAs to total fatty acids (n-3 pct), and the ratio of n-6 PUFAs to n-3 PUFAs (n-6 to n-3), and five measures of osteoporosis, including estimated bone mineral density (eBMD), forearm (FA) BMD, femoral neck (FN) BMD, lumbar spine (LS) BMD, and fracture, using two-sample MR analysis. In order to verify the direct effect between PUFAs and BMD, we chose interleukin-6 (IL-6), tumor necrosis factor-β (TNF-β), and bone morphogenetic proteins 7 (BMP-7), three markers or cytokines strongly related to BMD, as possible confounding factors, and analyzed the possible causal relationships between them and PUFAs or BMD by MR. Inverse variance weighting (IVW), MR-Egger, weighted and weighted median were conducted. MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) and MR-Egger regression methods were used to evaluate the potential pleiotropy of instrumental variables (IVs) and outliers were identified by MR-PRESSO. Cochran's Q statistic was used to detect the heterogeneity among IVs. Leave-one-out sensitivity analysis was used to find SNPs that have a significant impact on the results. All results were corrected by the Bonferroni correction. Results The IVW results showed that n-3 PUFAs (OR = 1.030, 95% CI: 1.013, 1.047, P = 0.001) and n-6 PUFAs (OR = 1.053, 95% CI: 1.034, 1.072, P < 0.001) were positively correlated with eBMD, while n-6 to n-3 (OR = 0.947, 95% CI: 0.924, 0.970, P < 0.001) were negatively correlated with eBMD. These casual relationships still existed after Bonferroni correction. There were positive effects of n-3 PUFAs on FA BMD (OR = 1.090, 95% CI: 1.011, 1.176, P = 0.025) and LS BMD (OR = 1.056, 95% CI: 1.011, 1.104, P = 0.014), n-3 pct on eBMD (OR = 1.028, 95% CI: 1.002, 1.055, P = 0.035) and FA BMD (OR = 1.090, 95% CI: 1.011, 1.174, P = 0.025), n-6 to n-3 on LS BMD (OR = 1.071, 95% CI: 1.021, 1.124, P = 0.005); negative effects of n-3 pct on fracture (OR = 0.953, 95% CI: 0.918, 0.988, P = 0.009) and n-6 to n-3 on FA BMD (OR = 0.910, 95% CI: 0.837, 0.988, P = 0.025). However, these causal effects all disappeared after Bonferroni correction (all P > 0.0025). None of IL-6, TNF-β, and BMP-7 had a causal effect on PUFA and BMD simultaneously (all P > 0.05). Conclusion Evidence from this MR study supports the genetically predicted causal effects of n-3, n-6, n-3 pct, and n-6 to n-3 on eBMD. In addition, n-3 not only associate with FA BMD and LS BMD through its own level and n-6 to n-3, but also link to fracture through n-3 pct.
Collapse
Affiliation(s)
- Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China,Experimental Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Peng Wang
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xin-Yi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,The First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Xin Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De-Guang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,*Correspondence: De-Guang Wang,
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China,Hai-Feng Pan, ,
| |
Collapse
|
36
|
Bezerra AF, Alves JPM, Fernandes CCL, Cavalcanti CM, Silva MRL, Conde AJH, Tetaping GM, Ferreira ACA, Melo LM, Rodrigues APR, Rondina D. Dyslipidemia induced by lipid diet in late gestation donor impact on growth kinetics and in vitro potential differentiation of umbilical cord Wharton's Jelly mesenchymal stem cells in goats. Vet Res Commun 2022; 46:1259-1270. [PMID: 36125693 DOI: 10.1007/s11259-022-09995-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Mesenchymal stem cells (MSC) from the umbilical cord (UC) have several attractive properties for clinical use. This study aimed to verify the impact of a lipid-rich diet during late gestation of donor goats on the growth and differentiation of MSCs from UC. From the 100th day of pregnancy to delivery, 22 goats were grouped based on their diet into the donor-lipid (DLD; n = 11) and donor-baseline (DBD; n = 11) diet groups. Diets were isonitrogenous and isoenergetic, differing in fat content (2.8% vs. 6.3% on a dry matter basis). Wharton's jelly (WJ) fragments were cultured. After primary culture, samples of WJ-MSCs were characterized by the expression of CD90, CD73, CD34, CD45, CD105, and Fas genes, mitochondrial activity using MitoTracker (MT) fluorescence probe, and growth kinetics. Population doubling time (PDT) was also determined. WJ-MSCs were differentiated into chondrocytes, adipocytes and osteocytes, and the mineralized area and adipocytes were determined. The lipid diet significantly increased triglyceride and cholesterol levels during pregnancy. The DLD group showed sub-expression of the CD90 gene, a high MT intensity, and a low proliferation rate at the end of the subculture. The mean PDT was 83.9 ± 1.3 h. Mineralized area and lipid droplet stain intensity from osteogenic and adipogenic differentiations, respectively, were greater in DLD. We conclude that in donor goats, dietary dyslipidemia during late pregnancy affects the ability of UC-derived MSCs to express their developmental potential in vitro, thus limiting their possible use for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | - Camila Muniz Cavalcanti
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil
| | | | | | - Gildas Mbemya Tetaping
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil
| | | | - Luciana Magalhães Melo
- School of Veterinary Medicine, Centro Universitario Fametro (UNIFAMETRO), Fortaleza, CE, 60010-470, Brazil
| | | | - Davide Rondina
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil.
| |
Collapse
|
37
|
Bispo DSC, Jesus CSH, Romek K, Marques IMC, Oliveira MB, Mano JF, Gil AM. An Intracellular Metabolic Signature as a Potential Donor-Independent Marker of the Osteogenic Differentiation of Adipose Tissue Mesenchymal Stem Cells. Cells 2022; 11:cells11233745. [PMID: 36497004 PMCID: PMC9739047 DOI: 10.3390/cells11233745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
This paper describes an untargeted NMR metabolomics study to identify potential intracellular donor-dependent and donor-independent metabolic markers of proliferation and osteogenic differentiation of human adipose mesenchymal stem cells (hAMSCs). The hAMSCs of two donors with distinct proliferating/osteogenic characteristics were fully characterized regarding their polar endometabolome during proliferation and osteogenesis. An 18-metabolites signature (including changes in alanine, aspartate, proline, tyrosine, ATP, and ADP, among others) was suggested to be potentially descriptive of cell proliferation, independently of the donor. In addition, a set of 11 metabolites was proposed to compose a possible donor-independent signature of osteogenesis, mostly involving changes in taurine, glutathione, methylguanidine, adenosine, inosine, uridine, and creatine/phosphocreatine, choline/phosphocholine and ethanolamine/phosphocholine ratios. The proposed signatures were validated for a third donor, although they require further validation in a larger donor cohort. We believe that this proof of concept paves the way to exploit metabolic markers to monitor (and potentially predict) cell proliferation and the osteogenic ability of different donors.
Collapse
|
38
|
Ugwuodo CJ, Colosimo F, Adhikari J, Shen Y, Badireddy AR, Mouser PJ. Salinity and hydraulic retention time induce membrane phospholipid acyl chain remodeling in Halanaerobium congolense WG10 and mixed cultures from hydraulically fractured shale wells. Front Microbiol 2022; 13:1023575. [PMID: 36439785 PMCID: PMC9687094 DOI: 10.3389/fmicb.2022.1023575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2023] Open
Abstract
Bacteria remodel their plasma membrane lipidome to maintain key biophysical attributes in response to ecological disturbances. For Halanaerobium and other anaerobic halotolerant taxa that persist in hydraulically fractured deep subsurface shale reservoirs, salinity, and hydraulic retention time (HRT) are important perturbants of cell membrane structure, yet their effects remain poorly understood. Membrane-linked activities underlie in situ microbial growth kinetics and physiologies which drive biogeochemical reactions in engineered subsurface systems. Hence, we used gas chromatography-mass spectrometry (GC-MS) to investigate the effects of salinity and HRT on the phospholipid fatty acid composition of H. congolense WG10 and mixed enrichment cultures from hydraulically fractured shale wells. We also coupled acyl chain remodeling to membrane mechanics by measuring bilayer elasticity using atomic force microscopy (AFM). For these experiments, cultures were grown in a chemostat vessel operated in continuous flow mode under strict anoxia and constant stirring. Our findings show that salinity and HRT induce significant changes in membrane fatty acid chemistry of H. congolense WG10 in distinct and complementary ways. Notably, under nonoptimal salt concentrations (7% and 20% NaCl), H. congolense WG10 elevates the portion of polyunsaturated fatty acids (PUFAs) in its membrane, and this results in an apparent increase in fluidity (homeoviscous adaptation principle) and thickness. Double bond index (DBI) and mean chain length (MCL) were used as proxies for membrane fluidity and thickness, respectively. These results provide new insight into our understanding of how environmental and engineered factors might disrupt the physical and biogeochemical equilibria of fractured shale by inducing physiologically relevant changes in the membrane fatty acid chemistry of persistent microbial taxa. GRAPHICAL ABSTRACTSalinity significantly alters membrane bilayer fluidity and thickness in Halanaerobium congolense WG10.
Collapse
Affiliation(s)
- Chika Jude Ugwuodo
- Natural Resources and Earth Systems Science, University of New Hampshire, Durham, NH, United States
- Department of Civil and Environmental Engineering, University of New Hampshire, Durham, NH, United States
| | | | - Jishnu Adhikari
- Sanborn, Head and Associates, Inc., Concord, NH, United States
| | - Yuxiang Shen
- Department of Civil and Environmental Engineering, University of Vermont, Burlington, VT, United States
| | - Appala Raju Badireddy
- Department of Civil and Environmental Engineering, University of Vermont, Burlington, VT, United States
| | - Paula J. Mouser
- Department of Civil and Environmental Engineering, University of New Hampshire, Durham, NH, United States
| |
Collapse
|
39
|
Li X, Zhou S, Lin X. Molecular View on the Impact of DHA Molecules on the Physical Properties of a Model Cell Membrane. J Chem Inf Model 2022; 62:2421-2431. [PMID: 35513897 DOI: 10.1021/acs.jcim.2c00074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Docosahexaenoic acid (DHA) is a ω-3 polyunsaturated fatty acid, which can be uptaken by cells and is essential for proper neuronal and retinal function. However, the detailed physical impact of DHA molecules on the plasma membrane is still unclear. Hence, in this work, we carried out μs-scale coarse-grained molecular dynamics (MD) simulations to reveal the interactions between DHA molecules and a model cell membrane. As is known, the cell membrane can segregate into liquid-ordered (Lo) and liquid-disordered (Ld) membrane domains due to the differential interactions between lipids and proteins. In order to capture this feature, we adopted the three-component phase-separated lipid membranes and considered both anionic and neutral DHA molecules in the current work. Our results showed that DHA molecules can spontaneously self-assemble into nanoclusters, fuse with lipid membranes, and localize preferably in Ld membrane domains. During the membrane fusion process, DHA molecules can change the intrinsic transmembrane potential of the lipid membrane, and the effects of anionic DHA molecules are much more significant. Besides, the presence of DHA molecules mainly in the Ld membrane domains could regulate the differences in the lipid chain order, membrane thickness, cholesterol preference, and cholesterol flip-flop basically in a concentration-dependent manner, which further promote the stability of the intraleaflet dynamics and inhibit the interleaflet dynamics (or promote membrane domain registration) of the membrane domains. In short, the impact of DHA molecules on the physical properties of a model cell membrane on the molecular level revealed in our work will provide useful insights for understanding the biological functions of DHA molecules.
Collapse
Affiliation(s)
- Xiu Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shiying Zhou
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
40
|
Endo- and Exometabolome Crosstalk in Mesenchymal Stem Cells Undergoing Osteogenic Differentiation. Cells 2022; 11:cells11081257. [PMID: 35455937 PMCID: PMC9024772 DOI: 10.3390/cells11081257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
This paper describes, for the first time to our knowledge, a lipidome and exometabolome characterization of osteogenic differentiation for human adipose tissue stem cells (hAMSCs) using nuclear magnetic resonance (NMR) spectroscopy. The holistic nature of NMR enabled the time-course evolution of cholesterol, mono- and polyunsaturated fatty acids (including ω-6 and ω-3 fatty acids), several phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins, and plasmalogens), and mono- and triglycerides to be followed. Lipid changes occurred almost exclusively between days 1 and 7, followed by a tendency for lipidome stabilization after day 7. On average, phospholipids and longer and more unsaturated fatty acids increased up to day 7, probably related to plasma membrane fluidity. Articulation of lipidome changes with previously reported polar endometabolome profiling and with exometabolome changes reported here in the same cells, enabled important correlations to be established during hAMSC osteogenic differentiation. Our results supported hypotheses related to the dynamics of membrane remodelling, anti-oxidative mechanisms, protein synthesis, and energy metabolism. Importantly, the observation of specific up-taken or excreted metabolites paves the way for the identification of potential osteoinductive metabolites useful for optimized osteogenic protocols.
Collapse
|
41
|
Sezgin E. Giant plasma membrane vesicles to study plasma membrane structure and dynamics. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183857. [PMID: 34990591 DOI: 10.1016/j.bbamem.2021.183857] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
The plasma membrane (PM) is a highly heterogenous structure intertwined with the cortical actin cytoskeleton and extracellular matrix. This complex architecture makes it difficult to study the processes taking place at the PM. Model membrane systems that are simple mimics of the PM overcome this bottleneck and allow us to study the biophysical principles underlying the processes at the PM. Among them, cell-derived giant plasma membrane vesicles (GPMVs) are considered the most physiologically relevant system, retaining the compositional complexity of the PM to a large extent. GPMVs have become a key tool in membrane research in the last few years. In this review, I will provide a brief overview of this system, summarize recent applications and discuss the limitations.
Collapse
Affiliation(s)
- Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden.
| |
Collapse
|
42
|
Yang X, Li J, Zhao L, Chen Y, Cui Z, Xu T, Li X, Wu S, Zhang Y. Targeting adipocytic discoidin domain receptor 2 impedes fat gain while increasing bone mass. Cell Death Differ 2022; 29:737-749. [PMID: 34645939 PMCID: PMC8990016 DOI: 10.1038/s41418-021-00887-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity is closely associated with low-bone-mass disorder. Discoidin domain receptor 2 (DDR2) plays essential roles in skeletal metabolism, and is probably involved in fat metabolism. To test the potential role of DDR2 in fat and fat-bone crosstalk, Ddr2 conditional knockout mice (Ddr2Adipo) were generated in which Ddr2 gene is exclusively deleted in adipocytes by Adipoq Cre. We found that Ddr2Adipo mice are protected from fat gain on high-fat diet, with significantly decreased adipocyte size. Ddr2Adipo mice exhibit significantly increased bone mass and mechanical properties, with enhanced osteoblastogenesis and osteoclastogenesis. Marrow adipocyte is diminished in the bone marrow of Ddr2Adipo mice, due to activation of lipolysis. Fatty acid in the bone marrow was reduced in Ddr2Adipo mice. RNA-Seq analysis identified adenylate cyclase 5 (Adcy5) as downstream molecule of Ddr2. Mechanically, adipocytic Ddr2 modulates Adcy5-cAMP-PKA signaling, and Ddr2 deficiency stimulates lipolysis and supplies fatty acid for oxidation in osteoblasts, leading to the enhanced osteoblast differentiation and bone mass. Treatment of Adcy5 specific inhibitor abolishes the increased bone mass gain in Ddr2Adipo mice. These observations establish, for the first time, that Ddr2 plays an essential role in the crosstalk between fat and bone. Targeting adipocytic Ddr2 may be a potential strategy for treating obesity and pathological bone loss simultaneously.
Collapse
Affiliation(s)
- Xiaoyu Yang
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China ,grid.452842.d0000 0004 8512 7544The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China ,grid.207374.50000 0001 2189 3846School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jing Li
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Liting Zhao
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yazhuo Chen
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Zhijun Cui
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China ,grid.47840.3f0000 0001 2181 7878Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA USA
| | - Taotao Xu
- grid.417400.60000 0004 1799 0055The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Xu Li
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shufang Wu
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yan Zhang
- grid.452438.c0000 0004 1760 8119Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
43
|
Bispo DC, Jesus CSH, Correia M, Ferreira F, Bonifazio G, Goodfellow BJ, Oliveira MB, Mano JF, Gil AM. NMR Metabolomics Assessment of Osteogenic Differentiation of Adipose-Tissue-Derived Mesenchymal Stem Cells. J Proteome Res 2022; 21:654-670. [PMID: 35061379 PMCID: PMC9776527 DOI: 10.1021/acs.jproteome.1c00832] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This Article presents, for the first time to our knowledge, an untargeted nuclear magnetic resonance (NMR) metabolomic characterization of the polar intracellular metabolic adaptations of human adipose-derived mesenchymal stem cells during osteogenic differentiation. The use of mesenchymal stem cells (MSCs) for bone regeneration is a promising alternative to conventional bone grafts, and untargeted metabolomics may unveil novel metabolic information on the osteogenic differentiation of MSCs, allowing their behavior to be understood and monitored/guided toward effective therapies. Our results unveiled statistically relevant changes in the levels of just over 30 identified metabolites, illustrating a highly dynamic process with significant variations throughout the whole 21-day period of osteogenic differentiation, mainly involving amino acid metabolism and protein synthesis; energy metabolism and the roles of glycolysis, the tricarboxylic acid cycle, and oxidative phosphorylation; cell membrane metabolism; nucleotide metabolism (including the specific involvement of O-glycosylation intermediates and NAD+); and metabolic players in protective antioxidative mechanisms (such as glutathione and specific amino acids). Different metabolic stages are proposed and are supported by putative biochemical explanations for the metabolite changes observed. This work lays the groundwork for the use of untargeted NMR metabolomics to find potential metabolic markers of osteogenic differentiation efficacy.
Collapse
Affiliation(s)
- Daniela
S. C. Bispo
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Catarina S. H. Jesus
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Marlene Correia
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Filipa Ferreira
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Giulia Bonifazio
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal,Department
of Biotechnology Lazzaro Spallanzani, University
of Pavia, Corso Str.
Nuova, 65, 27100 Pavia PV, Italy
| | - Brian J. Goodfellow
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Mariana B. Oliveira
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - João F. Mano
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal
| | - Ana M. Gil
- Department
of Chemistry, CICECO - Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitario de Santiago, 3810-193 Aveiro, Portugal,
| |
Collapse
|
44
|
Cai M, Liu Y, Tian Y, Liang Y, Xu Z, Liu F, Lai R, Zhou Z, Liu M, Dai J, Liu X. Osteogenic peptides in periodontal ligament stem cell-containing three-dimensional bioscaffolds promote bone healing. Biomater Sci 2022; 10:1765-1775. [PMID: 35212326 DOI: 10.1039/d1bm01673c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bone tissue engineering shows great potential in bone regeneration; however, the lack of bone growth factors with high biocompatibility and efficiency is a major concern. Oligopeptides have drawn great attention due to their high biological efficacy, low toxicity, and low molecular weight. The oligopeptide SDSSD promotes the osteogenesis of human periodontal ligament stem cells (hPDLSCs) in vitro. The SDSSD-modified three-dimensional (3D) bioscaffolds promote osteogenesis and bone formation in the subcutaneous pockets of BALB/c nude mice and facilitate bone healing in vivo. Mechanistically, SDSSD promoted bone formation by binding to G protein-coupled receptors and regulating the AKT signaling pathway. 3D-printing bioscaffolds with SDSSD may be potential bone tissue engineering materials for treating bone defects.
Collapse
Affiliation(s)
- Mingxiang Cai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Yaoyao Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Yinping Tian
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445099, China
| | - Yan Liang
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Zinan Xu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Fangchen Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Renfa Lai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Zhiying Zhou
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Minyi Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Jian Dai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
45
|
Li Y, Liu J, Zhou H, Liu J, Xue X, Wang L, Ren S. Liquid chromatography-mass spectrometry method for discovering the metabolic markers to reveal the potential therapeutic effects of naringin on osteoporosis. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1194:123170. [DOI: 10.1016/j.jchromb.2022.123170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 02/02/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
|
46
|
Gao X, Hong G, Zhan W, Liu T, Yan S, Deng M, Tu C, Li P. DPA promotes hBMSCs osteogenic differentiation by miR-9-5p/ERK/ALP signaling pathway. Int J Med Sci 2022; 19:1879-1887. [PMID: 36438924 PMCID: PMC9682506 DOI: 10.7150/ijms.77729] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023] Open
Abstract
Docosahexaenoic acid (DHA) has been reported potentiate osteogenic differentiation, while Docosapentaenoic acid (DPA), another Omega-3 fatty acid, its contribution to the osteogenic differentiation of human bone-marrow-derived mesenchymal stromal cells (hBMSCs) is not entirely elucidated. The Alizarin Red S (ARS) staining and the expression of osteogenesis‑associated genes were analyzed during osteogenic induction by DPA. Then, bioinformatics analysis and dual luciferase reporter assays were investigated to confirm the interactions between miR-9-5p and alkaline phosphatase (ALP). miR-9-5p mimics / inhibitor were transfected to human hBMSCs and the osteogenic assay above was also performed. Furthermore, DPA significantly promoted the phosphorylation of ERK via miR-9-5p. PD98059, a highly specific and potent ERK1/2 inhibitor, inhibited the activation of ALP and partially reversed the role of DPA during osteogenic differentiation. These data indicated that DPA promoted osteogenic differentiation of hBMSCs potentially through miR-9-5p/ERK/ALP signaling pathway, providing a potentially useful therapeutic strategy for patients to improve bone loss.
Collapse
Affiliation(s)
- Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Guanhao Hong
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.,Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Weiqiang Zhan
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.,Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Tianfeng Liu
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Shouquan Yan
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.,Scientific Research Department, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Mingzhu Deng
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.,Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chenlin Tu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.,Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Peng Li
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
47
|
Westra M, Gutierrez Y, MacGillavry HD. Contribution of Membrane Lipids to Postsynaptic Protein Organization. Front Synaptic Neurosci 2021; 13:790773. [PMID: 34887741 PMCID: PMC8649999 DOI: 10.3389/fnsyn.2021.790773] [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: 10/07/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
The precise subsynaptic organization of proteins at the postsynaptic membrane controls synaptic transmission. In particular, postsynaptic receptor complexes are concentrated in distinct membrane nanodomains to optimize synaptic signaling. However, despite the clear functional relevance of subsynaptic receptor organization to synaptic transmission and plasticity, the mechanisms that underlie the nanoscale organization of the postsynaptic membrane remain elusive. Over the last decades, the field has predominantly focused on the role of protein-protein interactions in receptor trafficking and positioning in the synaptic membrane. In contrast, the contribution of lipids, the principal constituents of the membrane, to receptor positioning at the synapse remains poorly understood. Nevertheless, there is compelling evidence that the synaptic membrane is enriched in specific lipid species and that deregulation of lipid homeostasis in neurons severely affects synaptic functioning. In this review we focus on how lipids are organized at the synaptic membrane, with special emphasis on how current models of membrane organization could contribute to protein distribution at the synapse and synaptic transmission. Finally, we will present an outlook on how novel technical developments could be applied to study the dynamic interplay between lipids and proteins at the postsynaptic membrane.
Collapse
Affiliation(s)
- Manon Westra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Yolanda Gutierrez
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Harold D MacGillavry
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
48
|
Carravilla P, Dasgupta A, Zhurgenbayeva G, Danylchuk DI, Klymchenko AS, Sezgin E, Eggeling C. Long-term STED imaging of membrane packing and dynamics by exchangeable polarity-sensitive dyes. BIOPHYSICAL REPORTS 2021; 1:None. [PMID: 34939048 PMCID: PMC8651516 DOI: 10.1016/j.bpr.2021.100023] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/08/2021] [Indexed: 12/28/2022]
Abstract
Understanding the plasma membrane nanoscale organization and dynamics in living cells requires microscopy techniques with high spatial and temporal resolution that permit for long acquisition times and allow for the quantification of membrane biophysical properties, such as lipid ordering. Among the most popular super-resolution techniques, stimulated emission depletion (STED) microscopy offers one of the highest temporal resolutions, ultimately defined by the scanning speed. However, monitoring live processes using STED microscopy is significantly limited by photobleaching, which recently has been circumvented by exchangeable membrane dyes that only temporarily reside in the membrane. Here, we show that NR4A, a polarity-sensitive exchangeable plasma membrane probe based on Nile red, permits the super-resolved quantification of membrane biophysical parameters in real time with high temporal and spatial resolution as well as long acquisition times. The potential of this polarity-sensitive exchangeable dye is showcased by live-cell real-time three-dimensional STED recordings of bleb formation and lipid exchange during membrane fusion as well as by STED-fluorescence correlation spectroscopy experiments for the simultaneous quantification of membrane dynamics and lipid packing that correlate in model and live-cell membranes.
Collapse
Affiliation(s)
- Pablo Carravilla
- Leibniz Institute of Photonic Technology e.V., Jena, Germany
- Faculty of Physics and Astronomy, Institute of Applied Optics and Biophysics, Friedrich Schiller University Jena, Jena, Germany
| | - Anindita Dasgupta
- Leibniz Institute of Photonic Technology e.V., Jena, Germany
- Faculty of Physics and Astronomy, Institute of Applied Optics and Biophysics, Friedrich Schiller University Jena, Jena, Germany
| | - Gaukhar Zhurgenbayeva
- Faculty of Physics and Astronomy, Institute of Applied Optics and Biophysics, Friedrich Schiller University Jena, Jena, Germany
- Jena School for Microbial Communication, Friedrich Schiller University Jena, Jena, Germany
| | - Dmytro I. Danylchuk
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | - Andrey S. Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Christian Eggeling
- Leibniz Institute of Photonic Technology e.V., Jena, Germany
- Faculty of Physics and Astronomy, Institute of Applied Optics and Biophysics, Friedrich Schiller University Jena, Jena, Germany
- Jena School for Microbial Communication, Friedrich Schiller University Jena, Jena, Germany
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Jena Center for Soft Matter, Jena, Germany
| |
Collapse
|
49
|
Agostini F, Vicinanza C, Biolo G, Spessotto P, Da Ros F, Lombardi E, Durante C, Mazzucato M. Nucleofection of Adipose Mesenchymal Stem/Stromal Cells: Improved Transfection Efficiency for GMP Grade Applications. Cells 2021; 10:cells10123412. [PMID: 34943920 PMCID: PMC8700287 DOI: 10.3390/cells10123412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022] Open
Abstract
Nucleofection (NF) is a safe, non-viral transfection method, compatible with Good Manufacturing Practice guidelines. Such a technique is useful to improve therapeutic effectiveness of adipose tissue mesenchymal stem cells (ASC) in clinical settings, but improvement of NF efficiency is mandatory. Supernatant rich in growth factors (SRGF) is a clinical-grade medium additive for ASC expansion. We showed a dramatically increased NF efficiency and post-transfection viability in ASC expanded in presence of SRGF (vs. fetal bovine serum). SRGF expanded ASC were characterized by increased vesicle endocytosis but lower phagocytosis properties. SRGF increased n-6/n-3 ratio, reduced membrane lipid raft occurrence, and lowered intracellular actin content in ASC. A statistical correlation between NF efficiency and lipid raft availability on cell membranes was shown, even though a direct relationship could not be demonstrated: attempts to selectively modulate lipid rafts levels were, in fact, limited by technical constraints. In conclusion, we reported for the first time that tuning clinical-grade compatible cell culture conditions can significantly improve ASC transfection efficiency by a non-viral and safe approach. A deep mechanistic characterization is extremely complex, but we can hypothesize that integrated changes in membrane structure and intracellular actin content could contribute to explain SRGF impact on ASC NF efficiency.
Collapse
Affiliation(s)
- Francesco Agostini
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
- Correspondence: ; Tel.: +39-0434-659095
| | - Carla Vicinanza
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
| | - Gianni Biolo
- Unit of Internal Medicine, Clinica Medica, Department of Medical Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34100 Trieste, Italy;
| | - Paola Spessotto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy;
| | - Francesco Da Ros
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
| | - Elisabetta Lombardi
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
| | - Cristina Durante
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
| | - Mario Mazzucato
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy; (C.V.); (F.D.R.); (E.L.); (C.D.); (M.M.)
| |
Collapse
|
50
|
Matias MI, Yong CS, Foroushani A, Goldsmith C, Mongellaz C, Sezgin E, Levental KR, Talebi A, Perrault J, Rivière A, Dehairs J, Delos O, Bertand-Michel J, Portais JC, Wong M, Marie JC, Kelekar A, Kinet S, Zimmermann VS, Levental I, Yvan-Charvet L, Swinnen JV, Muljo SA, Hernandez-Vargas H, Tardito S, Taylor N, Dardalhon V. Regulatory T cell differentiation is controlled by αKG-induced alterations in mitochondrial metabolism and lipid homeostasis. Cell Rep 2021; 37:109911. [PMID: 34731632 PMCID: PMC10167917 DOI: 10.1016/j.celrep.2021.109911] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 08/18/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Suppressive regulatory T cell (Treg) differentiation is controlled by diverse immunometabolic signaling pathways and intracellular metabolites. Here we show that cell-permeable α-ketoglutarate (αKG) alters the DNA methylation profile of naive CD4 T cells activated under Treg polarizing conditions, markedly attenuating FoxP3+ Treg differentiation and increasing inflammatory cytokines. Adoptive transfer of these T cells into tumor-bearing mice results in enhanced tumor infiltration, decreased FoxP3 expression, and delayed tumor growth. Mechanistically, αKG leads to an energetic state that is reprogrammed toward a mitochondrial metabolism, with increased oxidative phosphorylation and expression of mitochondrial complex enzymes. Furthermore, carbons from ectopic αKG are directly utilized in the generation of fatty acids, associated with lipidome remodeling and increased triacylglyceride stores. Notably, inhibition of either mitochondrial complex II or DGAT2-mediated triacylglyceride synthesis restores Treg differentiation and decreases the αKG-induced inflammatory phenotype. Thus, we identify a crosstalk between αKG, mitochondrial metabolism and triacylglyceride synthesis that controls Treg fate.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Diacylglycerol O-Acyltransferase/metabolism
- Energy Metabolism/drug effects
- Fibrosarcoma/genetics
- Fibrosarcoma/immunology
- Fibrosarcoma/metabolism
- Fibrosarcoma/therapy
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Homeostasis
- Humans
- Immunotherapy, Adoptive
- Ketoglutaric Acids/pharmacology
- Lipid Metabolism/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/drug effects
- Mitochondria/genetics
- Mitochondria/metabolism
- Phenotype
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Signal Transduction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Mice
Collapse
Affiliation(s)
- Maria I Matias
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Carmen S Yong
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Amir Foroushani
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Chloe Goldsmith
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Cédric Mongellaz
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Julie Perrault
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Anais Rivière
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Océane Delos
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France; I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Justine Bertand-Michel
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France; I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Jean-Charles Portais
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Madeline Wong
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Julien C Marie
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Ameeta Kelekar
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | | | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Stefan A Muljo
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Saverio Tardito
- Cancer Research UK, Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA.
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France.
| |
Collapse
|