1
|
Gurjar S, Bhat A R, Upadhya R, Shenoy RP. Extracellular vesicle-mediated approaches for the diagnosis and therapy of MASLD: current advances and future prospective. Lipids Health Dis 2025; 24:5. [PMID: 39773634 PMCID: PMC11705780 DOI: 10.1186/s12944-024-02396-3] [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: 09/11/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an asymptomatic, multifaceted condition often associated with various risk factors, including fatigue, obesity, insulin resistance, metabolic syndrome, and sleep apnea. The increasing burden of MASLD underscores the critical need for early diagnosis and effective therapies. Owing to the lack of efficient therapies for MASLD, early diagnosis is crucial. Consequently, noninvasive biomarkers and imaging techniques are essential for analyzing disease risk and play a pivotal role in the global diagnostic process. The use of extracellular vesicles has emerged as promising for early diagnosis and therapy of various liver ailments. Herein, a comprehensive summary of the current diagnostic modalities for MASLD is presented, highlighting their advantages and limitations while exploring the potential of extracellular vesicles (EVs) as innovative diagnostic and therapeutic tools for MASLD. With this aim, this review emphasizes an in-depth understanding of the origin of EVs and the pathophysiological alterations of these ectosomes and exosomes in various liver diseases. This review also explores the therapeutic potential of EVs as key components in the future management of liver disease. The dual role of EVs as biomarkers and their therapeutic utility in MASLD essentially highlights their clinical integration to improve MASLD diagnosis and treatment. While EV-based therapies are still in their early stages of development and require substantial research to increase their therapeutic value before they can be used clinically, the diagnostic application of EVs has been extensively explored. Moving forward, developing diagnostic devices leveraging EVs will be crucial in advancing MASLD diagnosis. Thus, the literature summarized provides suitable grounds for clinicians and researchers to explore EVs for devising diagnostic and treatment strategies for MASLD.
Collapse
Affiliation(s)
- Swasthika Gurjar
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India
| | - Ramanarayana Bhat A
- Manipal Centre for Biotherapeutics Research, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India
| | - Raghavendra Upadhya
- Manipal Centre for Biotherapeutics Research, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India.
| | - Revathi P Shenoy
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India.
| |
Collapse
|
2
|
Łapińska Z, Rembiałkowska N, Szewczyk A, Przystupski D, Drąg-Zalesińska M, Novickij V, Saczko J, Kulbacka J, Baczyńska D. The additive effect of 17β-estradiol on the modulation of electrochemotherapy with calcium ions or cisplatin in human clear carcinoma cells. Biomed Pharmacother 2024; 181:117708. [PMID: 39608316 DOI: 10.1016/j.biopha.2024.117708] [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: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024] Open
Abstract
Calcium electroporation (CaEP) is an efficient approach for ovarian cancer treatment. It causes cell death by introducing elevated levels of calcium into cells. In this work, the research focused on two types of cell lines: CHO-K1, representing normal ovary cells, and OvBH-1, representing ovarian clear carcinoma cells. Those cell lines exhibited distinct reactions to calcium electroporation (CaEP). Also, we have evaluated the effects of 17β-estradiol following CaEP and electrochemotherapy (ECT) with cisplatin (CPP). The combination of ECT with CPP and CaEP with prior E2 preincubation resulted in approximately 23.55 % and 39 % decreases in cell survival compared to the control cells (exposed to CPP and CaCl2 alone) for ovarian cancer cells. The obtained results showed that ovarian cancer cells preincubated with 17β-estradiol after exposure to pulsed electric fields undergo primary necrosis. Additionally, preincubation of ovarian cancer cells with 17β-estradiol can significantly improve the effectiveness of both chemotherapy and electrochemotherapy involving cisplatin and calcium chloride.
Collapse
Affiliation(s)
- Zofia Łapińska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland.
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius LT-08406, Lithuania
| | - Dawid Przystupski
- Department of Pediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, Wroclaw 50-556, Poland
| | - Małgorzata Drąg-Zalesińska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Vitalij Novickij
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius LT-08406, Lithuania; Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius LT-10105, Lithuania
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius LT-08406, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| |
Collapse
|
3
|
Shabanpour Y, Hajipour-Verdom B, Abdolmaleki P, Alipour M. Protein-free domains in native and ferroptosis-driven oxidized cell membranes: a molecular dynamics study of biophysical properties and doxorubicin uptake. Front Mol Biosci 2024; 11:1494257. [PMID: 39611002 PMCID: PMC11602475 DOI: 10.3389/fmolb.2024.1494257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Ferroptosis is a regulated form of cell death characterized by iron-dependent lipid peroxidation of polyunsaturated fatty acids (PUFAs). Despite its significance, the precise molecular mechanisms underlying ferroptosis remain elusive, particularly concerning their impact on membrane properties. This study aimed to investigate the biophysical changes in plasma membranes due to lipid peroxidation during ferroptosis and their impact on the uptake of doxorubicin (DOX), a potent anticancer agent linked to ferroptosis. Using all-atom molecular dynamics simulations, we compared native red blood cell membranes (protein-free domains) with a ferroptosis model, in which PUFAs were replaced with hydroperoxide derivatives. Our findings reveal that the ferroptotic membrane exhibits decreased thickness and increased lipid area while maintaining overall integrity. The hydroperoxide groups localized in the disordered tail regions, enhancing tail mobility and facilitating hydrogen bonding. Lipid lateral diffusion was significantly altered, both layers of the ferroptotic membrane exhibited slower diffusion rates compared to the native membrane. Furthermore, lipid oxidation affected diffusion activation energies. Importantly, we found that DOX could penetrate the oxidized ferroptosis membrane with a lower free-energy barrier (∆GPB) of approximately 38 kJ.mol-1. Consequently, DOX's permeability was approximately seven orders of magnitude higher than that of the native membrane. In summary, lipid peroxidation during ferroptosis induces extensive structural and dynamic changes, influencing membrane behavior and potentially offering insights that could inform future therapeutic strategies.
Collapse
Affiliation(s)
- Yaser Shabanpour
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behnam Hajipour-Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mozhgan Alipour
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
5
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
6
|
Mavuduru VA, Vadupu L, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial phospholipid transport: Role of contact sites and lipid transport proteins. Prog Lipid Res 2024; 94:101268. [PMID: 38195013 DOI: 10.1016/j.plipres.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/11/2024]
Abstract
One of the major constituents of mitochondrial membranes is the phospholipids, which play a key role in maintaining the structure and the functions of the mitochondria. However, mitochondria do not synthesize most of the phospholipids in situ, necessitating the presence of phospholipid import pathways. Even for the phospholipids, which are synthesized within the inner mitochondrial membrane (IMM), the phospholipid precursors must be imported from outside the mitochondria. Therefore, the mitochondria heavily rely on the phospholipid transport pathways for its proper functioning. Since, mitochondria are not part of a vesicular trafficking network, the molecular mechanisms of how mitochondria receive its phospholipids remain a relevant question. One of the major ways that hydrophobic phospholipids can cross the aqueous barrier of inter or intraorganellar spaces is by apposing membranes, thereby decreasing the distance of transport, or by being sequestered by lipid transport proteins (LTPs). Therefore, with the discovery of LTPs and membrane contact sites (MCSs), we are beginning to understand the molecular mechanisms of phospholipid transport pathways in the mitochondria. In this review, we will present a brief overview of the recent findings on the molecular architecture and the importance of the MCSs, both the intraorganellar and interorganellar contact sites, in facilitating the mitochondrial phospholipid transport. In addition, we will also discuss the role of LTPs for trafficking phospholipids through the intermembrane space (IMS) of the mitochondria. Mechanistic insights into different phospholipid transport pathways of mitochondria could be exploited to vary the composition of membrane phospholipids and gain a better understanding of their precise role in membrane homeostasis and mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Vijay Aditya Mavuduru
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Lavanya Vadupu
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522502, India
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore; Department of Clinical Neuroscience, Karolinska Institute, Stockholm 17176, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore.
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India.
| |
Collapse
|
7
|
Sarmento MJ, Llorente A, Petan T, Khnykin D, Popa I, Nikolac Perkovic M, Konjevod M, Jaganjac M. The expanding organelle lipidomes: current knowledge and challenges. Cell Mol Life Sci 2023; 80:237. [PMID: 37530856 PMCID: PMC10397142 DOI: 10.1007/s00018-023-04889-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
Lipids in cell membranes and subcellular compartments play essential roles in numerous cellular processes, such as energy production, cell signaling and inflammation. A specific organelle lipidome is characterized by lipid synthesis and metabolism, intracellular trafficking, and lipid homeostasis in the organelle. Over the years, considerable effort has been directed to the identification of the lipid fingerprints of cellular organelles. However, these fingerprints are not fully characterized due to the large variety and structural complexity of lipids and the great variability in the abundance of different lipid species. The process becomes even more challenging when considering that the lipidome differs in health and disease contexts. This review summarizes the information available on the lipid composition of mammalian cell organelles, particularly the lipidome of the nucleus, mitochondrion, endoplasmic reticulum, Golgi apparatus, plasma membrane and organelles in the endocytic pathway. The lipid compositions of extracellular vesicles and lamellar bodies are also described. In addition, several examples of subcellular lipidome dynamics under physiological and pathological conditions are presented. Finally, challenges in mapping organelle lipidomes are discussed.
Collapse
Affiliation(s)
- Maria J Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167, Oslo, Norway
- Faculty of Medicine, Centre for Cancer Cell Reprogramming, University of Oslo, Montebello, 0379, Oslo, Norway
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Denis Khnykin
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Iuliana Popa
- Pharmacy Department, Bâtiment Henri Moissan, University Paris-Saclay, 17 Avenue des Sciences, 91400, Orsay, France
| | | | - Marcela Konjevod
- Division of Molecular Medicine, Ruder Boskovic Institute, 10000, Zagreb, Croatia
| | - Morana Jaganjac
- Division of Molecular Medicine, Ruder Boskovic Institute, 10000, Zagreb, Croatia.
| |
Collapse
|
8
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
9
|
Distinguishing Plasmin-Generating Microvesicles: Tiny Messengers Involved in Fibrinolysis and Proteolysis. Int J Mol Sci 2023; 24:ijms24021571. [PMID: 36675082 PMCID: PMC9860915 DOI: 10.3390/ijms24021571] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
A number of stressors and inflammatory mediators (cytokines, proteases, oxidative stress mediators) released during inflammation or ischemia stimulate and activate cells in blood, the vessel wall or tissues. The most well-known functional and phenotypic responses of activated cells are (1) the immediate expression and/or release of stored or newly synthesized bioactive molecules, and (2) membrane blebbing followed by release of microvesicles. An ultimate response, namely the formation of extracellular traps by neutrophils (NETs), is outside the scope of this work. The main objective of this article is to provide an overview on the mechanism of plasminogen reception and activation at the surface of cell-derived microvesicles, new actors in fibrinolysis and proteolysis. The role of microvesicle-bound plasmin in pathological settings involving inflammation, atherosclerosis, angiogenesis, and tumour growth, remains to be investigated. Further studies are necessary to determine if profibrinolytic microvesicles are involved in a finely regulated equilibrium with pro-coagulant microvesicles, which ensures a balanced haemostasis, leading to the maintenance of vascular patency.
Collapse
|
10
|
Ramos-Martín F, Herrera-León C, D'Amelio N. Bombyx mori Cecropin D could trigger cancer cell apoptosis by interacting with mitochondrial cardiolipin. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184003. [PMID: 35850261 DOI: 10.1016/j.bbamem.2022.184003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
Cecropin D is an antimicrobial peptide from Bombyx mori displaying anticancer and pro-apoptotic activities and, together with Cecropin XJ and Cecropin A, one of the very few peptides targeting esophageal cancer. Cecropin D displays poor similarity to other cecropins but a remarkable similarity in the structure and activity spectrum with Cecropin A and Cecropin XJ, offering the possibility to highlight key motifs at the base of the biological activity. In this work we show by NMR and MD simulations that Cecropin D is partially structured in solution and stabilizes its two-helix folding upon interaction with biomimetic membranes. Simulations show that Cecropin D strongly interacts with the surface of cancer cell biomimetic bilayers where it recognises the phosphatidylserine headgroup often exposed in the outer leaflet of cancerous cells by means of specific salt bridges. Cecropin D is also able to penetrate deeply in bilayers containing cardiolipin, a phospholipid found in mitochondria, causing significant destabilization in the lipid packing which might account for its pro-apoptotic activity. In bacterial membranes, phosphatidylglycerol and phosphatidylethanolamine act synergically by electrostatically attracting cecropin D and providing access to the membrane core, respectively.
Collapse
Affiliation(s)
- Francisco Ramos-Martín
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France.
| | - Claudia Herrera-León
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
| | - Nicola D'Amelio
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France.
| |
Collapse
|
11
|
Shaik GM, Draberova L, Cernohouzova S, Tumova M, Bugajev V, Draber P. Pentacyclic triterpenoid ursolic acid interferes with mast cell activation via a lipid-centric mechanism affecting FcεRI signalosome functions. J Biol Chem 2022; 298:102497. [PMID: 36115460 PMCID: PMC9587013 DOI: 10.1016/j.jbc.2022.102497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022] Open
Abstract
Pentacyclic triterpenoids, including ursolic acid (UA), are bioactive compounds with multiple biological activities involving anti-inflammatory effects. However, the mode of their action on mast cells, key players in the early stages of allergic inflammation, and underlying molecular mechanisms remain enigmatic. To better understand the effect of UA on mast cell signaling, here we examined the consequences of short-term treatment of mouse bone marrow-derived mast cells with UA. Using IgE-sensitized and antigen- or thapsigargin-activated cells, we found that 15 min exposure to UA inhibited high affinity IgE receptor (FcεRI)–mediated degranulation, calcium response, and extracellular calcium uptake. We also found that UA inhibited migration of mouse bone marrow-derived mast cells toward antigen but not toward prostaglandin E2 and stem cell factor. Compared to control antigen-activated cells, UA enhanced the production of tumor necrosis factor-α at the mRNA and protein levels. However, secretion of this cytokine was inhibited. Further analysis showed that UA enhanced tyrosine phosphorylation of the SYK kinase and several other proteins involved in the early stages of FcεRI signaling, even in the absence of antigen activation, but inhibited or reduced their further phosphorylation at later stages. In addition, we show that UA induced changes in the properties of detergent-resistant plasma membrane microdomains and reduced antibody-mediated clustering of the FcεRI and glycosylphosphatidylinositol-anchored protein Thy-1. Finally, UA inhibited mobility of the FcεRI and cholesterol. These combined data suggest that UA exerts its effects, at least in part, via lipid-centric plasma membrane perturbations, hence affecting the functions of the FcεRI signalosome.
Collapse
Affiliation(s)
- Gouse M Shaik
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Sara Cernohouzova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Magda Tumova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Viktor Bugajev
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
12
|
Himbert S, Rheinstädter MC. Structural and mechanical properties of the red blood cell's cytoplasmic membrane seen through the lens of biophysics. Front Physiol 2022; 13:953257. [PMID: 36171967 PMCID: PMC9510598 DOI: 10.3389/fphys.2022.953257] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/15/2022] [Indexed: 11/27/2022] Open
Abstract
Red blood cells (RBCs) are the most abundant cell type in the human body and critical suppliers of oxygen. The cells are characterized by a simple structure with no internal organelles. Their two-layered outer shell is composed of a cytoplasmic membrane (RBC cm ) tethered to a spectrin cytoskeleton allowing the cell to be both flexible yet resistant against shear stress. These mechanical properties are intrinsically linked to the molecular composition and organization of their shell. The cytoplasmic membrane is expected to dominate the elastic behavior on small, nanometer length scales, which are most relevant for cellular processes that take place between the fibrils of the cytoskeleton. Several pathologies have been linked to structural and compositional changes within the RBC cm and the cell's mechanical properties. We review current findings in terms of RBC lipidomics, lipid organization and elastic properties with a focus on biophysical techniques, such as X-ray and neutron scattering, and Molecular Dynamics simulations, and their biological relevance. In our current understanding, the RBC cm 's structure is patchy, with nanometer sized liquid ordered and disordered lipid, and peptide domains. At the same time, it is surprisingly soft, with bending rigidities κ of 2-4 kBT. This is in strong contrast to the current belief that a high concentration of cholesterol results in stiff membranes. This extreme softness is likely the result of an interaction between polyunsaturated lipids and cholesterol, which may also occur in other biological membranes. There is strong evidence in the literature that there is no length scale dependence of κ of whole RBCs.
Collapse
Affiliation(s)
- Sebastian Himbert
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| | - Maikel C. Rheinstädter
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
13
|
Gabrielle PH. Lipid metabolism and retinal diseases. Acta Ophthalmol 2022; 100 Suppl 269:3-43. [PMID: 36117363 DOI: 10.1111/aos.15226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/24/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The retina has enormous lipids demands and must meet those needs. Retinal lipid homeostasis appears to be based on the symbiosis between neurons, Müller glial cells (MGC), and retinal pigment epithelium (RPE) cells, which can be impacted in several retinal diseases. The current research challenge is to better understand lipid-related mechanisms involved in retinal diseases, such as age-related macular degeneration (AMD) and glaucoma. RESULTS In a first axis, in vitro and focus on Müller glial cell, we aimed to characterize whether the 24S-hydroxycholesterol (24S-OHC), an overexpressed end-product of cholesterol elimination pathway in neural tissue and likely produced by suffering retinal ganglion cells in glaucoma, may modulate MGC membrane organization, such as lipid rafts, to trigger cellular signalling pathways related to retinal gliosis. We have found that lipid composition appears to be a key factor of membrane architecture, especially for lipid raft microdomain formation, in MGC. However, 24S-OHC did not appear to trigger retinal gliosis via the modulation of lipid or protein composition within lipid rafts microdomains. This study provided a better understanding of the complex mechanisms involved in the pathophysiology of glaucoma. On a second clinical ax, we focused on the lipid-related mechanisms involved in the dysfunction of aging RPE and the appearance of drusenoid deposits in AMD. Using the Montrachet population-based study, we intend to report the frequency of reticular pseudodrusen (RPD) and its ocular and systemic risk factors, particularly related to lipid metabolisms, such as plasma lipoprotein levels, carotenoids levels, and lipid-lowering drug intake. Our study showed that RPD was less common in subjects taking lipid-lowering drugs. Lipid-lowering drugs, such as statins, may reduce the risk of RPD through their effect on the production and function of lipoproteins. This observation highlights the potential role of retinal lipid trafficking via lipoproteins between photoreceptors and retinal pigment epithelium cells in RPD formation. Those findings have been complemented with preliminary results on the analysis of plasma fatty acid (FA) profile, a surrogate marker of short-term dietary lipid intake, according to the type of predominant drusenoid deposit, soft drusen or RPD, in age-related maculopathy. CONCLUSION Further research on lipid metabolism in retinal diseases is warranted to better understand the pathophysiology of retinal diseases and develop new promising diagnostic, prognostic, and therapeutic tools for our patients.
Collapse
Affiliation(s)
- Pierre-Henry Gabrielle
- Eye and Nutrition Research Group, Center for Taste and Feeding Behaviour, AgroSup Dijon, CNRS, INRAe, The University Bourgogne Franche-Comté, Dijon, France.,Department of Ophthalmology, Dijon University Hospital, Dijon, France.,The Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Ramos-Martín F, D'Amelio N. Biomembrane lipids: When physics and chemistry join to shape biological activity. Biochimie 2022; 203:118-138. [PMID: 35926681 DOI: 10.1016/j.biochi.2022.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022]
Abstract
Biomembranes constitute the first lines of defense of cells. While small molecules can often permeate cell walls in bacteria and plants, they are generally unable to penetrate the barrier constituted by the double layer of phospholipids, unless specific receptors or channels are present. Antimicrobial or cell-penetrating peptides are in fact highly specialized molecules able to bypass this barrier and even discriminate among different cell types. This capacity is made possible by the intrinsic properties of its phospholipids, their distribution between the internal and external leaflet, and their ability to mutually interact, modulating the membrane fluidity and the exposition of key headgroups. Although common phospholipids can be found in the membranes of most organisms, some are characteristic of specific cell types. Here, we review the properties of the most common lipids and describe how they interact with each other in biomembrane. We then discuss how their assembly in bilayers determines some key physical-chemical properties such as permeability, potential and phase status. Finally, we describe how the exposition of specific phospholipids determines the recognition of cell types by membrane-targeting molecules.
Collapse
Affiliation(s)
- Francisco Ramos-Martín
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens, 80039, France.
| | - Nicola D'Amelio
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens, 80039, France.
| |
Collapse
|
15
|
Lipid Specific Membrane Interaction of Aptamers and Cytotoxicity. MEMBRANES 2021; 12:membranes12010037. [PMID: 35054563 PMCID: PMC8780203 DOI: 10.3390/membranes12010037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 11/17/2022]
Abstract
We aim to discover diagnostic tools to detect phosphatidylserine (PS) externalization on apoptotic cell surface using PS binding aptamers, AAAGAC and TAAAGA, and hence to understand chemotherapy drug efficacy when inducing apoptosis into cancer cells. The entropic fragment-based approach designed aptamers have been investigated to inspect three aspects: lipid specificity in aptamers' membrane binding and bilayer physical properties-induced regulation of binding mechanisms, the apoptosis-induced cancer cell surface binding of aptamers, and the aptamer-induced cytotoxicity. The liposome binding assays show preferred membrane binding of aptamers due to presence of PS in predominantly phosphatidylcholine-contained liposomes. Two membrane stiffness reducing amphiphiles triton X-100 and capsaicin were found to enhance membrane's aptamer adsorption suggesting that bilayer physical properties influence membrane's adsorption of drugs. Microscopic images of fluorescence-tagged aptamer treated LoVo cells show strong fluorescence intensity only if apoptosis is induced. Aptamers find enhanced PS molecules to bind with on the surface of apoptotic over nonapoptotic cells. In cytotoxicity experiments, TAAAGA (over poor PS binding aptamer CAGAAAAAAAC) was found cytotoxic towards RBL cells due to perhaps binding with nonapoptotic externalized PS randomly and thus slowly breaching plasma membrane integrity. In these three experimental investigations, we found aptamers to act on membranes at comparable concentrations and specifically with PS binding manner. Earlier, we reported the origins of actions through molecular mechanism studies-aptamers interact with lipids using mainly charge-based interactions. Lipids and aptamers hold distinguishable charge properties, and hence, lipid-aptamer association follows distinguishable energetics due to electrostatic and van der Waals interactions. We discover that our PS binding aptamers, due to lipid-specific interactions, appear as diagnostic tools capable of detecting drug-induced apoptosis in cancer cells.
Collapse
|
16
|
Acoba MG, Senoo N, Claypool SM. Phospholipid ebb and flow makes mitochondria go. J Cell Biol 2021; 219:151918. [PMID: 32614384 PMCID: PMC7401802 DOI: 10.1083/jcb.202003131] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 01/19/2023] Open
Abstract
Mitochondria, so much more than just being energy factories, also have the capacity to synthesize macromolecules including phospholipids, particularly cardiolipin (CL) and phosphatidylethanolamine (PE). Phospholipids are vital constituents of mitochondrial membranes, impacting the plethora of functions performed by this organelle. Hence, the orchestrated movement of phospholipids to and from the mitochondrion is essential for cellular integrity. In this review, we capture recent advances in the field of mitochondrial phospholipid biosynthesis and trafficking, highlighting the significance of interorganellar communication, intramitochondrial contact sites, and lipid transfer proteins in maintaining membrane homeostasis. We then discuss the physiological functions of CL and PE, specifically how they associate with protein complexes in mitochondrial membranes to support bioenergetics and maintain mitochondrial architecture.
Collapse
Affiliation(s)
- Michelle Grace Acoba
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nanami Senoo
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
17
|
Production of erythrocyte microparticles in a sub-hemolytic environment. J Artif Organs 2021; 24:135-145. [PMID: 33420875 DOI: 10.1007/s10047-020-01231-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/25/2020] [Indexed: 01/14/2023]
Abstract
Microparticles are produced by various cells due to a number of different stimuli in the circulatory system. Shear stress has been shown to injure red blood cells resulting in hemolysis or non-reversible sub-hemolytic damage. We hypothesized that, in the sub-hemolytic shear range, there exist sufficient mechanical stimuli for red blood cells to respond with production of microparticles. Red blood cells isolated from blood of healthy volunteers were exposed to high shear stress in a microfluidic channel to mimic mechanical trauma similar to that occurring in ventricular assist devices. Utilizing flow cytometry techniques, both an increase of shear rate and exposure time showed higher concentrations of red blood cell microparticles. Controlled shear rate exposure shows that red blood cell microparticle concentration may be indicative of sub-hemolytic damage to red blood cells. In addition, properties of these red blood cell microparticles produced by shear suggest that mechanical trauma may underlie some complications for cardiovascular patients.
Collapse
|
18
|
Comparative lipidomic analysis of mammalian retinal ganglion cells and Müller glia in situ and in vitro using High-Resolution Imaging Mass Spectrometry. Sci Rep 2020; 10:20053. [PMID: 33208898 PMCID: PMC7674471 DOI: 10.1038/s41598-020-77087-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/03/2020] [Indexed: 01/02/2023] Open
Abstract
In order to better understand retinal physiology, alterations to which underlie some ocular diseases, we set out to establish the lipid signature of two fundamental cell types in the retina, Müller Glia and Retinal Ganglion Cells (RGCs). Moreover, we compared the lipid signature of these cells in sections (in situ), as well as after culturing the cells and isolating their cell membranes (in vitro). The lipidome of Müller glia and RGCs was analyzed in porcine retinal sections using Matrix Assisted Laser Desorption Ionization Imaging Mass Spectrometry (MALDI-IMS). Isolated membranes, as well as whole cells from primary cell cultures of RGCs and Müller glia, were printed onto glass slides using a non-contact microarrayer (Nano Plotter), and a LTQ-Orbitrap XL analyzer was used to scan the samples in negative ion mode, thereafter identifying the RGCs and Müller cells immunohistochemically. The spectra acquired were aligned and normalized against the total ion current, and a statistical analysis was carried out to select the lipids specific to each cell type in the retinal sections and microarrays. The peaks of interest were identified by MS/MS analysis. A cluster analysis of the MS spectra obtained from the retinal sections identified regions containing RGCs and Müller glia, as confirmed by immunohistochemistry in the same sections. The relative density of certain lipids differed significantly (p-value ≤ 0.05) between the areas containing Müller glia and RGCs. Likewise, different densities of lipids were evident between the RGC and Müller glia cultures in vitro. Finally, a comparative analysis of the lipid profiles in the retinal sections and microarrays identified six peaks that corresponded to a collection of 10 lipids characteristic of retinal cells. These lipids were identified by MS/MS. The analyses performed on the RGC layer of the retina, on RGCs in culture and using cell membrane microarrays of RGCs indicate that the lipid composition of the retina detected in sections is preserved in primary cell cultures. Specific lipid species were found in RGCs and Müller glia, allowing both cell types to be identified by a lipid fingerprint. Further studies into these specific lipids and of their behavior in pathological conditions may well help identify novel therapeutic targets for ocular diseases.
Collapse
|
19
|
Osman A, Benameur T, Korashy HM, Zeidan A, Agouni A. Interplay between Endoplasmic Reticulum Stress and Large Extracellular Vesicles (Microparticles) in Endothelial Cell Dysfunction. Biomedicines 2020; 8:E409. [PMID: 33053883 PMCID: PMC7599704 DOI: 10.3390/biomedicines8100409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/26/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
Upon increased demand for protein synthesis, accumulation of misfolded and/or unfolded proteins within the endoplasmic reticulum (ER), a pro-survival response is activated termed unfolded protein response (UPR), aiming at restoring the proper function of the ER. Prolonged activation of the UPR leads, however, to ER stress, a cellular state that contributes to the pathogenesis of various chronic diseases including obesity and diabetes. ER stress response by itself can result in endothelial dysfunction, a hallmark of cardiovascular disease, through various cellular mechanisms including apoptosis, insulin resistance, inflammation and oxidative stress. Extracellular vesicles (EVs), particularly large EVs (lEVs) commonly referred to as microparticles (MPs), are membrane vesicles. They are considered as a fingerprint of their originating cells, carrying a variety of molecular components of their parent cells. lEVs are emerging as major contributors to endothelial cell dysfunction in various metabolic disease conditions. However, the mechanisms underpinning the role of lEVs in endothelial dysfunction are not fully elucidated. Recently, ER stress emerged as a bridging molecular link between lEVs and endothelial cell dysfunction. Therefore, in the current review, we summarized the roles of lEVs and ER stress in endothelial dysfunction and discussed the molecular crosstalk and relationship between ER stress and lEVs in endothelial dysfunction.
Collapse
Affiliation(s)
- Aisha Osman
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, P.O. Box 400, Al Ahsa 31982, Saudi Arabia;
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| | - Asad Zeidan
- Department of Basic Medical Sciences, College of Medicine, QU health, Qatar University, Doha 2713, Qatar;
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| |
Collapse
|
20
|
Ahmed A, Sarwar S, Hu Y, Munir MU, Nisar MF, Ikram F, Asif A, Rahman SU, Chaudhry AA, Rehman IU. Surface-modified polymeric nanoparticles for drug delivery to cancer cells. Expert Opin Drug Deliv 2020; 18:1-24. [PMID: 32905714 DOI: 10.1080/17425247.2020.1822321] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The utilization of polymeric nanoparticles, as drug payloads, has been extensively prevailed in cancer therapy. However, the precise distribution of these nanocarriers is restrained by various physiological and cellular obstacles. Nanoparticles must avoid nonspecific interactions with healthy cells and in vivo compartments to circumvent these barriers. Since in vivo interactions of nanoparticles are mainly dependent on surface properties of nanoparticles, efficient control on surface constituents is necessary for the determination of nanoparticles' fate in the body. AREAS COVERED In this review, the surface-modified polymeric nanoparticles and their utilization in cancer treatment were elaborated. First, the interaction of nanoparticles with numerous in vivo barriers was highlighted. Second, different strategies to overcome these obstacles were described. Third, some inspiring examples of surface-modified nanoparticles were presented. Later, fabrication and characterization methods of surface-modified nanoparticles were discussed. Finally, the applications of these nanoparticles in different routes of treatments were explored. EXPERT OPINION Surface modification of anticancer drug-loaded polymeric nanoparticles can enhance the efficacy, selective targeting, and biodistribution of the anticancer drug at the tumor site.
Collapse
Affiliation(s)
- Arsalan Ahmed
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Shumaila Sarwar
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan.,Faculty of Pharmacy, University of Sargodha , Sargodha, Pakistan
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu, China
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University , Sakaka, Aljouf, Saudi Arabia
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences , Bahawalpur, Pakistan
| | - Fakhera Ikram
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Anila Asif
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Saeed Ur Rahman
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Aqif Anwar Chaudhry
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Ihtasham Ur Rehman
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan.,Bioengineering, Engineering Department, Lancaster University , Lancaster, UK
| |
Collapse
|
21
|
Gu S, Song X, Xie R, Ouyang C, Xie L, Li Q, Su T, Xu M, Xu T, Huang D, Liang B. Berberine inhibits cancer cells growth by suppressing fatty acid synthesis and biogenesis of extracellular vesicles. Life Sci 2020; 257:118122. [PMID: 32702446 DOI: 10.1016/j.lfs.2020.118122] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 02/05/2023]
Abstract
AIMS Berberine is an isoquinoline alkaloid extracted from the root, rhizome and stem bark of Coptidis Rhizoma. Previous studies have revealed the anti-tumor potential of berberine against various types of cancer cells. However, the underlying mechanisms are not yet fully understood. In this study, we focused on the effects of berberine on fatty acid synthesis and extracellular vesicles formation in cancer cells, and revealed the internal mechanism of berberine inhibition on cancer cell proliferation. MATERIALS AND METHODS Anti-proliferative activity of berberine was determined by cell counting and microscope observation and cell cycle analysis. Activities of AMPK and ACC, expression of extracellular vesicles markers were detected by western blotting. 13C labeling metabolic flux analysis was used for determination of de novo synthesis of fatty acids. The excreted extracellular vesicles in culture mediums were separated by both polyethylene glycol enrichment of extracellular vesicles and differential centrifugation separation. KEY FINDINGS Among our early experiments, 5-10 μmol/L berberine exhibited the substantial anti-proliferative effect against human colon cancer cell line HCT116, cervical cancer cell line HeLa and other cancer cells. It was also revealed that, through activating AMPK, berberine inhibited ACC activity then suppressed intracellular fatty acid synthesis, finally decreased the biogenesis of extracellular vesicles. Moreover, supplement with citrate acid, palmitic acid, as well as exogenous extracellular vesicles, could rescue the inhibitory effect of berberine on cell proliferation, suggesting that inhibited ACC activity, suppressed fatty acid synthesis and decreased extracellular vesicles production were important mechanisms account for berberine inhibiting cancer cell proliferation. SIGNIFICANCE Our study indicates that berberine suppresses cancer cell proliferation through inhibiting the synthesis of fatty acids and decreasing biogenesis and secretion of extracellular vesicles, suggests that berberine is a promising candidate for the development of new therapies for cancer.
Collapse
Affiliation(s)
- Songgang Gu
- Department of Biliary-Pancreatic Minimally Invasive Surgery, the First Affiliated Hospital, Shantou University Medical College, Guangdong, China; Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Xuhong Song
- Center for Cancer Research, Shantou University Medical College, Guangdong, China
| | - Rufei Xie
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Cong Ouyang
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Lingzhu Xie
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China; Biomedical Research Center, Shantou University Medical College, Guangdong, China
| | - Qidong Li
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Ting Su
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Man Xu
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Tian Xu
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China
| | - Dongyang Huang
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China.
| | - Bin Liang
- Section of Cell Biology and Genetics, Shantou University Medical College, Guangdong, China; Biomedical Research Center, Shantou University Medical College, Guangdong, China.
| |
Collapse
|
22
|
Kaczmarska M, Grosicki M, Bulat K, Mardyla M, Szczesny-Malysiak E, Blat A, Dybas J, Sacha T, Marzec KM. Temporal sequence of the human RBCs' vesiculation observed in nano-scale with application of AFM and complementary techniques. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102221. [PMID: 32438105 DOI: 10.1016/j.nano.2020.102221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/27/2020] [Accepted: 04/26/2020] [Indexed: 12/22/2022]
Abstract
Based on the multimodal characterization of human red blood cells (RBCs), the link between the storage-related sequence of the nanoscale changes in RBC membranes in the relation to their biochemical profile as well as mechanical and functional properties was presented. On the background of the accumulation of RBCs waste products, programmed cell death and impaired rheological properties, progressive alterations in the RBC membranes including changes in their height and diameter as well as the in situ characterization of RBC-derived microparticles (RMPs) on the RBCs surface were presented. The advantage of atomic force microscopy (AFM) in RMPs visualization, even at the very early stage of vesiculation, was shown based on the results revealed by other reference techniques. The nanoscale characterization of RMPs was correlated with a decrease in cholesterol and triglycerides levels in the RBC membranes, proving the link between the lipids leakage from RBCs and the process of vesiculation.
Collapse
Affiliation(s)
- Magdalena Kaczmarska
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Marek Grosicki
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Katarzyna Bulat
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Mateusz Mardyla
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland; Faculty of Motor Rehabilitation, University of Physical Education, Krakow, Poland
| | - Ewa Szczesny-Malysiak
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Aneta Blat
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Jakub Dybas
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Tomasz Sacha
- Chair and Department of Hematology, Jagiellonian University Hospital, Krakow, Poland
| | - Katarzyna M Marzec
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
23
|
Galle JN, Hegemann JH. Exofacial phospholipids at the plasma membrane: ill-defined targets for early infection processes. Biol Chem 2020; 400:1323-1334. [PMID: 31408428 DOI: 10.1515/hsz-2019-0187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/06/2019] [Indexed: 02/04/2023]
Abstract
The eukaryotic plasma membrane (PM) consists largely of phospholipids and proteins, and separates the intracellular compartments from the extracellular space. It also serves as a signaling platform for cell-to-cell communication and an interaction platform for the molecular crosstalk between pathogens and their target cells. Much research has been done to elucidate the interactions between pathogens and host membrane proteins. However, little is known about the interactions between pathogens and membrane phospholipids, although reports have described a contribution of phospholipids to cell recognition and/or invasion during early infection by diverse pathogens. Thus, during adhesion to the host cell, the obligate intracellular bacterial pathogens Chlamydia spp., the facultative intracellular pathogen Helicobacter pylori and the facultative aerobic pathogen Vibrio parahaemolyticus, interact with exofacial phospholipids. This review focuses on several prominent instances of pathogen interaction with host-cell phospholipids.
Collapse
Affiliation(s)
- Jan N Galle
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Johannes H Hegemann
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
24
|
A Mechanosensitive Channel Governs Lipid Flippase-Mediated Echinocandin Resistance in Cryptococcus neoformans. mBio 2019; 10:mBio.01952-19. [PMID: 31822582 PMCID: PMC6904872 DOI: 10.1128/mbio.01952-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Echinocandins show fungicidal activity against common invasive mycoses but are ineffective against cryptococcosis. The underlying mechanism for echinocandin resistance in Cryptococcus neoformans remains poorly understood but has been shown to involve Cdc50, the regulatory subunit of lipid flippase. In a forward genetic screen for cdc50Δ suppressor mutations that are caspofungin resistant, we identified Crm1 (caspofungin resistant mutation 1), a homolog of mechanosensitive channel proteins, and showed that crm1Δ restored caspofungin resistance in cdc50Δ cells. Caspofungin-treated cdc50Δ cells exhibited abnormally high intracellular calcium levels ([Ca2+]c) and heightened activation of the calcineurin pathway. Deletion of CRM1 in the cdc50Δ background normalized the abnormally high [Ca2+]c. Cdc50 interacts with Crm1 to maintain cellular calcium homeostasis. Analysis of chitin/chitosan content showed that deleting CRM1 reversed the decreased chitosan production of cdc50Δ cells. Together, these results demonstrate that Cdc50 and Crm1 regulation of the calcineurin pathway and cytoplasmic calcium homeostasis may underlie caspofungin resistance in C. neoformans IMPORTANCE Cryptococcus neoformans is the leading cause of fungal meningitis, accounting for ∼15% of HIV/AIDS-related deaths, but treatment options for cryptococcosis are limited. Echinocandins are the newest fungicidal drug class introduced but are ineffective in treating cryptococcosis. Our previous study identified the lipid flippase subunit Cdc50 as a contributor to echinocandin resistance in C. neoformans Here, we further elucidated the mechanism of Cdc50-mediated caspofungin drug resistance. We discovered that Cdc50 interacts with the mechanosensitive calcium channel protein Crm1 to regulate calcium homeostasis and caspofungin resistance via calcium/calcineurin signaling. These results provide novel insights into echinocandin resistance in this pathogen, which may lead to new treatment options, as well as inform echinocandin resistance mechanisms in other fungal organisms and, hence, advance our understanding of modes of antifungal drug susceptibility and resistance.
Collapse
|
25
|
Ezzati M, Shanehbandi D, Hamdi K, Rahbar S, Pashaiasl M. Influence of cryopreservation on structure and function of mammalian spermatozoa: an overview. Cell Tissue Bank 2019; 21:1-15. [DOI: 10.1007/s10561-019-09797-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022]
|
26
|
Taus F, Meneguzzi A, Castelli M, Minuz P. Platelet-Derived Extracellular Vesicles as Target of Antiplatelet Agents. What Is the Evidence? Front Pharmacol 2019; 10:1256. [PMID: 31780927 PMCID: PMC6857039 DOI: 10.3389/fphar.2019.01256] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived large extracellular vesicles (often referred to as microparticles in the field of cardiovascular disease) have been identified as effector in the atherothrombotic process, therefore representing a target of pharmacological intervention of potential interest. Despite that, limited evidence is so far available concerning the effects of antiplatelet agents on the release of platelet-derived extracellular vesicles. In the present narrative review, the mechanisms leading to vesiculation in platelets and the pathophysiological processes implicated will be discussed. This will be followed by a summary of the present evidence concerning the effects of antiplatelet agents under experimental conditions and in clinical settings.
Collapse
Affiliation(s)
- Francesco Taus
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Alessandra Meneguzzi
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Marco Castelli
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Pietro Minuz
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| |
Collapse
|
27
|
Carden MA, Fasano RM, Meier ER. Not all red cells sickle the same: Contributions of the reticulocyte to disease pathology in sickle cell anemia. Blood Rev 2019; 40:100637. [PMID: 31735458 DOI: 10.1016/j.blre.2019.100637] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/05/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022]
Abstract
Sickle cell anemia (SCA) is associated with morbidity and early death. While the switch from fetal to sickle hemoglobin during the first months of life results in hemolytic anemia with reticulocytosis, the role of the reticulocyte in the pathophysiology and prognosis of SCA is not well-defined. Reticulocytes have unique cytoskeletal and membrane components that allow them to be distinguished from mature sickle erythrocytes in the circulation. Reticulocytes in patients with SCA are less dense than more mature and 'sickled' erythrocytes, and have increased adhesive properties. The circulating reticulocyte number in peripheral blood may assist in predicting disease severity in SCA; characterization of patient-specific reticulocyte properties during infancy and childhood may assist in predicting therapeutic response to therapies. Here, we review the biological and clinical data regarding reticulocytes and their potential impact on SCA pathophysiology and disease severity.
Collapse
Affiliation(s)
- Marcus A Carden
- Departments of Pediatrics and Medicine, UNC School of Medicine, UNC Blood Research Center, 170 Manning Drive, POB-CB#7236, Chapel Hill, North Carolina 27599, USA.
| | - Ross M Fasano
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1405 Clifton Road NE, Atlanta, GA 30322, USA.
| | - Emily Riehm Meier
- Indiana Hemophilia and Thrombosis Center, 8326 Naab Road, Indianapolis, Indiana 46220, USA.
| |
Collapse
|
28
|
Huang M, Dong Y, Zhang Y, Chen Q, Xie J, Xu C, Zhao Q, Li E. Growth and Lipidomic Responses of Juvenile Pacific White Shrimp Litopenaeus vannamei to Low Salinity. Front Physiol 2019; 10:1087. [PMID: 31507450 PMCID: PMC6716509 DOI: 10.3389/fphys.2019.01087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
The Pacific white shrimp (Litopenaeus vannamei), a euryhaline penaeid species, can tolerate a wide range of salinities, but little is known on its strategies to cope with low salinity fluctuations from the aspect of lipidomics. Thus, in this study, L. vannamei were grown in two different salinities [3 and 30‰ (control)] for 8 weeks, and then an liquid chromatography (LC)-mass spectrometry (MS)-based lipidomics analysis was performed to reveal the lipid profile differences in gill and muscle. L. vannamei under low salinity had lower weight gain and condition factor than the control shrimp at 30‰, but no differences were found in survival and hepatopancreas index. A higher number of differential lipid metabolites were identified in gill than in muscle in L. vannamei at salinity 3‰ relative to the control shrimp at salinity of 30‰ (159 versus 37), which belonged to 11 and 6 lipids classes, respectively. Of these lipids, phosphatidylcholine (PC), phosphatidylinositol (PI), phosphatidic acid (PA), phosphatidylethanolamine (PE), and triglyceride (TG) were the main lipids in both shrimp gill and muscle, regardless of salinities. Compared with the control shrimp at salinity 30‰, the percentage of PC significantly reduced, but TG and PA significantly increased in gill of shrimp at salinity 3‰. Moreover, the relative fatty acid abundances showed significant changes in L. vannamei between the two salinity groups, but the patterns of the changes were complex and were fatty acid dependent. Neither lipid nor fatty acid composition in muscle was affected by salinity. Further pathway analysis showed that these metabolites were closely related to lipid and fatty acid metabolic pathways. All the findings in this study reveal that the lipid variations are closely related to bio-membrane structure, mitochondrial function, energy supply, or organic osmolyte contents in hemolymph for improving osmoregulatory capacity of L. vannamei under low salinity.
Collapse
Affiliation(s)
- Maoxian Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Yangfan Dong
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Yan Zhang
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Qinsheng Chen
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Jia Xie
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Chang Xu
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Qun Zhao
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| | - Erchao Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, China
| |
Collapse
|
29
|
Regulation of boar sperm functionality by the nitric oxide synthase/nitric oxide system. J Assist Reprod Genet 2019; 36:1721-1736. [PMID: 31325069 PMCID: PMC6707978 DOI: 10.1007/s10815-019-01526-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose Nitric oxide (NO) is a free radical synthesized mainly by nitric oxide synthases (NOSs). NO regulates many aspects in sperm physiology in different species. However, in vitro studies investigating NOS distribution, and how NO influences sperm capacitation and fertilization (IVF) in porcine, have been lacking. Therefore, our study aimed to clarify these aspects. Methods Two main experiments were conducted: (i) boar spermatozoa were capacitated in the presence/absence of S-nitrosoglutathione (GSNO), a NO donor, and two NOS inhibitors, NG-nitro-L-arginine methyl ester hydrochloride (L-NAME) and aminoguanidine hemisulfate salt (AG), and (ii) IVF was performed in the presence or not of these supplements, but neither the oocytes nor the sperm were previously incubated in the supplemented media. Results Our results suggest that NOS distribution could be connected to pathways which lead to capacitation. Treatments showed significant differences after 30 min of incubation, compared to time zero in almost all motility parameters (P < 0.05). When NOSs were inhibited, three protein kinase A (PKA) substrates (~ 75, ~ 55, and ~50 kDa) showed lower phosphorylation levels between treatments (P < 0.05). No differences were observed in total tyrosine phosphorylation levels evaluated by Western blotting nor in situ. The percentage of acrosome-reacted sperm and phosphatidylserine translocation was significantly lower with L-NAME. Both inhibitors reduced sperm intracellular calcium concentration and IVF parameters, but L-NAME impaired sperm ability to penetrate denuded oocytes. Conclusions These findings point out to the importance of both sperm and cumulus-oocyte-derived NO in the IVF outcome in porcine. Electronic supplementary material The online version of this article (10.1007/s10815-019-01526-6) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Muller MP, Jiang T, Sun C, Lihan M, Pant S, Mahinthichaichan P, Trifan A, Tajkhorshid E. Characterization of Lipid-Protein Interactions and Lipid-Mediated Modulation of Membrane Protein Function through Molecular Simulation. Chem Rev 2019; 119:6086-6161. [PMID: 30978005 PMCID: PMC6506392 DOI: 10.1021/acs.chemrev.8b00608] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The cellular membrane constitutes one of the most fundamental compartments of a living cell, where key processes such as selective transport of material and exchange of information between the cell and its environment are mediated by proteins that are closely associated with the membrane. The heterogeneity of lipid composition of biological membranes and the effect of lipid molecules on the structure, dynamics, and function of membrane proteins are now widely recognized. Characterization of these functionally important lipid-protein interactions with experimental techniques is however still prohibitively challenging. Molecular dynamics (MD) simulations offer a powerful complementary approach with sufficient temporal and spatial resolutions to gain atomic-level structural information and energetics on lipid-protein interactions. In this review, we aim to provide a broad survey of MD simulations focusing on exploring lipid-protein interactions and characterizing lipid-modulated protein structure and dynamics that have been successful in providing novel insight into the mechanism of membrane protein function.
Collapse
Affiliation(s)
- Melanie P. Muller
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- College of Medicine
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tao Jiang
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chang Sun
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Muyun Lihan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shashank Pant
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Paween Mahinthichaichan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Anda Trifan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- College of Medicine
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
31
|
Molecular Mechanisms Underpinning Microparticle-Mediated Cellular Injury in Cardiovascular Complications Associated with Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6475187. [PMID: 30915196 PMCID: PMC6399542 DOI: 10.1155/2019/6475187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/13/2019] [Indexed: 12/15/2022]
Abstract
Microparticles (MPs) are small vesicles shed from the cytoplasmic membrane of healthy, activated, or apoptotic cells. MPs are very heterogeneous in size (100–1,000 nm), and they harbor proteins and surface antigens specific to cells they originate from. Virtually, all cells can shed MPs, and therefore, they can be found in all body fluids, but also entrapped in tissues. Of interest and because of their easy detection using a variety of techniques, circulating MPs were recognized as biomarkers for cell activation. MPs were also found to mediate critical actions in intercellular communication and transmitting biological messages by acting as paracrine vehicles. High plasma numbers of MPs were reported in many cardiovascular and metabolic disturbances that are closely associated with insulin resistance and low-grade inflammation and have been linked to adverse actions on cardiovascular function. This review highlights the involvement of MPs in cardiovascular complications associated with diabetes and discusses the molecular mechanisms that underpin the pathophysiological role of MPs in the onset and progression of cellular injury in diabetes.
Collapse
|
32
|
Liu Z, Barber C, Gupta A, Wan L, Won YW, Furenlid LR, Chen Q, Desai AA, Zhao M, Bull DA, Unger EC, Martin DR. Imaging assessment of cardioprotection mediated by a dodecafluoropentane oxygen-carrier administered during myocardial infarction. Nucl Med Biol 2019; 70:67-77. [PMID: 30772168 DOI: 10.1016/j.nucmedbio.2019.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/18/2018] [Accepted: 01/14/2019] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The objective of this study was to investigate the cardioprotective effects of a dodecafluoropentane (DDFP)-based perfluorocarbon emulsion (DDFPe) as an artificial carrier for oxygen delivery to ischemic myocardium, using 99mTc-duramycin SPECT imaging. METHODS Rat hearts with Ischemia-reperfusion (I/R) was prepared by coronary ligation for 45-min followed by reperfusion. The feasibility of 99mTc-duramycin in detecting myocardial I/R injury and its kinetic profile were first verified in the ischemic hearts with 2-h reperfusion (n = 6). DDFPe (0.6 mL/kg) was intravenously administered at 10 min after coronary ligation in fifteen rats and saline was given in thirteen rats as controls. 99mTc-duramycin SPECT images were acquired in the DDFPe-treated hearts and saline controls at 2-h (DDFPe-2 h, n = 7 and Saline-2 h, n = 6) or 24-h (DDFPe-24 h, n = 8 and Saline-24 h, n = 7) of reperfusion. RESULTS SPECT images, showing "hot-spot" 99mTc-duramycin uptake in the ischemic myocardium, exhibited significantly lower radioactive retention and smaller hot-spot size in the DDFPe-2 h and DDFPe-24 h hearts compared to controls. The infarcts in the Saline-24 h hearts extended significantly relative to measurements in the Saline-2 h. The extension of infarct size did not reach a statistical difference between the DDFPe-2 h and DDFPe-24 h hearts. Ex vivo measurement of 99mTc-duramycin activity (%ID/g) was lower in the ischemic area of DDFPe-2 h and DDFPe-24 h than that of the Saline-2 h and Saline-24 h hearts (P < 0.05). The area of injured myocardium, delineated by the uptake of 99mTc-duramycin, extended more substantially outside the infarct zone in the controls. CONCLUSIONS Significant reduction in myocardial I/R injury, as assessed by 99mTc-duramycin cell death imaging and histopathological analysis, was induced by DDFPe treatment after acute myocardial ischemia. 99mTc-duramycin imaging can reveal myocardial cell death in ischemic hearts and may provide a tool for the non-invasive assessment of cardioprotective interventions.
Collapse
Affiliation(s)
- Zhonglin Liu
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America.
| | - Christy Barber
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Akash Gupta
- Division of Cardiology of Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Li Wan
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Young-Wook Won
- Division of Cardiothoracic Surgery of Department of Surgery, University of Arizona, Tucson, AZ, United States of America
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Qin Chen
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States of America
| | - Ankit A Desai
- Division of Cardiology of Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Ming Zhao
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - David A Bull
- Division of Cardiothoracic Surgery of Department of Surgery, University of Arizona, Tucson, AZ, United States of America
| | - Evan C Unger
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; NuvOx Pharma, LLC., Tucson, AZ, United States of America
| | - Diego R Martin
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
33
|
|
34
|
Haskali MB, Denoyer D, Roselt PD, Hicks RJ, Hutton CA. Radiosynthesis and preliminary in vivo evaluation of 18F-labelled glycosylated duramycin peptides for imaging of phosphatidylethanolamine during apoptosis. MEDCHEMCOMM 2019. [DOI: 10.1039/c9md00354a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[18F]-Labelled duramycin derivatives incorporating hydrophilic aminogalacturonic acid moieties were prepared as tracers for in vivo imaging of phosphatidylethanolamine during apoptosis.
Collapse
Affiliation(s)
- Mohammad B. Haskali
- School of Chemistry
- The University of Melbourne
- Australia
- Bio21 Molecular Science and Biotechnology Institute
- The University of Melbourne
| | - Delphine Denoyer
- The Centre for Molecular Imaging and Translational Research Laboratory
- The Peter MacCallum Cancer Centre
- Melbourne
- Australia
| | - Peter D. Roselt
- The Centre for Molecular Imaging and Translational Research Laboratory
- The Peter MacCallum Cancer Centre
- Melbourne
- Australia
| | - Rodney J. Hicks
- The Centre for Molecular Imaging and Translational Research Laboratory
- The Peter MacCallum Cancer Centre
- Melbourne
- Australia
- The Sir Peter MacCallum Department of Oncology
| | - Craig A. Hutton
- School of Chemistry
- The University of Melbourne
- Australia
- Bio21 Molecular Science and Biotechnology Institute
- The University of Melbourne
| |
Collapse
|
35
|
Ahyayauch H, García-Arribas AB, Sot J, González-Ramírez EJ, Busto JV, Monasterio BG, Jiménez-Rojo N, Contreras FX, Rendón-Ramírez A, Martin C, Alonso A, Goñi FM. Pb(II) Induces Scramblase Activation and Ceramide-Domain Generation in Red Blood Cells. Sci Rep 2018; 8:7456. [PMID: 29748552 PMCID: PMC5945622 DOI: 10.1038/s41598-018-25905-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/19/2018] [Indexed: 01/01/2023] Open
Abstract
The mechanisms of Pb(II) toxicity have been studied in human red blood cells using confocal microscopy, immunolabeling, fluorescence-activated cell sorting and atomic force microscopy. The process follows a sequence of events, starting with calcium entry, followed by potassium release, morphological change, generation of ceramide, lipid flip-flop and finally cell lysis. Clotrimazole blocks potassium channels and the whole process is inhibited. Immunolabeling reveals the generation of ceramide-enriched domains linked to a cell morphological change, while the use of a neutral sphingomyelinase inhibitor greatly delays the process after the morphological change, and lipid flip-flop is significantly reduced. These facts point to three major checkpoints in the process: first the upstream exchange of calcium and potassium, then ceramide domain formation, and finally the downstream scramblase activation necessary for cell lysis. In addition, partial non-cytotoxic cholesterol depletion of red blood cells accelerates the process as the morphological change occurs faster. Cholesterol could have a role in modulating the properties of the ceramide-enriched domains. This work is relevant in the context of cell death, heavy metal toxicity and sphingolipid signaling.
Collapse
Affiliation(s)
- Hasna Ahyayauch
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Institut Supérieur des Professions Infirmières et des Techniques de Santé, Rabat, Morocco.,Neuroendocrinology Unit, Laboratory of Genetics, Neuroendocrinology and Biotechnology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Aritz B García-Arribas
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Jesús Sot
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain
| | - Emilio J González-Ramírez
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Jon V Busto
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Bingen G Monasterio
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Noemi Jiménez-Rojo
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain.,NCCR Chemical Biology, Department of Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - F Xabier Contreras
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Adela Rendón-Ramírez
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Cesar Martin
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Alicia Alonso
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain.,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain
| | - Félix M Goñi
- Instituto Biofisika (CSIC, UPV/EHU), 48080, Bilbao, Spain. .,Departamento de Bioquímica, University of the Basque Country (UPV/EHU), 48080, Bilbao, Spain.
| |
Collapse
|
36
|
Willms E, Cabañas C, Mäger I, Wood MJA, Vader P. Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression. Front Immunol 2018; 9:738. [PMID: 29760691 PMCID: PMC5936763 DOI: 10.3389/fimmu.2018.00738] [Citation(s) in RCA: 663] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
Cells release membrane enclosed nano-sized vesicles termed extracellular vesicles (EVs) that function as mediators of intercellular communication by transferring biological information between cells. Tumor-derived EVs have emerged as important mediators in cancer development and progression, mainly through transfer of their bioactive content which can include oncoproteins, oncogenes, chemokine receptors, as well as soluble factors, transcripts of proteins and miRNAs involved in angiogenesis or inflammation. This transfer has been shown to influence the metastatic behavior of primary tumors. Moreover, tumor-derived EVs have been shown to influence distant cellular niches, establishing favorable microenvironments that support growth of disseminated cancer cells upon their arrival at these pre-metastatic niches. It is generally accepted that cells release a number of major EV populations with distinct biophysical properties and biological functions. Exosomes, microvesicles, and apoptotic bodies are EV populations most widely studied and characterized. They are discriminated based primarily on their intracellular origin. However, increasing evidence suggests that even within these EV populations various subpopulations may exist. This heterogeneity introduces an extra level of complexity in the study of EV biology and function. For example, EV subpopulations could have unique roles in the intricate biological processes underlying cancer biology. Here, we discuss current knowledge regarding the role of subpopulations of EVs in cancer development and progression and highlight the relevance of EV heterogeneity. The position of tetraspanins and integrins therein will be highlighted. Since addressing EV heterogeneity has become essential for the EV field, current and novel techniques for isolating EV subpopulations will also be discussed. Further dissection of EV heterogeneity will advance our understanding of the critical roles of EVs in health and disease.
Collapse
Affiliation(s)
- Eduard Willms
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Department of Microbiology I (Immunology), Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Institute of Technology, University of Tartu, Tartu, Estonia
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pieter Vader
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
37
|
Samanta S, Rajasingh S, Drosos N, Zhou Z, Dawn B, Rajasingh J. Exosomes: new molecular targets of diseases. Acta Pharmacol Sin 2018; 39:501-513. [PMID: 29219950 PMCID: PMC5888687 DOI: 10.1038/aps.2017.162] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) comprise apoptotic bodies, microvesicles and exosomes, and they perform as key regulators in cell-to-cell communication in normal as well as diseased states. EVs contain natural cargo molecules, such as miRNA, mRNA and proteins, and transfer these functional cargos to neighboring cells or more distant cells through circulation. These functionally active molecules then affect distinct signaling cascades. The message conveyed to the recipient cells is dependent upon the composition of the EV, which is determined by the parent cell and the EV biogenesis. Because of their properties such as increased stability in circulation, biocompatibility, low immunogenicity and toxicity, EVs have drawn attention as attractive delivery systems for therapeutics. This review focuses on the functional use of exosomes in therapy and the potential advantages and challenges in using exosomes for therapeutic purposes.
Collapse
Affiliation(s)
- Saheli Samanta
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Sheeja Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Nicholas Drosos
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Zhigang Zhou
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Buddhadeb Dawn
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Johnson Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
38
|
Rybczynska AA, Boersma HH, de Jong S, Gietema JA, Noordzij W, Dierckx RAJO, Elsinga PH, van Waarde A. Avenues to molecular imaging of dying cells: Focus on cancer. Med Res Rev 2018. [PMID: 29528513 PMCID: PMC6220832 DOI: 10.1002/med.21495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to “patient‐tailored therapy.” Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single‐photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target‐to‐nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.
Collapse
Affiliation(s)
- Anna A Rybczynska
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Hendrikus H Boersma
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy & Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Walter Noordzij
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Philip H Elsinga
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
39
|
Said AS, Rogers SC, Doctor A. Physiologic Impact of Circulating RBC Microparticles upon Blood-Vascular Interactions. Front Physiol 2018; 8:1120. [PMID: 29379445 PMCID: PMC5770796 DOI: 10.3389/fphys.2017.01120] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Here, we review current data elucidating the role of red blood cell derived microparticles (RMPs) in normal vascular physiology and disease progression. Microparticles (MPs) are submicron-size, membrane-encapsulated vesicles derived from various parent cell types. MPs are produced in response to numerous stimuli that promote a sequence of cytoskeletal and membrane phospholipid changes and resulting MP genesis. MPs were originally considered as potential biomarkers for multiple disease processes and more recently are recognized to have pleiotropic biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in initiating apoptosis. RMPs, specifically, form normally during RBC maturation in response to injury during circulation, and are copiously produced during processing and storage for transfusion. Notably, several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs markedly from that of intact RBCs and the nature/composition of RMP components are affected by the specific circumstances of RMP genesis. Described RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion as well as influence upon vasoregulation via influence upon nitric oxide (NO) bioavailability. Of particular relevance, RMPs scavenge NO more avidly than do intact RBCs; this physiology has been proposed to contribute to the impaired oxygen delivery homeostasis that may be observed following transfusion. In summary, RMPs are submicron particles released from RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in normal and patho-physiology and in transfusion recipients is an area of continued investigation.
Collapse
Affiliation(s)
- Ahmed S Said
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Stephen C Rogers
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Allan Doctor
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States.,Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
40
|
Bouchet S, Tang R, Fava F, Legrand O, Bauvois B. The CNGRC-GG-D(KLAKLAK)2 peptide induces a caspase-independent, Ca2+-dependent death in human leukemic myeloid cells by targeting surface aminopeptidase N/CD13. Oncotarget 2017; 7:19445-67. [PMID: 26655501 PMCID: PMC4991394 DOI: 10.18632/oncotarget.6523] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/16/2015] [Indexed: 02/06/2023] Open
Abstract
The CD13 antigen's binding site for the Asn-Gly-Arg (NGR) motif enables NGR-containing chemotherapeutic drugs to be delivered to CD13-positive tumours. Human CD13-positive acute myeloid leukemia (AML) cells proliferate abnormally and escape death. Here, we show that the CNGRC-GG-D(KLAKLAK)2 peptide induces death in AML cell lines (U937, THP-1, NB4, HL-60) and primary blood cells from AML patients. Cell death was characterized as a caspase-independent mechanism, without DNA fragmentation, but phosphatidylserine externalization and membrane disruption. Our results demonstrate in U937 cells that (i) the NGR-peptide triggers the loss of mitochondrial potential(ΔΨm) and generates superoxide anion (O2-), (ii) N-acetyl-L-cysteine (NAC) and extra/intracellular Ca2+ chelators (BAPTA) prevent both O2- production and cell death, (iii) the Ca2+-channel blocker nifedipine prevents cell death (indicating that Ca2+ influx is the initial death trigger), and (iv) BAPTA, but not NAC, prevents ΔΨm loss (suggesting O2- is a mitochondrial downstream effector). AML cell lines and primary blasts responding to the lethal action of NGR-peptide express promatrix metalloproteinase-12 (proMMP-12) and its substrate progranulin (an 88 kDa cell survival factor). A cell-free assay highlighted proMMP-12 activation by O2-. Accordingly, NGR-peptide's downregulation of 88 kDa progranulin protein was prevented by BAPTA and NAC. Conversely, AML blast resistance to NGR-peptide is associated with the expression of a distinct, 105 kDa progranulin isoform. These results indicate that CNGRC-GG-D(KLAKLAK)2 induces death in AML cells through the Ca2+-mitochondria-O2.-pathway, and support the link between proMMP-12 activation and progranulin cleavage during cell death. Our findings may have implications for the understanding of tumour biology and treatment.
Collapse
Affiliation(s)
- Sandrine Bouchet
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France.,Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Ruoping Tang
- Centre de Recherche de Saint-Antoine, INSERM UMRS 938, Service d'Hématologie, Hôpital St Antoine, Paris, France.,Sorbonne Universités UPMC Paris 06, Paris, France
| | - Fanny Fava
- Centre de Recherche de Saint-Antoine, INSERM UMRS 938, Service d'Hématologie, Hôpital St Antoine, Paris, France.,Sorbonne Universités UPMC Paris 06, Paris, France
| | - Ollivier Legrand
- Centre de Recherche de Saint-Antoine, INSERM UMRS 938, Service d'Hématologie, Hôpital St Antoine, Paris, France.,Sorbonne Universités UPMC Paris 06, Paris, France
| | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| |
Collapse
|
41
|
Cholesterol metabolism and glaucoma: Modulation of Muller cell membrane organization by 24S-hydroxycholesterol. Chem Phys Lipids 2017; 207:179-191. [DOI: 10.1016/j.chemphyslip.2017.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 02/04/2023]
|
42
|
Thom SR, Bhopale VM, Yu K, Huang W, Kane MA, Margolis DJ. Neutrophil microparticle production and inflammasome activation by hyperglycemia due to cytoskeletal instability. J Biol Chem 2017; 292:18312-18324. [PMID: 28972154 DOI: 10.1074/jbc.m117.802629] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/11/2017] [Indexed: 12/23/2022] Open
Abstract
Microparticles are lipid bilayer-enclosed vesicles produced by cells under oxidative stress. MP production is elevated in patients with diabetes, but the underlying cellular mechanisms are poorly understood. We hypothesized that raising glucose above the physiological level of 5.5 mm would stimulate leukocytes to produce MPs and activate the nucleotide-binding domain, leucine-rich repeat pyrin domain-containing 3 (NLRP3) inflammasome. We found that when incubated in buffer with up to 20 mm glucose, human and murine neutrophils, but not monocytes, generate progressively more MPs with high interleukin (IL)-1β content. Enhanced MP production required generation of reactive chemical species by mitochondria, NADPH oxidase, and type 2 nitric-oxide synthase (NOS-2) and resulted in S-nitrosylation of actin. Depleting cells of capon (C-terminal PDZ ligand of neuronal nitric-oxide synthase protein), apoptosis-associated speck-like protein containing C-terminal caspase recruitment domain (ASC), or pro-IL-1β prevented the hyperglycemia-induced enhancement of reactive species production, MP generation, and IL-1β synthesis. Additional components required for these responses included inositol 1,3,5-triphosphate receptors, PKC, and enhancement of filamentous-actin turnover. Numerous proteins become localized to short filamentous actin in response to S-nitrosylation, including vasodilator-stimulated phosphoprotein, focal adhesion kinase, the membrane phospholipid translocation enzymes flippase and floppase, capon, NLRP3, and ASC. We conclude that an interdependent oxidative stress response to hyperglycemia perturbs neutrophil cytoskeletal stability leading to MP production and IL-1β synthesis.
Collapse
Affiliation(s)
- Stephen R Thom
- From the Department of Emergency Medicine, School of Medicine, and
| | - Veena M Bhopale
- From the Department of Emergency Medicine, School of Medicine, and
| | - Kevin Yu
- From the Department of Emergency Medicine, School of Medicine, and
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201 and
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201 and
| | - David J Margolis
- the Department of Dermatology and Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
43
|
Influence of red blood cell-derived microparticles upon vasoregulation. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2017; 15:522-534. [PMID: 28686154 DOI: 10.2450/2017.0353-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022]
Abstract
Here we review recent data and the evolving understanding of the role of red blood cell-derived microparticles (RMPs) in normal physiology and in disease progression. Microparticles (MPs) are small membrane vesicles derived from various parent cell types. MPs are produced in response to a variety of stimuli through several cytoskeletal and membrane phospholipid changes. MPs have been investigated as potential biomarkers for multiple disease processes and are thought to have biological effects, most notably in: promotion of coagulation, production and handling of reactive oxygen species, immune modulation, angiogenesis, and in apoptosis. Specifically, RMPs are produced normally during RBC maturation and their production is accelerated during processing and storage for transfusion. Several factors during RBC storage are known to trigger RMP production, including: increased intracellular calcium, increased potassium leakage, and energy failure with ATP depletion. Of note, RMP composition differs from that of intact RBCs, and the nature and composition of RMP components are affected by both storage duration and the character of storage solutions. Recognised RMP bioactivities include: promotion of coagulation, immune modulation, and promotion of endothelial adhesion, as well as influence upon vasoregulation via nitric oxide (NO) scavenging. Of particular relevance, RMPs are more avid NO scavengers than intact RBCs and this feature has been proposed as a mechanism for the impaired oxygen delivery homeostasis that has been observed following transfusion. Preliminary human studies demonstrate that circulating RMP abundance increases with RBC transfusion and is associated with altered plasma vasoactivity and abnormal vasoregulation. In summary, RMPs are submicron particles released from stored RBCs, with demonstrated vasoactive properties that appear to disturb oxygen delivery homeostasis. The clinical impact of RMPs in transfusion recipients is an area of continued investigation.
Collapse
|
44
|
Selmaj I, Mycko MP, Raine CS, Selmaj KW. The role of exosomes in CNS inflammation and their involvement in multiple sclerosis. J Neuroimmunol 2017; 306:1-10. [PMID: 28385180 DOI: 10.1016/j.jneuroim.2017.02.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/03/2017] [Accepted: 02/03/2017] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is a putative autoimmune disease of the central nervous system (CNS) in which autoreactive immune cells recognizing myelin antigens lead to demyelination and axonal injury. Mechanisms relevant to the pathogenesis of MS have not been fully elucidated, particularly those underlying initiation of immune system dysfunction. For example, it is not known how reactivity against CNS components is generated within the peripheral immune system. In this review, we propose that a significant contribution to the immunoregulatory events may derive from a cell-to-cell communication system involving the production, secretion and transfer of extracellular vesicles known as exosomes. Herein, we discuss in detail the biogenesis and roles of these cell surface-generated vesicles from the standpoint of receptors and their cargo, microRNA. It is well known that exosomes can cross the blood-brain barrier and thus may contribute to the spread of brain antigens to the periphery. Further understanding of exosome-dependent mechanisms in MS should provide a novel angle to the analysis of the pathogenesis of this disease. Finally, we launch the idea that exosomes and their contents may serve as biomarkers in MS.
Collapse
Affiliation(s)
- Igor Selmaj
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| | - Marcin P Mycko
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| | - Cedric S Raine
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Krzysztof W Selmaj
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
45
|
Muratori C, Pakhomov AG, Gianulis EC, Jensen SD, Pakhomova ON. The cytotoxic synergy of nanosecond electric pulses and low temperature leads to apoptosis. Sci Rep 2016; 6:36835. [PMID: 27833151 PMCID: PMC5104977 DOI: 10.1038/srep36835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022] Open
Abstract
Electroporation by nanosecond electric pulses (nsEP) is an emerging modality for tumor ablation. Here we show the efficient induction of apoptosis even by a non-toxic nsEP exposure when it is followed by a 30-min chilling on ice. This chilling itself had no impact on the survival of U-937 or HPAF-II cells, but caused more than 75% lethality in nsEP-treated cells (300 ns, 1.8-7 kV/cm, 50-700 pulses). The cell death was largely delayed by 5-23 hr and was accompanied by a 5-fold activation of caspase 3/7 (compared to nsEP without chilling) and more than 60% cleavage of poly-ADP ribose polymerase (compared to less than 5% in controls or after nsEP or chilling applied separately). When nsEP caused a transient permeabilization of 83% of cells to propidium iodide, cells placed at 37 °C resealed in 10 min, whereas 60% of cells placed on ice remained propidium-permeable even in 30 min. The delayed membrane resealing caused cell swelling, which could be blocked by an isosmotic addition of a pore-impermeable solute (sucrose). However, the block of swelling did not prevent the delayed cell death by apoptosis. The potent enhancement of nsEP cytotoxicity by subsequent non-damaging chilling may find applications in tumor ablation therapies.
Collapse
Affiliation(s)
- Claudia Muratori
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Elena C Gianulis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Sarah Damsbo Jensen
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Olga N Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| |
Collapse
|
46
|
Probing phosphoethanolamine-containing lipids in membranes with duramycin/cinnamycin and aegerolysin proteins. Biochimie 2016; 130:81-90. [DOI: 10.1016/j.biochi.2016.09.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023]
|
47
|
The interaction of sorafenib and regorafenib with membranes is modulated by their lipid composition. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2871-2881. [PMID: 27581086 DOI: 10.1016/j.bbamem.2016.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/29/2016] [Accepted: 08/25/2016] [Indexed: 11/22/2022]
Abstract
Sorafenib and regorafenib are small-molecule kinase inhibitors approved for the treatment of locally recurrent or metastatic, progressive, differentiated thyroid carcinoma, renal cell carcinoma, and hepatocellular carcinoma (sorafenib) and of colorectal cancer (regorafenib). As of now, the mechanisms, which are responsible for their antitumor activities, are not completely understood. Given the lipophilic nature of the molecules, it can be hypothesized that the pharmacological impact is mediated by the interaction with cellular membranes as it is true for many pharmacologically active molecules. However, an interaction of sorafenib or regorafenib with lipid membranes has not yet been investigated in detail. Here, we characterized the interaction of both drugs with lipid membranes by applying a variety of biophysical approaches including nuclear magnetic resonance, electron spin resonance, and fluorescence spectroscopy. We found that sorafenib and regorafenib bind to lipid membranes by inserting into the lipid-water interface of the bilayer. This membrane embedding causes a disturbance of bilayer structure leading to an increased permeability of the membrane for polar molecules. One approach shows that the extent of the effects depends on the membrane lipid composition underlining a particular role of phosphatidylcholine and cholesterol. Our data for the first time characterize the impact of sorafenib and regorafenib on the lipid membrane structure and dynamics, which may contribute to a better understanding of their effectiveness in the treatment of malignancies as well as of their side effects.
Collapse
|
48
|
Flamant S, Tamarat R. Extracellular Vesicles and Vascular Injury: New Insights for Radiation Exposure. Radiat Res 2016; 186:203-18. [PMID: 27459703 DOI: 10.1667/rr14482.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This article reviews our current knowledge about cell-derived extracellular vesicles (EVs), including microparticles and exosomes, and their emergence as mediators of a new important mechanism of cell-to-cell communication. Particular emphasis has been given to the increasing involvement of EVs in the field of radiation-induced vascular injury. Although EVs have been considered for a long time as cell "dust", they in fact precisely reflect the physiological state of the cells. The role of microparticles and exosomes in mediating vascular dysfunction suggests that they may represent novel pathways in short- or long-distance paracrine intercellular signaling in vascular environment. In this article, the mechanisms involved in the biogenesis of microparticles and exosomes, their composition and participation in the pathogenesis of vascular dysfunction are discussed. Furthermore, this article highlights the concept of EVs as potent vectors of biological information and protagonists of an intercellular communication network. Special emphasis is made on EV-mediated microRNA transfer and on the principal consequences of such signal exchange on vascular injury and radiation-induced nontargeted effect. The recent progress in elucidating the biology of EVs has provided new insights for the field of radiation, advancing their use as diagnostic biomarkers or in therapeutic interventions.
Collapse
Affiliation(s)
- Stéphane Flamant
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Radia Tamarat
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| |
Collapse
|
49
|
Giesler KE, Marengo J, Liotta DC. Reduction Sensitive Lipid Conjugates of Tenofovir: Synthesis, Stability, and Antiviral Activity. J Med Chem 2016; 59:7097-110. [PMID: 27405794 DOI: 10.1021/acs.jmedchem.6b00428] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The therapeutic value of numerous small molecules hinges on their ability to permeate the plasma membrane. This is particularly true for tenofovir (TFV), adefovir, and other antiviral nucleosides that demonstrate potent antiviral activity but poor bioavailability. Using TFV as a model substrate, we hybridized two disparate prodrug strategies to afford novel reduction-sensitive lipid conjugates of TFV that exhibit subnanomolar activity toward HIV-1 and are stable in human plasma for more than 24 h with a therapeutic index approaching 30000. These compounds significantly rival the clinically approved formulation of TFV and revitalize the potential of disulfide-bearing prodrugs which have seen limited in vitro and in vivo success since their debut over 20 years ago. We further demonstrate the utility of these conjugates as a tool to indirectly probe the enzymatic hydrolysis of phosphonomonoesters that may further advance the development of other prodrug strategies for nucleosides, peptides, and beyond.
Collapse
Affiliation(s)
- Kyle E Giesler
- Department of Chemistry, Emory University , 1521 Dickey Drive NE, Atlanta, Georgia 30322, United States
| | - Jose Marengo
- Emory Institute for Drug Development , 954 Gatewood Road, Atlanta, Georgia 30329, United States
| | - Dennis C Liotta
- Department of Chemistry, Emory University , 1521 Dickey Drive NE, Atlanta, Georgia 30322, United States.,Emory Institute for Drug Development , 954 Gatewood Road, Atlanta, Georgia 30329, United States
| |
Collapse
|
50
|
Scheidt HA, Haralampiev I, Theisgen S, Schirbel A, Sbiera S, Huster D, Kroiss M, Müller P. The adrenal specific toxicant mitotane directly interacts with lipid membranes and alters membrane properties depending on lipid composition. Mol Cell Endocrinol 2016; 428:68-81. [PMID: 27002491 DOI: 10.1016/j.mce.2016.03.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/26/2016] [Accepted: 03/16/2016] [Indexed: 11/20/2022]
Abstract
Mitotane (o,p'.-DDD) is an orphan drug approved for the treatment of adrenocortical carcinoma. The mechanisms, which are responsible for this activity of the drug, are not completely understood. It can be hypothesized that an impact of mitotane is mediated by the interaction with cellular membranes. However, an interaction of mitotane with (lipid) membranes has not yet been investigated in detail. Here, we characterized the interaction of mitotane and its main metabolite o,p'-dichlorodiphenyldichloroacetic acid (o,p'-DDA) with lipid membranes by applying a variety of biophysical approaches of nuclear magnetic resonance, electron spin resonance, and fluorescence spectroscopy. We found that mitotane and o,p'-DDA bind to lipid membranes by inserting into the lipid-water interface of the bilayer. Mitotane but not o,p'-DDA directly causes a disturbance of bilayer structure leading to an increased permeability of the membrane for polar molecules. Mitotane induced alterations of the membrane integrity required the presence of phosphatidylethanolamine and/or cholesterol. Collectively, our data for the first time characterize the impact of mitotane on the lipid membrane structure and dynamics, which may contribute to a better understanding of specific mitotane effects and side effects.
Collapse
Affiliation(s)
- Holger A Scheidt
- University of Leipzig, Institute of Medical Physics and Biophysics, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Ivan Haralampiev
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, 10115 Berlin, Germany
| | - Stephan Theisgen
- University of Leipzig, Institute of Medical Physics and Biophysics, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Andreas Schirbel
- University Hospital Würzburg, Department of Nuclear Medicine, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Silviu Sbiera
- University Hospital Würzburg, Department of Internal Medicine I, Endocrinology and Diabetes Unit, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Daniel Huster
- University of Leipzig, Institute of Medical Physics and Biophysics, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Matthias Kroiss
- University Hospital Würzburg, Department of Internal Medicine I, Endocrinology and Diabetes Unit, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Peter Müller
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, 10115 Berlin, Germany.
| |
Collapse
|