1
|
Ye J, Yang D, Shi C, Zhou F, Wang P. Designer
DNA
Nanostructures and Their Cellular Uptake Behaviors. DNA NANOTECHNOLOGY FOR CELL RESEARCH 2024:375-399. [DOI: 10.1002/9783527840816.ch16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
2
|
Matkivska R, Samborska I, Maievskyi O. Effect of animal venom toxins on the main links of the homeostasis of mammals (Review). Biomed Rep 2024; 20:16. [PMID: 38144889 PMCID: PMC10739175 DOI: 10.3892/br.2023.1704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023] Open
Abstract
The human body is affected by environmental factors. The dynamic balance between the organism and its environment results from the influence of natural, anthropogenic and social aspects. The factors of exogenous origin determine development of adaptive changes. The present article summarises the mechanisms of animal venom toxins and homeostasis disruption in the body of mammals. The mechanisms underlying pathological changes are associated with shifts in biochemical reactions. Components of the immune, nervous and endocrine systems are key in the host defense and adaptation processes in response to venom by triggering signalling pathways (PI3kinase pathway, arachidonic acid cascade). Animal venom toxins initiate the development of inflammatory processes, the synthesis of pro-inflammatory mediators (cytokines), ROS, proteolytic enzymes, activate the migration of leukocytes and macrophages. Keratinocytes and endothelial cells act as protective barriers under the action of animal venom toxins on the body of mammals. In addition, the formation of pores in cell membranes, structural changes in cell ion channels are characteristic of the action of animal venom toxins.
Collapse
Affiliation(s)
- Ruzhena Matkivska
- Department of Descriptive and Clinical Anatomy, Bogomolets National Medical University, Kyiv 03680, Ukraine
| | - Inha Samborska
- Department of Biological and General Chemistry, National Pirogov Memorial Medical University, Vinnytsya 21018, Ukraine
| | - Oleksandr Maievskyi
- Department of Clinical Medicine, Educational and Scientific Center ‘Institute of Biology and Medicine’ of Taras Shevchenko National University of Kyiv, Kyiv 03127, Ukraine
| |
Collapse
|
3
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
4
|
Meléndez AV, Velasco Cárdenas RMH, Lagies S, Strietz J, Siukstaite L, Thomas OS, Tomisch J, Weber W, Kammerer B, Römer W, Minguet S. Novel lectin-based chimeric antigen receptors target Gb3-positive tumour cells. Cell Mol Life Sci 2022; 79:513. [PMID: 36097202 PMCID: PMC9468074 DOI: 10.1007/s00018-022-04524-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 07/19/2022] [Accepted: 07/31/2022] [Indexed: 11/05/2022]
Abstract
The link between cancer and aberrant glycosylation has recently become evident. Glycans and their altered forms, known as tumour-associated carbohydrate antigens (TACAs), are diverse, complex and difficult to target therapeutically. Lectins are naturally occurring glycan-binding proteins that offer a unique opportunity to recognise TACAs. T cells expressing chimeric antigen receptors (CARs) have proven to be a successful immunotherapy against leukaemias, but so far have shown limited success in solid tumours. We developed a panel of lectin-CARs that recognise the glycosphingolipid globotriaosylceramide (Gb3), which is overexpressed in various cancers, such as Burkitt's lymphoma, colorectal, breast and pancreatic. We have selected the following lectins: Shiga toxin's B-subunit from Shigella dysenteriae, LecA from Pseudomonas aeruginosa, and the engineered lectin Mitsuba from Mytilus galloprovincialis as antigen-binding domains and fused them to a well-known second-generation CAR. The Gb3-binding lectin-CARs have demonstrated target-specific cytotoxicity against Burkitt's lymphoma-derived cell lines as well as solid tumour cells from colorectal and triple-negative breast cancer. Our findings reveal the big potential of lectin-based CARs as therapeutical applications to target Gb3 and other TACAs expressed in haematological malignancies and solid tumours.
Collapse
Affiliation(s)
- Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Rubí M-H Velasco Cárdenas
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Simon Lagies
- Institute of Organic Chemistry, Albert-Ludwigs-University Freiburg, Albertstraße 21, 79102, Freiburg, Germany
| | | | - Lina Siukstaite
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Oliver S Thomas
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Jana Tomisch
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Wilfried Weber
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Bernd Kammerer
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute of Organic Chemistry, Albert-Ludwigs-University Freiburg, Albertstraße 21, 79102, Freiburg, Germany
- Centre for Integrative Signalling Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
- Center of Chronic Immunodeficiency (CCI), University Clinics and Medical Faculty, Freiburg, Germany.
| |
Collapse
|
5
|
Enterohemorrhagic Escherichia coli and a Fresh View on Shiga Toxin-Binding Glycosphingolipids of Primary Human Kidney and Colon Epithelial Cells and Their Toxin Susceptibility. Int J Mol Sci 2022; 23:ijms23136884. [PMID: 35805890 PMCID: PMC9266556 DOI: 10.3390/ijms23136884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are the human pathogenic subset of Shiga toxin (Stx)-producing E. coli (STEC). EHEC are responsible for severe colon infections associated with life-threatening extraintestinal complications such as the hemolytic-uremic syndrome (HUS) and neurological disturbances. Endothelial cells in various human organs are renowned targets of Stx, whereas the role of epithelial cells of colon and kidneys in the infection process has been and is still a matter of debate. This review shortly addresses the clinical impact of EHEC infections, novel aspects of vesicular package of Stx in the intestine and the blood stream as well as Stx-mediated extraintestinal complications and therapeutic options. Here follows a compilation of the Stx-binding glycosphingolipids (GSLs), globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) and their various lipoforms present in primary human kidney and colon epithelial cells and their distribution in lipid raft-analog membrane preparations. The last issues are the high and extremely low susceptibility of primary renal and colonic epithelial cells, respectively, suggesting a large resilience of the intestinal epithelium against the human-pathogenic Stx1a- and Stx2a-subtypes due to the low content of the high-affinity Stx-receptor Gb3Cer in colon epithelial cells. The review closes with a brief outlook on future challenges of Stx research.
Collapse
|
6
|
Need for more focus on lipid species in studies of biological and model membranes. Prog Lipid Res 2022; 86:101160. [DOI: 10.1016/j.plipres.2022.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/06/2022] [Indexed: 11/23/2022]
|
7
|
Rosso DA, Rosato M, Gómez FD, Álvarez RS, Shiromizu CM, Keitelman IA, Ibarra C, Amaral MM, Jancic CC. Human Glomerular Endothelial Cells Treated With Shiga Toxin Type 2 Activate γδ T Lymphocytes. Front Cell Infect Microbiol 2021; 11:765941. [PMID: 34900753 PMCID: PMC8656354 DOI: 10.3389/fcimb.2021.765941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/19/2021] [Indexed: 12/02/2022] Open
Abstract
The hemolytic uremic syndrome associated with diarrhea, a consequence of Shiga toxin (Stx)-producing Escherichia coli infection, is a common cause of pediatric acute renal failure in Argentina. Stx type 2a (Stx2a) causes direct damage to renal cells and induces local inflammatory responses that involve secretion of inflammatory mediators and the recruitment of innate immune cells. γδ T cells constitute a subset of T lymphocytes, which act as early sensors of cellular stress and infection. They can exert cytotoxicity against infected and transformed cells, and produce cytokines and chemokines. In this study, we investigated the activation of human peripheral γδ T cells in response to the incubation with Stx2a-stimulated human glomerular endothelial cells (HGEC) or their conditioned medium, by analyzing in γδ T lymphocytes, the expression of CD69, CD107a, and perforin, and the production of TNF-α and IFN-γ. In addition, we evaluated by confocal microscopy the contact between γδ T cells and HGEC. This analysis showed an augmentation in cellular interactions in the presence of Stx2a-stimulated HGEC compared to untreated HGEC. Furthermore, we observed an increase in cytokine production and CD107a expression, together with a decrease in intracellular perforin when γδ T cells were incubated with Stx2a-treated HGEC or their conditioned medium. Interestingly, the blocking of TNF-α by Etanercept reversed the changes in the parameters measured in γδ T cells incubated with Stx2a-treated HGEC supernatants. Altogether, our results suggest that soluble factors released by Stx2a-stimulated HGEC modulate the activation of γδ T cells, being TNF-α a key player during this process.
Collapse
Affiliation(s)
- David Antonio Rosso
- Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina., Buenos Aires, Argentina
| | - Micaela Rosato
- Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina., Buenos Aires, Argentina
| | - Fernando Daniel Gómez
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Romina Soledad Álvarez
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Maiumi Shiromizu
- Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina., Buenos Aires, Argentina
| | - Irene Angélica Keitelman
- Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina., Buenos Aires, Argentina
| | - Cristina Ibarra
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Marta Amaral
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Cristina Jancic
- Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina., Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
8
|
Róg T, Girych M, Bunker A. Mechanistic Understanding from Molecular Dynamics in Pharmaceutical Research 2: Lipid Membrane in Drug Design. Pharmaceuticals (Basel) 2021; 14:1062. [PMID: 34681286 PMCID: PMC8537670 DOI: 10.3390/ph14101062] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
We review the use of molecular dynamics (MD) simulation as a drug design tool in the context of the role that the lipid membrane can play in drug action, i.e., the interaction between candidate drug molecules and lipid membranes. In the standard "lock and key" paradigm, only the interaction between the drug and a specific active site of a specific protein is considered; the environment in which the drug acts is, from a biophysical perspective, far more complex than this. The possible mechanisms though which a drug can be designed to tinker with physiological processes are significantly broader than merely fitting to a single active site of a single protein. In this paper, we focus on the role of the lipid membrane, arguably the most important element outside the proteins themselves, as a case study. We discuss work that has been carried out, using MD simulation, concerning the transfection of drugs through membranes that act as biological barriers in the path of the drugs, the behavior of drug molecules within membranes, how their collective behavior can affect the structure and properties of the membrane and, finally, the role lipid membranes, to which the vast majority of drug target proteins are associated, can play in mediating the interaction between drug and target protein. This review paper is the second in a two-part series covering MD simulation as a tool in pharmaceutical research; both are designed as pedagogical review papers aimed at both pharmaceutical scientists interested in exploring how the tool of MD simulation can be applied to their research and computational scientists interested in exploring the possibility of a pharmaceutical context for their research.
Collapse
Affiliation(s)
- Tomasz Róg
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Alex Bunker
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland;
| |
Collapse
|
9
|
Ran G, Feng XL, Xie YL, Zheng QY, Guo PP, Yang M, Feng YL, Ling C, Zhu LQ, Zhong C. The use of miR122 and its target sequence in adeno-associated virus-mediated trichosanthin gene therapy. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:515-525. [PMID: 34538767 DOI: 10.1016/j.joim.2021.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Plant-derived cytotoxic transgene expression, such as trichosanthin (tcs), regulated by recombinant adeno-associated virus (rAAV) vector is a promising cancer gene therapy. However, the cytotoxic transgene can hamper the vector production in the rAAV producer cell line, human embryonic kidney (HEK293) cells. Here, we explored microRNA-122 (miR122) and its target sequence to limit the expression of the cytotoxic gene in the rAAV producer cells. METHODS A miR122 target (122T) sequence was incorporated into the 3' untranslated region of the tcs cDNA sequence. The firefly luciferase (fluc) transgene was used as an appropriate control. Cell line HEK293-mir122 was generated by the lentiviral vector-mediated genome integration of the mir122 gene in parental HEK293 cells. The effects of miR122 overexpression on cell growth, transgene expression, and rAAV production were determined. RESULTS The presence of 122T sequence significantly reduced transgene expression in the miR122-enriched Huh7 cell line (in vitro), fresh human hepatocytes (ex vivo), and mouse liver (in vivo). Also, the normal liver physiology was unaffected by delivery of 122T sequence by rAAV vectors. Compared with the parental cells, the miR122-overexpressing HEK293-mir122 cell line showed similar cell growth rate and expression of transgene without 122T, as well as the ability to produce liver-targeting rAAV vectors. Fascinatingly, the yield of rAAV vectors carrying the tcs-122T gene was increased by 77.7-fold in HEK293-mir122 cells. Moreover, the tcs-122T-containing rAAV vectors significantly reduced the proliferation of hepatocellular carcinoma cells without affecting the normal liver cells. CONCLUSION HEK293-mir122 cells along with the 122T sequence provide a potential tool to attenuate the cytotoxic transgene expression, such as tcs, during rAAV vector production.
Collapse
Affiliation(s)
- Gai Ran
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Xi-Lin Feng
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yi-Lin Xie
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Qing-Yun Zheng
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Peng-Peng Guo
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army 971 Hospital, Qingdao 266071, Shandong Province, China
| | - Ming Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Ying-Lu Feng
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army 971 Hospital, Qingdao 266071, Shandong Province, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Li-Qing Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chen Zhong
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China.
| |
Collapse
|
10
|
Detzner J, Klein AL, Pohlentz G, Krojnewski E, Humpf HU, Mellmann A, Karch H, Müthing J. Primary Human Renal Proximal Tubular Epithelial Cells (pHRPTEpiCs): Shiga Toxin (Stx) Glycosphingolipid Receptors, Stx Susceptibility, and Interaction with Membrane Microdomains. Toxins (Basel) 2021; 13:toxins13080529. [PMID: 34437399 PMCID: PMC8402424 DOI: 10.3390/toxins13080529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Tubular epithelial cells of the human kidney are considered as targets of Shiga toxins (Stxs) in the Stx-mediated pathogenesis of hemolytic–uremic syndrome (HUS) caused by Stx-releasing enterohemorrhagic Escherichia coli (EHEC). Analysis of Stx-binding glycosphingolipids (GSLs) of primary human renal proximal tubular epithelial cells (pHRPTEpiCs) yielded globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) with Cer (d18:1, C16:0), Cer (d18:1, C22:0), and Cer (d18:1, C24:1/C24:0) as the dominant lipoforms. Investigation of detergent-resistant membranes (DRMs) and nonDRMs, serving as equivalents for the liquid-ordered and liquid-disordered membrane phase, respectively, revealed the prevalence of Gb3Cer and Gb4Cer together with cholesterol and sphingomyelin in DRMs, suggesting lipid raft association. Stx1a and Stx2a exerted strong cellular damage with half-maximal cytotoxic doses (CD50) of 1.31 × 102 pg/mL and 1.66 × 103 pg/mL, respectively, indicating one order of magnitude higher cellular cytotoxicity of Stx1a. Surface acoustic wave (SAW) real-time interaction analysis using biosensor surfaces coated with DRM or nonDRM fractions gave stronger binding capability of Stx1a versus Stx2a that correlated with the lower cytotoxicity of Stx2a. Our study underlines the substantial role of proximal tubular epithelial cells of the human kidney being associated with the development of Stx-mediated HUS at least for Stx1a, while the impact of Stx2a remains somewhat ambiguous.
Collapse
Affiliation(s)
- Johanna Detzner
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Anna-Lena Klein
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Gottfried Pohlentz
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Elisabeth Krojnewski
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Münster, D-48149 Münster, Germany;
| | - Alexander Mellmann
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Helge Karch
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Johannes Müthing
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
- Correspondence:
| |
Collapse
|
11
|
Smuggle tau through a secret(ory) pathway. Biochem J 2021; 478:2921-2925. [PMID: 34319403 DOI: 10.1042/bcj20210324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 11/17/2022]
Abstract
Secretion of misfolded tau, a microtubule-binding protein enriched in nerve cells, is linked to the progression of tau pathology. However, the molecular mechanisms underlying tau secretion are poorly understood. Recent work by Lee et al. [Biochemical J. (2021) 478: 1471-1484] demonstrated that the transmembrane domains of syntaxin6 and syntaxin8 could be exploited for tau release, setting a stage for testing a novel hypothesis that has profound implications in tauopathies (e.g. Alzheimer's disease, FTDP-17, and CBD/PSP) and other related neurodegenerative diseases. The present commentary highlights the importance and limitations of the study, and discusses opportunities and directions for future investigations.
Collapse
|
12
|
The Protein Toxins Ricin and Shiga Toxin as Tools to Explore Cellular Mechanisms of Internalization and Intracellular Transport. Toxins (Basel) 2021; 13:toxins13060377. [PMID: 34070659 PMCID: PMC8227415 DOI: 10.3390/toxins13060377] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/22/2021] [Indexed: 12/18/2022] Open
Abstract
Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.
Collapse
|
13
|
Rádis-Baptista G. Cell-Penetrating Peptides Derived from Animal Venoms and Toxins. Toxins (Basel) 2021; 13:147. [PMID: 33671927 PMCID: PMC7919042 DOI: 10.3390/toxins13020147] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/31/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-penetrating peptides (CPPs) comprise a class of short polypeptides that possess the ability to selectively interact with the cytoplasmic membrane of certain cell types, translocate across plasma membranes and accumulate in the cell cytoplasm, organelles (e.g., the nucleus and mitochondria) and other subcellular compartments. CPPs are either of natural origin or de novo designed and synthesized from segments and patches of larger proteins or designed by algorithms. With such intrinsic properties, along with membrane permeation, translocation and cellular uptake properties, CPPs can intracellularly convey diverse substances and nanomaterials, such as hydrophilic organic compounds and drugs, macromolecules (nucleic acids and proteins), nanoparticles (nanocrystals and polyplexes), metals and radionuclides, which can be covalently attached via CPP N- and C-terminals or through preparation of CPP complexes. A cumulative number of studies on animal toxins, primarily isolated from the venom of arthropods and snakes, have revealed the cell-penetrating activities of venom peptides and toxins, which can be harnessed for application in biomedicine and pharmaceutical biotechnology. In this review, I aimed to collate examples of peptides from animal venoms and toxic secretions that possess the ability to penetrate diverse types of cells. These venom CPPs have been chemically or structurally modified to enhance cell selectivity, bioavailability and a range of target applications. Herein, examples are listed and discussed, including cysteine-stabilized and linear, α-helical peptides, with cationic and amphipathic character, from the venom of insects (e.g., melittin, anoplin, mastoparans), arachnids (latarcin, lycosin, chlorotoxin, maurocalcine/imperatoxin homologs and wasabi receptor toxin), fish (pardaxins), amphibian (bombesin) and snakes (crotamine and cathelicidins).
Collapse
Affiliation(s)
- Gandhi Rádis-Baptista
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| |
Collapse
|
14
|
Detzner J, Krojnewski E, Pohlentz G, Steil D, Humpf HU, Mellmann A, Karch H, Müthing J. Shiga Toxin (Stx)-Binding Glycosphingolipids of Primary Human Renal Cortical Epithelial Cells (pHRCEpiCs) and Stx-Mediated Cytotoxicity. Toxins (Basel) 2021; 13:toxins13020139. [PMID: 33673393 PMCID: PMC7918848 DOI: 10.3390/toxins13020139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Human kidney epithelial cells are supposed to be directly involved in the pathogenesis of the hemolytic–uremic syndrome (HUS) caused by Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). The characterization of the major and minor Stx-binding glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), respectively, of primary human renal cortical epithelial cells (pHRCEpiCs) revealed GSLs with Cer (d18:1, C16:0), Cer (d18:1, C22:0), and Cer (d18:1, C24:1/C24:0) as the dominant lipoforms. Using detergent-resistant membranes (DRMs) and non-DRMs, Gb3Cer and Gb4Cer prevailed in the DRM fractions, suggesting their association with microdomains in the liquid-ordered membrane phase. A preference of Gb3Cer and Gb4Cer endowed with C24:0 fatty acid accompanied by minor monounsaturated C24:1-harboring counterparts was observed in DRMs, whereas the C24:1 fatty acid increased in relation to the saturated equivalents in non-DRMs. A shift of the dominant phospholipid phosphatidylcholine with saturated fatty acids in the DRM to unsaturated species in the non-DRM fractions correlated with the GSL distribution. Cytotoxicity assays gave a moderate susceptibility of pHRCEpiCs to the Stx1a and Stx2a subtypes when compared to highly sensitive Vero-B4 cells. The results indicate that presence of Stx-binding GSLs per se and preferred occurrence in microdomains do not necessarily lead to a high cellular susceptibility towards Stx.
Collapse
Affiliation(s)
- Johanna Detzner
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Elisabeth Krojnewski
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Gottfried Pohlentz
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Daniel Steil
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Münster, D-48149 Münster, Germany;
| | - Alexander Mellmann
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Helge Karch
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
| | - Johannes Müthing
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (E.K.); (G.P.); (D.S.); (A.M.); (H.K.)
- Correspondence:
| |
Collapse
|
15
|
Brandel A, Aigal S, Lagies S, Schlimpert M, Meléndez AV, Xu M, Lehmann A, Hummel D, Fisch D, Madl J, Eierhoff T, Kammerer B, Römer W. The Gb3-enriched CD59/flotillin plasma membrane domain regulates host cell invasion by Pseudomonas aeruginosa. Cell Mol Life Sci 2021; 78:3637-3656. [PMID: 33555391 PMCID: PMC8038999 DOI: 10.1007/s00018-021-03766-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa has gained precedence over the years due to its ability to develop resistance to existing antibiotics, thereby necessitating alternative strategies to understand and combat the bacterium. Our previous work identified the interaction between the bacterial lectin LecA and its host cell glycosphingolipid receptor globotriaosylceramide (Gb3) as a crucial step for the engulfment of P. aeruginosa via the lipid zipper mechanism. In this study, we define the LecA-associated host cell membrane domain by pull-down and mass spectrometry analysis. We unraveled a predilection of LecA for binding to saturated, long fatty acyl chain-containing Gb3 species in the extracellular membrane leaflet and an induction of dynamic phosphatidylinositol (3,4,5)-trisphosphate (PIP3) clusters at the intracellular leaflet co-localizing with sites of LecA binding. We found flotillins and the GPI-anchored protein CD59 not only to be an integral part of the LecA-interacting membrane domain, but also majorly influencing bacterial invasion as depletion of either of these host cell proteins resulted in about 50% reduced invasiveness of the P. aeruginosa strain PAO1. In summary, we report that the LecA-Gb3 interaction at the extracellular leaflet induces the formation of a plasma membrane domain enriched in saturated Gb3 species, CD59, PIP3 and flotillin thereby facilitating efficient uptake of PAO1.
Collapse
Affiliation(s)
- Annette Brandel
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Sahaja Aigal
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Simon Lagies
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Manuel Schlimpert
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Maokai Xu
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Anika Lehmann
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Daniel Hummel
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Department of Biochemistry, University of Geneva, 30 Quai Ernest-Ansermet, 1211, Geneva, Switzerland
| | - Daniel Fisch
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK
| | - Josef Madl
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Elsässer Straße 2q, 79110, Freiburg, Germany
| | - Thorsten Eierhoff
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert Schweitzer Campus 1, 48149, Münster, Germany
| | - Bernd Kammerer
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
| |
Collapse
|
16
|
Detzner J, Pohlentz G, Müthing J. Thin-Layer Chromatography in Structure and Recognition Studies of Shiga Toxin Glycosphingolipid Receptors. Methods Mol Biol 2021; 2291:229-252. [PMID: 33704756 DOI: 10.1007/978-1-0716-1339-9_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glycosphingolipids (GSLs) consist of a ceramide (Cer) lipid anchor, which is typically composed of the long-chain aminoalcohol sphingosine (d18:1) and a fatty acid (mostly C16-C24) and a sugar moiety harboring to a great extent one to five monosaccharides. GSLs of the globo-series are well-recognized receptors of Shiga toxins (Stxs) released by Stx-producing Escherichia coli (STEC). Receptors for the Stx subtypes Stx1a and Stx2a are globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), whereby Gb3Cer represents their high-affinity and Gb4Cer their low-affinity receptor. In addition to Gb3Cer and Gb4Cer, Gb5Cer and Forssman GSL are further receptors of the Stx2e subtype rendering Stx2e unique among the various Stx subtypes. Thin-layer chromatography (TLC) is a convenient and ubiquitously employed method for analyzing GSL mixtures of unknown composition. In particular, TLC immunochemical overlay detection allows for sensitive identification of Stx-binding GSLs in complex mixtures directly on the TLC plate. For this purpose, specific anti-GSL antibodies or Stxs themselves in conjunction with anti-Stx antibodies can be used. The described protocols of antibody-mediated detection of TLC-separated globo-series GSLs and corresponding identification of Stx-binding globo-series GSLs will provide detailed advice for successful GSL analysis and particularly highlight the power of the TLC overlay technique.
Collapse
Affiliation(s)
- Johanna Detzner
- Institute for Hygiene, University of Münster, Münster, Germany
| | | | | |
Collapse
|
17
|
Ansari S, Yamaoka Y. Role of vacuolating cytotoxin A in Helicobacter pylori infection and its impact on gastric pathogenesis. Expert Rev Anti Infect Ther 2020; 18:987-996. [PMID: 32536287 DOI: 10.1080/14787210.2020.1782739] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction Helicobacter pylori causes, via the influence of several virulence factors, persistent infection of the stomach, which leads to severe complications. Vacuolating cytotoxin A (VacA) is observed in almost all clinical strains of H. pylori; however, only some strains produce the toxigenic and pathogenic VacA, which is influenced by the gene sequence variations. VacA exerts its action by causing cell vacuolation and apoptosis. We performed a PubMed search to review the latest literatures published in English language. Areas covered Articles regarding H. pylori VacA and its genotypes, architecture, internalization, and role in gastric infection and pathogenicity are reviewed. We included the search for recently published literature until January 2020. Expert opinion H. pylori VacA plays a crucial role in severe gastric pathogenicity. In addition, VacA mediated in vivo bacterial survival leads to persistent infection and an enhanced bacterial evasion from the action of antibiotics and the innate host defense system, which leads to drug evasion. VacA as a co-stimulator for the CagA phosphorylation may exert a synergistic effect playing an important role in the CagA-mediated pathogenicity.
Collapse
Affiliation(s)
- Shamshul Ansari
- Department of Microbiology, Chitwan Medical College , Bharatpur, Nepal
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine , Yufu, Oita, Japan.,Global Oita Medical Advanced Research Center for Health , Yufu, Oita, Japan.,Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine , Houston, TX, USA.,Borneo Medical and Health Research Centre, Universiti Malaysia Sabah , Kota Kinabaru, Malaysia
| |
Collapse
|
18
|
Valid Presumption of Shiga Toxin-Mediated Damage of Developing Erythrocytes in EHEC-Associated Hemolytic Uremic Syndrome. Toxins (Basel) 2020; 12:toxins12060373. [PMID: 32512916 PMCID: PMC7354503 DOI: 10.3390/toxins12060373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
The global emergence of clinical diseases caused by enterohemorrhagic Escherichia coli (EHEC) is an issue of great concern. EHEC release Shiga toxins (Stxs) as their key virulence factors, and investigations on the cell-damaging mechanisms toward target cells are inevitable for the development of novel mitigation strategies. Stx-mediated hemolytic uremic syndrome (HUS), characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal injury, is the most severe outcome of an EHEC infection. Hemolytic anemia during HUS is defined as the loss of erythrocytes by mechanical disruption when passing through narrowed microvessels. The formation of thrombi in the microvasculature is considered an indirect effect of Stx-mediated injury mainly of the renal microvascular endothelial cells, resulting in obstructions of vessels. In this review, we summarize and discuss recent data providing evidence that HUS-associated hemolytic anemia may arise not only from intravascular rupture of erythrocytes, but also from the extravascular impairment of erythropoiesis, the development of red blood cells in the bone marrow, via direct Stx-mediated damage of maturing erythrocytes, leading to “non-hemolytic” anemia.
Collapse
|
19
|
Lingelem ABD, Kavaliauskiene S, Halsne R, Klokk TI, Surma MA, Klose C, Skotland T, Sandvig K. Diacylglycerol kinase and phospholipase D inhibitors alter the cellular lipidome and endosomal sorting towards the Golgi apparatus. Cell Mol Life Sci 2020; 78:985-1009. [PMID: 32447426 PMCID: PMC7897626 DOI: 10.1007/s00018-020-03551-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
The membrane lipids diacylglycerol (DAG) and phosphatidic acid (PA) are important second messengers that can regulate membrane transport by recruiting proteins to the membrane and by altering biophysical membrane properties. DAG and PA are involved in the transport from the Golgi apparatus to endosomes, and we have here investigated whether changes in these lipids might be important for regulation of transport to the Golgi using the protein toxin ricin. Modulation of DAG and PA levels using DAG kinase (DGK) and phospholipase D (PLD) inhibitors gave a strong increase in retrograde ricin transport, but had little impact on ricin recycling or degradation. Inhibitor treatment strongly affected the endosome morphology, increasing endosomal tubulation and size. Furthermore, ricin was present in these tubular structures together with proteins known to regulate retrograde transport. Using siRNA to knock down different isoforms of PLD and DGK, we found that several isoforms of PLD and DGK are involved in regulating ricin transport to the Golgi. Finally, by performing lipidomic analysis we found that the DGK inhibitor gave a weak, but expected, increase in DAG levels, while the PLD inhibitor gave a strong and unexpected increase in DAG levels, showing that it is important to perform lipidomic analysis when using inhibitors of lipid metabolism.
Collapse
Affiliation(s)
- Anne Berit Dyve Lingelem
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Forensic Biology, Oslo University Hospital, Oslo, Norway
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ruth Halsne
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Forensic Biology, Oslo University Hospital, Oslo, Norway
| | - Tove Irene Klokk
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Regional Committees for Medical and Health Research Ethics, University of Oslo, Oslo, Norway
| | | | | | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway. .,Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
20
|
Acosta W, Cramer CL. Targeting Macromolecules to CNS and Other Hard-to-Treat Organs Using Lectin-Mediated Delivery. Int J Mol Sci 2020; 21:ijms21030971. [PMID: 32024082 PMCID: PMC7037663 DOI: 10.3390/ijms21030971] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
The greatest challenges for therapeutic efficacy of many macromolecular drugs that act on intracellular are delivery to key organs and tissues and delivery into cells and subcellular compartments. Transport of drugs into critical cells associated with disease, including those in organs protected by restrictive biological barriers such as central nervous system (CNS), bone, and eye remains a significant hurdle to drug efficacy and impacts commercial risk and incentives for drug development for many diseases. These limitations expose a significant need for the development of novel strategies for macromolecule delivery. RTB lectin is the non-toxic carbohydrate-binding subunit B of ricin toxin with high affinity for galactose/galactosamine-containing glycolipids and glycoproteins common on human cell surfaces. RTB mediates endocytic uptake into mammalian cells by multiple routes exploiting both adsorptive-mediated and receptor-mediated mechanisms. In vivo biodistribution studies in lysosomal storage disease models provide evidence for the theory that the RTB-lectin transports corrective doses of enzymes across the blood–brain barrier to treat CNS pathologies. These results encompass significant implications for protein-based therapeutic approaches to address lysosomal and other diseases having strong CNS involvement.
Collapse
|
21
|
Skotland T, Sagini K, Sandvig K, Llorente A. An emerging focus on lipids in extracellular vesicles. Adv Drug Deliv Rev 2020; 159:308-321. [PMID: 32151658 DOI: 10.1016/j.addr.2020.03.002] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/02/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles contain a lipid bilayer membrane that protects the encapsulated material, such as proteins, nucleic acids, lipids and metabolites, from the extracellular environment. These vesicles are released from cells via different mechanisms. During recent years extracellular vesicles have been studied as possible biomarkers for different diseases, as biological nanoparticles for drug delivery, and in basic studies as a tool to understand the structure of biological membranes and the mechanisms involved in vesicular trafficking. Lipids are essential molecular components of extracellular vesicles, but at the moment our knowledge about the lipid composition and the function of lipids in these vesicles is limited. However, the interest of the research community in these molecules is increasing as their role in extracellular vesicles is starting to be acknowledged. In this review, we will present the status of the field and describe what is needed to bring it forward.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway.
| |
Collapse
|
22
|
Detzner J, Gloerfeld C, Pohlentz G, Legros N, Humpf HU, Mellmann A, Karch H, Müthing J. Structural Insights into Escherichia coli Shiga Toxin (Stx) Glycosphingolipid Receptors of Porcine Renal Epithelial Cells and Inhibition of Stx-Mediated Cellular Injury Using Neoglycolipid-Spiked Glycovesicles. Microorganisms 2019; 7:microorganisms7110582. [PMID: 31752441 PMCID: PMC6920957 DOI: 10.3390/microorganisms7110582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 11/18/2022] Open
Abstract
Shiga toxin (Stx) producing Escherichia coli (STEC) cause the edema disease in pigs by releasing the swine-pathogenic Stx2e subtype as the key virulence factor. Stx2e targets endothelial cells of animal organs including the kidney harboring the Stx receptor glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer, Galα1-4Galβ1-4Glcβ1-1Cer) and globotetraosylceramide (Gb4Cer, GalNAcβ1-3Galα1-4Galβ1-4Glcβ1-1Cer). Since the involvement of renal epithelial cells in the edema disease is unknown, in this study, we analyzed the porcine kidney epithelial cell lines, LLC-PK1 and PK-15, regarding the presence of Stx-binding GSLs, their sensitivity towards Stx2e, and the inhibitory potential of Gb3- and Gb4-neoglycolipids, carrying phosphatidylethanolamine (PE) as the lipid anchor, towards Stx2e. Immunochemical and mass spectrometric analysis revealed various Gb3Cer and Gb4Cer lipoforms as the dominant Stx-binding GSLs in both LLC-PK1 and PK-15 cells. A dihexosylceramide with proposed Galα1-4Gal-sequence (Gal2Cer) was detected in PK-15 cells, whereas LLC-PK1 cells lacked this compound. Both cell lines were susceptible towards Stx2e with LLC-PK1 representing an extremely Stx2e-sensitive cell line. Gb3-PE and Gb4-PE applied as glycovesicles significantly reduced the cytotoxic activity of Stx2e towards LLC-PK1 cells, whereas only Gb4-PE exhibited some protection against Stx2e for PK-15 cells. This is the first report identifying Stx2e receptors of porcine kidney epithelial cells and providing first data on their Stx2e-mediated damage suggesting possible involvement in the edema disease.
Collapse
Affiliation(s)
- Johanna Detzner
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Caroline Gloerfeld
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Gottfried Pohlentz
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Nadine Legros
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Hans-Ulrich Humpf
- Institute for Food Chemistry, University of Münster, 48149 Münster, Germany;
| | - Alexander Mellmann
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Helge Karch
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, 48149 Münster, Germany; (J.D.); (C.G.); (G.P.); (N.L.); (A.M.); (H.K.)
- Correspondence: ; Tel.: +49-(0)251-8355192
| |
Collapse
|
23
|
Skotland T, Sandvig K. The role of PS 18:0/18:1 in membrane function. Nat Commun 2019; 10:2752. [PMID: 31227693 PMCID: PMC6588574 DOI: 10.1038/s41467-019-10711-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
Various studies have demonstrated that the two leaflets of cellular membranes interact, potentially through so-called interdigitation between the fatty acyl groups. While the molecular mechanism underlying interleaflet coupling remains to be fully understood, recent results suggest interactions between the very-long-chain sphingolipids in the outer leaflet, and phosphatidylserine PS18:0/18:1 in the inner leaflet, and an important role for cholesterol for these interactions. Here we review the evidence that cross-linking of sphingolipids may result in clustering of phosphatidylserine and transfer of signals to the cytosol. Although much remains to be uncovered, the molecular properties and abundance of PS 18:0/18:1 suggest a unique role for this lipid. There are several lines of evidence for interactions between the two membrane leaflets in cells. In this review the authors discuss the transmembrane coupling of lipids, the involvement of phosphatidyl serine species PS 18:0/18:1, and their importance for various cellular processes.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.,Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
24
|
Ricin: An Ancient Story for a Timeless Plant Toxin. Toxins (Basel) 2019; 11:toxins11060324. [PMID: 31174319 PMCID: PMC6628454 DOI: 10.3390/toxins11060324] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
Abstract
The castor plant (Ricinus communis L.) has been known since time immemorial in traditional medicine in the pharmacopeia of Mediterranean and eastern ancient cultures. Moreover, it is still used in folk medicine worldwide. Castor bean has been mainly recommended as anti-inflammatory, anthelmintic, anti-bacterial, laxative, abortifacient, for wounds, ulcers, and many other indications. Many cases of human intoxication occurred accidentally or voluntarily with the ingestion of castor seeds or derivatives. Ricinus toxicity depends on several molecules, among them the most important is ricin, a protein belonging to the family of ribosome-inactivating proteins. Ricin is the most studied of this category of proteins and it is also known to the general public, having been used for several biocrimes. This manuscript intends to give the reader an overview of ricin, focusing on the historical path to the current knowledge on this protein. The main steps of ricin research are here reported, with particular regard to its enzymatic activity, structure, and cytotoxicity. Moreover, we discuss ricin toxicity for animals and humans, as well as the relation between bioterrorism and ricin and its impact on environmental toxicity. Ricin has also been used to develop immunotoxins for the elimination of unwanted cells, mainly cancer cells; some of these immunoconjugates gave promising results in clinical trials but also showed critical limitation.
Collapse
|
25
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
26
|
Sandvig K, Kavaliauskiene S, Skotland T. Clathrin-independent endocytosis: an increasing degree of complexity. Histochem Cell Biol 2018; 150:107-118. [PMID: 29774430 PMCID: PMC6096564 DOI: 10.1007/s00418-018-1678-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2018] [Indexed: 11/03/2022]
Abstract
This article aims at providing an update on the complexity of clathrin-independent endocytosis. It is now almost 30 years since we first wrote a review about its existence; at that time many people believed that with the exception of macropinocytosis, which will only be briefly mentioned in this review, all uptake could be accounted for by clathrin-dependent endocytosis. Now it is generally accepted that there are different clathrin-independent mechanisms, some of them regulated by ligands and membrane lipid composition. They can be both dynamin-dependent and -independent, meaning that the uptake cannot be accounted for by caveolae and other dynamin-dependent processes such as tubular structures that can be induced by toxins, e.g. Shiga toxin, or the fast endophilin mediated endocytosis recently described. Caveolae seem to be mostly quite stable structures with other functions than endocytosis, but evidence suggests that they may have cell-type dependent functions. Although several groups have been working on endocytic mechanisms for years, and new advanced methods have improved our ability to study mechanistic details, there are still a number of important questions we need to address, such as: How many endocytic mechanisms does a cell have? How quantitatively important are they? What about the complexity in polarized cells where clathrin-independent endocytosis is differentially regulated on the apical and basolateral poles? These questions are not easy to answer since one and the same molecule may contribute to more than one process, and manipulating one mechanism can affect another. Also, several inhibitors of endocytic processes commonly used turn out to be less specific than originally thought. We will here describe the current view of clathrin-independent endocytic processes and the challenges in studying them.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway.
- Department of Molecular Biosciences, University of Oslo, 0316, Oslo, Norway.
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway
| |
Collapse
|
27
|
Sandhoff R, Sandhoff K. Emerging concepts of ganglioside metabolism. FEBS Lett 2018; 592:3835-3864. [PMID: 29802621 DOI: 10.1002/1873-3468.13114] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 11/12/2022]
Abstract
Gangliosides (GGs) are sialic acid-containing glycosphingolipids (GSLs) and major membrane components enriched on cellular surfaces. Biosynthesis of mammalian GGs starts at the cytosolic leaflet of endoplasmic reticulum (ER) membranes with the formation of their hydrophobic ceramide anchors. After intracellular ceramide transfer to Golgi and trans-Golgi network (TGN) membranes, anabolism of GGs, as well as of other GSLs, is catalyzed by membrane-spanning glycosyltransferases (GTs) along the secretory pathway. Combined activity of only a few promiscuous GTs allows for the formation of cell-type-specific glycolipid patterns. Following an exocytotic vesicle flow to the cellular plasma membranes, GGs can be modified by metabolic reactions at or near the cellular surface. For degradation, GGs are endocytosed to reach late endosomes and lysosomes. Whereas membrane-spanning enzymes of the secretory pathway catalyze GSL and GG formation, a cooperation of soluble glycosidases, lipases and lipid-binding cofactors, namely the sphingolipid activator proteins (SAPs), act as the main players of GG and GSL catabolism at intralysosomal luminal vesicles (ILVs).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group (G131), German Cancer Research Center, Heidelberg, Germany
| | | |
Collapse
|
28
|
Legros N, Pohlentz G, Steil D, Kouzel IU, Liashkovich I, Mellmann A, Karch H, Müthing J. Membrane assembly of Shiga toxin glycosphingolipid receptors and toxin refractiveness of MDCK II epithelial cells. J Lipid Res 2018; 59:1383-1401. [PMID: 29866658 DOI: 10.1194/jlr.m083048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Shiga toxins (Stxs) are the major virulence factors of Stx-producing Escherichia coli (STEC), which cause hemorrhagic colitis and severe extraintestinal complications due to injury of renal endothelial cells, resulting in kidney failure. Since kidney epithelial cells are suggested additional targets for Stxs, we analyzed Madin-Darby canine kidney (MDCK) II epithelial cells for presence of Stx-binding glycosphingolipids (GSLs), determined their distribution to detergent-resistant membranes (DRMs), and ascertained the lipid composition of DRM and non-DRM preparations. Globotriaosylceramide and globotetraosylceramide, known as receptors for Stx1a, Stx2a, and Stx2e, and Forssman GSL as a specific receptor for Stx2e, were found to cooccur with SM and cholesterol in DRMs of MDCK II cells, which was shown using TLC overlay assay detection combined with mass spectrometry. The various lipoforms of GSLs were found to mainly harbor ceramide moieties composed of sphingosine (d18:1) and C24:1/C24:0 or C16:0 FA. The cells were highly refractory toward Stx1a, Stx2a, and Stx2e, most likely due to the absence of Stx-binding GSLs in the apical plasma membrane determined by immunofluorescence confocal laser scanning microscopy. The results suggest that the cellular content of Stx receptor GSLs and their biochemical detection in DRM preparations alone are inadequate to predict cellular sensitivity toward Stxs.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Ivan U Kouzel
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, D-48149 Münster, Germany
| | - Alexander Mellmann
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany .,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| |
Collapse
|
29
|
Legros N, Pohlentz G, Runde J, Dusny S, Humpf HU, Karch H, Müthing J. Colocalization of receptors for Shiga toxins with lipid rafts in primary human renal glomerular endothelial cells and influence of D-PDMP on synthesis and distribution of glycosphingolipid receptors. Glycobiology 2018; 27:947-965. [PMID: 28535204 DOI: 10.1093/glycob/cwx048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/17/2017] [Indexed: 12/12/2022] Open
Abstract
Damage of human renal glomerular endothelial cells (HRGECs) of the kidney represents the linchpin in the pathogenesis of the hemolytic uremic syndrome caused by Shiga toxins of enterohemorrhagic Escherichia coli (EHEC). We performed a comprehensive structural analysis of the Stx-receptor glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1Cer) and their distribution in lipid raft analog detergent-resistant membranes (DRMs) and nonDRMs prepared from primary HRGECs. Predominant receptor lipoforms were Gb3Cer and Gb4Cer with Cer (d18:1, C16:0), Cer (d18:1, C22:0) and Cer (d18:1, C24:1/C24:0). Stx-receptor GSLs co-distribute with sphingomyelin (SM) and cholesterol as well as flotillin-2 in DRMs, representing the liquid-ordered membrane phase and indicating lipid raft association. Lyso-phosphatidylcholine (lyso-PC) was identified as a nonDRM marker phospholipid of the liquid-disordered membrane phase. Exposure of primary HRGECs to the ceramide analogon d-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP) reduced total Gb3Cer and Gb4Cer content, roughly calculated from two biological replicates, down to half and quarter of its primordial content, respectively, but strengthened their prevalence and cholesterol preponderance in DRMs. At the same time, the distribution of PC, SM and lyso-PC to subcellular membrane fractions remained unaffected by D-PDMP treatment. Defining the GSL composition and precise microdomain structures of primary HRGECs may help to develop novel therapeutic options to combat life-threatening EHEC infections.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Gottfried Pohlentz
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Jana Runde
- Institute for Food Chemistry, University of Münster, D-48149 Münster, Germany
| | - Stefanie Dusny
- Institute for Food Chemistry, University of Münster, D-48149 Münster, Germany
| | - Hans-Ulrich Humpf
- Institute for Food Chemistry, University of Münster, D-48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| |
Collapse
|
30
|
Raghunathan K, Kenworthy AK. Dynamic pattern generation in cell membranes: Current insights into membrane organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2018-2031. [PMID: 29752898 DOI: 10.1016/j.bbamem.2018.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
It has been two decades since the lipid raft hypothesis was first presented. Even today, whether these nanoscale cholesterol-rich domains are present in cell membranes is not completely resolved. However, especially in the last few years, a rich body of literature has demonstrated both the presence and the importance of non-random distribution of biomolecules on the membrane, which is the focus of this review. These new developments have pushed the experimental limits of detection and have brought us closer to observing lipid domains in the plasma membrane of live cells. Characterization of biomolecules associated with lipid rafts has revealed a deep connection between biological regulation and function and membrane compositional heterogeneities. Finally, tantalizing new developments in the field have demonstrated that lipid domains might not just be associated with the plasma membrane of eukaryotes but could potentially be a ubiquitous membrane-organizing principle in several other biological systems. This article is part of a Special Issue entitled: Emergence of Complex Behavior in Biomembranes edited by Marjorie Longo.
Collapse
Affiliation(s)
- Krishnan Raghunathan
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, PA 15224, USA.
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
31
|
Abstract
Helicobacter pylori, a Gram-negative bacterium, is a well-known risk factor for gastric cancer. H. pylori vacuolating cytotoxin A (VacA) is a secreted pore-forming toxin that induces a wide range of cellular responses. Like many other bacterial toxins, VacA has been hypothesized to utilize lipid rafts to gain entry into host cells. Here, we used giant plasma membrane vesicles (GPMVs) as a model system to understand the preferential partitioning of VacA into lipid rafts. We show that a wild-type (WT) toxin predominantly associates with the raft phase. Acid activation of VacA enhances binding of the toxin to GPMVs but is not required for raft partitioning. VacA mutant proteins with alterations at the amino terminus (resulting in impaired membrane channel formation) and a nonoligomerizing VacA mutant protein retain the ability to preferentially associate with lipid rafts. Consistent with these results, the isolated VacA p55 domain was capable of binding to lipid rafts. We conclude that the affinity of VacA for rafts is independent of its capacity to oligomerize or form membrane channels.
Collapse
|
32
|
Kavaliauskiene S, Torgersen ML, Lingelem ABD, Klokk TI, Lintonen T, Simolin H, Ekroos K, Skotland T, Sandvig K. Cellular effects of fluorodeoxyglucose: Global changes in the lipidome and alteration in intracellular transport. Oncotarget 2018; 7:79885-79900. [PMID: 27829218 PMCID: PMC5346758 DOI: 10.18632/oncotarget.13089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/21/2016] [Indexed: 11/29/2022] Open
Abstract
2-fluoro-2-deoxy-D-glucose (FDG), labeled with 18F radioisotope, is the most common imaging agent used for positron emission tomography (PET) in oncology. However, little is known about the cellular effects of FDG. Another glucose analogue, 2-deoxy-D-glucose (2DG), has been shown to affect many cellular functions, including intracellular transport and lipid metabolism, and has been found to improve the efficacy of cancer chemotherapeutic agents in vivo. Thus, in the present study, we have investigated cellular effects of FDG with the focus on changes in cellular lipids and intracellular transport. By quantifying more than 200 lipids from 17 different lipid classes in HEp-2 cells and by analyzing glycosphingolipids from MCF-7, HT-29 and HBMEC cells, we have discovered that FDG treatment inhibits glucosylceramide synthesis and thus reduces cellular levels of glycosphingolipids. In addition, in HEp-2 cells the levels and/or species composition of other lipid classes, namely diacylglycerols, phosphatidic acids and phosphatidylinositols, were found to change upon treatment with FDG. Furthermore, we show here that FDG inhibits retrograde Shiga toxin transport and is much more efficient in protecting cells against the toxin than 2DG. In summary, our data reveal novel effects of FDG on cellular transport and glycosphingolipid metabolism, which suggest a potential clinical application of FDG as an adjuvant for cancer chemotherapy.
Collapse
Affiliation(s)
- Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Anne Berit Dyve Lingelem
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Tove Irene Klokk
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | | | | | | | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
33
|
Ou L, Przybilla MJ, Koniar B, Whitley CB. RTB lectin-mediated delivery of lysosomal α-l-iduronidase mitigates disease manifestations systemically including the central nervous system. Mol Genet Metab 2018; 123:105-111. [PMID: 29198892 PMCID: PMC5808854 DOI: 10.1016/j.ymgme.2017.11.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 01/09/2023]
Abstract
Mucopolysaccharidosis type I (MPS I) is a lysosomal disease resulting from deficiency in the α-L-iduronidase (IDUA) hydrolase and subsequent accumulation of glycosaminoglycan (GAG). Clinically, enzyme replacement therapy (ERT) with IDUA achieves negligible neurological benefits presumably due to blood-brain-barrier (BBB) limitations. To investigate the plant lectin ricin B chain (RTB) as a novel carrier for enzyme delivery to the brain, an IDUA:RTB fusion protein (IDUAL), produced in N. benthamiana leaves, was tested in a murine model of Hurler syndrome (MPS I). Affect mice (n=3 for each group) were intravenously injected with a single dose of IDUAL (0.58, 2 or 5.8mgIDUAequivalents/kg) and analyzed after 24h. IDUA activities in liver, kidney and spleen increased significantly, and liver GAG levels were significantly reduced in all three groups. Plasma IDUA levels for all treated groups were high at 1h after injection and decreased by 95% at 4h, indicating efficient distribution into tissues. For long-term evaluations, IDUAL (0.58 or 2mg/kg, 8 weekly injections) was intravenously injected into MPS I mice (n=12 for each group). Thirteen days after the 8th injection, significant IDUA activity was detected in the liver and spleen. GAG levels in tissues including the brain cortex and cerebellum were significantly reduced in treated animals. Treated MPS I mice also showed significant improvement in neurocognitive testing. ELISA results showed that while there was a significant antibody response against IDUAL and plant-derived IDUA, there was no significant antibody response to RTB. No major toxicity or adverse events were observed. Together, these results showed that infusion of IDUAL allowed for significant IDUA levels and GAG reduction in the brain and subsequent neurological benefits. This RTB-mediated delivery may have significant implications for therapeutic protein delivery impacting a broad spectrum of lysosomal, and potentially neurological diseases.
Collapse
Affiliation(s)
- Li Ou
- Gene Therapy Center, Department of Pediatrics, United States
| | | | - Brenda Koniar
- Research Animal Resources, University of Minnesota, Minneapolis, MN 55455, United States
| | | |
Collapse
|
34
|
Sandhoff R, Schulze H, Sandhoff K. Ganglioside Metabolism in Health and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:1-62. [DOI: 10.1016/bs.pmbts.2018.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
35
|
Danne R, Poojari C, Martinez-Seara H, Rissanen S, Lolicato F, Róg T, Vattulainen I. doGlycans-Tools for Preparing Carbohydrate Structures for Atomistic Simulations of Glycoproteins, Glycolipids, and Carbohydrate Polymers for GROMACS. J Chem Inf Model 2017; 57:2401-2406. [PMID: 28906114 PMCID: PMC5662928 DOI: 10.1021/acs.jcim.7b00237] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Indexed: 11/30/2022]
Abstract
Carbohydrates constitute a structurally and functionally diverse group of biological molecules and macromolecules. In cells they are involved in, e.g., energy storage, signaling, and cell-cell recognition. All of these phenomena take place in atomistic scales, thus atomistic simulation would be the method of choice to explore how carbohydrates function. However, the progress in the field is limited by the lack of appropriate tools for preparing carbohydrate structures and related topology files for the simulation models. Here we present tools that fill this gap. Applications where the tools discussed in this paper are particularly useful include, among others, the preparation of structures for glycolipids, nanocellulose, and glycans linked to glycoproteins. The molecular structures and simulation files generated by the tools are compatible with GROMACS.
Collapse
Affiliation(s)
- Reinis Danne
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Chetan Poojari
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department
of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Hector Martinez-Seara
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Institute
of Organic Chemistry and Biochemistry, Academy
of Sciences of the Czech Republic, Flemingovo nám. 2, 16610 Prague 6, Czech Republic
| | - Sami Rissanen
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Fabio Lolicato
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department
of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Tomasz Róg
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department
of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Ilpo Vattulainen
- Laboratory
of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department
of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- MEMPHYS−Center
for Biomembrane Physics, University of Southern
Denmark, 5230 Odense, Denmark
| |
Collapse
|
36
|
Park JY, Jeong YJ, Park SK, Yoon SJ, Choi S, Jeong DG, Chung SW, Lee BJ, Kim JH, Tesh VL, Lee MS, Park YJ. Shiga Toxins Induce Apoptosis and ER Stress in Human Retinal Pigment Epithelial Cells. Toxins (Basel) 2017; 9:toxins9100319. [PMID: 29027919 PMCID: PMC5666366 DOI: 10.3390/toxins9100319] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 01/03/2023] Open
Abstract
Shiga toxins (Stxs) produced by Shiga toxin-producing bacteria Shigella dysenteriae serotype 1 and select serotypes of Escherichia coli are the most potent known virulence factors in the pathogenesis of hemorrhagic colitis progressing to potentially fatal systemic complications such as acute renal failure, blindness and neurological abnormalities. Although numerous studies have defined apoptotic responses to Shiga toxin type 1 (Stx1) or Shiga toxin type 2 (Stx2) in a variety of cell types, the potential significance of Stx-induced apoptosis of photoreceptor and pigmented cells of the eye following intoxication is unknown. We explored the use of immortalized human retinal pigment epithelial (RPE) cells as an in vitro model of Stx-induced retinal damage. To the best of our knowledge, this study is the first report that intoxication of RPE cells with Stxs activates both apoptotic cell death signaling and the endoplasmic reticulum (ER) stress response. Using live-cell imaging analysis, fluorescently labeled Stx1 or Stx2 were internalized and routed to the RPE cell endoplasmic reticulum. RPE cells were significantly sensitive to wild type Stxs by 72 h, while the cells survived challenge with enzymatically deficient mutant toxins (Stx1A− or Stx2A−). Upon exposure to purified Stxs, RPE cells showed activation of a caspase-dependent apoptotic program involving a reduction of mitochondrial transmembrane potential (Δψm), increased activation of ER stress sensors IRE1, PERK and ATF6, and overexpression CHOP and DR5. Finally, we demonstrated that treatment of RPE cells with Stxs resulted in the activation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK), suggesting that the ribotoxic stress response may be triggered. Collectively, these data support the involvement of Stx-induced apoptosis in ocular complications of intoxication. The evaluation of apoptotic responses to Stxs by cells isolated from multiple organs may reveal unique functional patterns of the cytotoxic actions of these toxins in the systemic complications that follow ingestion of toxin-producing bacteria.
Collapse
Affiliation(s)
- Jun-Young Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| | - Yu-Jin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biochemistry, College of Medicine, Konyang University, 158 Gwanjeo-ro, Daejeon 35365, South Korea.
| | - Sung-Kyun Park
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Sung-Jin Yoon
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Song Choi
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Dae Gwin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Su Wol Chung
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Ulsan 44610, South Korea.
| | - Byung Joo Lee
- Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea.
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea.
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| | - Moo-Seung Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| | - Young-Jun Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| |
Collapse
|
37
|
Simm R, Kvalvaag AS, van Deurs B, Lindbäck T, Sandvig K. Benzyl alcohol induces a reversible fragmentation of the Golgi apparatus and inhibits membrane trafficking between endosomes and the trans-Golgi network. Exp Cell Res 2017; 357:67-78. [DOI: 10.1016/j.yexcr.2017.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/23/2017] [Accepted: 04/24/2017] [Indexed: 01/01/2023]
|
38
|
Antimisiaris S, Mourtas S, Papadia K. Targeted si-RNA with liposomes and exosomes (extracellular vesicles): How to unlock the potential. Int J Pharm 2017; 525:293-312. [PMID: 28163221 DOI: 10.1016/j.ijpharm.2017.01.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
The concept of RNA interference therapeutics has been initiated 18 years ago, and the main bottleneck for translation of the technology into therapeutic products remains the delivery of functional RNA molecules into the cell cytoplasm. In the present review article after an introduction about the theoretical basis of RNAi therapy and the main challenges encountered for its realization, an overview of the different types of delivery systems or carriers, used as potential systems to overcome RNAi delivery issues, will be provided. Characteristic examples or results obtained with the most promising systems will be discussed. Focus will be given mostly on the applications of liposomes or other types of lipid carriers, such as exosomes, towards improved delivery of RNAi to therapeutic targets. Finally the approach of integrating the advantages of these two vesicular systems, liposomes and exosomes, as a potential solution to realize RNAi therapy, will be proposed.
Collapse
Affiliation(s)
- Sophia Antimisiaris
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece; Institute of Chemical Engineering, FORTH/ICE-HT, Rio 26504, Greece.
| | - Spyridon Mourtas
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece
| | - Konstantina Papadia
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece
| |
Collapse
|
39
|
Kavaliauskiene S, Dyve Lingelem AB, Skotland T, Sandvig K. Protection against Shiga Toxins. Toxins (Basel) 2017; 9:E44. [PMID: 28165371 PMCID: PMC5331424 DOI: 10.3390/toxins9020044] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 12/12/2022] Open
Abstract
Shiga toxins consist of an A-moiety and five B-moieties able to bind the neutral glycosphingolipid globotriaosylceramide (Gb3) on the cell surface. To intoxicate cells efficiently, the toxin A-moiety has to be cleaved by furin and transported retrogradely to the Golgi apparatus and to the endoplasmic reticulum. The enzymatically active part of the A-moiety is then translocated to the cytosol, where it inhibits protein synthesis and in some cell types induces apoptosis. Protection of cells can be provided either by inhibiting binding of the toxin to cells or by interfering with any of the subsequent steps required for its toxic effect. In this article we provide a brief overview of the interaction of Shiga toxins with cells, describe some compounds and conditions found to protect cells against Shiga toxins, and discuss whether they might also provide protection in animals and humans.
Collapse
Affiliation(s)
- Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, N-0379 Oslo, Norway.
| | - Anne Berit Dyve Lingelem
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, N-0379 Oslo, Norway.
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, N-0379 Oslo, Norway.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, N-0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, N-0379 Oslo, Norway.
- Department of Biosciences, University of Oslo, N-0316 Oslo, Norway.
| |
Collapse
|
40
|
Becker B, Schnöder T, Schmitt MJ. Yeast Reporter Assay to Identify Cellular Components of Ricin Toxin A Chain Trafficking. Toxins (Basel) 2016; 8:toxins8120366. [PMID: 27929418 PMCID: PMC5198560 DOI: 10.3390/toxins8120366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 01/08/2023] Open
Abstract
RTA, the catalytic A-subunit of the ribosome inactivating A/B toxin ricin, inhibits eukaryotic protein biosynthesis by depurination of 28S rRNA. Although cell surface binding of ricin holotoxin is mainly mediated through its B-subunit (RTB), sole application of RTA is also toxic, albeit to a significantly lower extent, suggesting alternative pathways for toxin uptake and transport. Since ricin toxin trafficking in mammalian cells is still not fully understood, we developed a GFP-based reporter assay in yeast that allows rapid identification of cellular components required for RTA uptake and subsequent transport through a target cell. We hereby show that Ypt6p, Sft2p and GARP-complex components play an important role in RTA transport, while neither the retromer complex nor COPIB vesicles are part of the transport machinery. Analyses of yeast knock-out mutants with chromosomal deletion in genes whose products regulate ADP-ribosylation factor GTPases (Arf-GTPases) and/or retrograde Golgi-to-ER (endoplasmic reticulum) transport identified Sso1p, Snc1p, Rer1p, Sec22p, Erv46p, Gea1p and Glo3p as novel components in RTA transport, suggesting the developed reporter assay as a powerful tool to dissect the multistep processes of host cell intoxication in yeast.
Collapse
Affiliation(s)
- Björn Becker
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken D-66123, Germany.
| | - Tina Schnöder
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken D-66123, Germany.
| | - Manfred J Schmitt
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken D-66123, Germany.
| |
Collapse
|
41
|
IgG trafficking in the adult pig small intestine: one- or bidirectional transfer across the enterocyte brush border? Histochem Cell Biol 2016; 147:399-411. [PMID: 27646280 DOI: 10.1007/s00418-016-1492-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2016] [Indexed: 12/18/2022]
Abstract
Immunoglobulin G (IgG) transfer in opposite directions across the small intestinal brush border serves different purposes in early life and in adulthood. In the neonate, maternal IgG is taken up from the gut lumen into the blood, conferring passive immunity to the offspring, whereas in the adult immunoglobulins, including IgG made by plasma cells in the lamina propria, are secreted via the brush border to the lumen as part of the mucosal defense. Here, IgG has been proposed to perform a luminal immune surveillance which eventually includes a reuptake through the brush border as pathogen-containing immune complexes. In the present work, we studied luminal uptake of FITC-conjugated and gold-conjugated IgG in cultured pig jejunal mucosal explants. After 1 h, binding to the brush border was seen in upper crypts and lower parts of the villi. However, no endocytotic uptake into EEA-1-positive compartments was detected, neither at neutral nor acidic pH, despite an ongoing constitutive endocytosis from the brush border, visualized by the polar tracer CF594. The 40-kDa neonatal Fc receptor, FcRn, was present in the microvillus fraction, but noteworthy, a 37 kDa band, most likely a proteolytic cleavage product, bound IgG in a pH-dependent manner more efficiently than did the full-length FcRn. In conclusion, our work does not support the theory that bidirectional transfer of IgG across the intestinal brush border is part of the luminal immune surveillance in the adult.
Collapse
|
42
|
Ailte I, Lingelem ABD, Kavaliauskiene S, Bergan J, Kvalvaag AS, Myrann AG, Skotland T, Sandvig K. Addition of lysophospholipids with large head groups to cells inhibits Shiga toxin binding. Sci Rep 2016; 6:30336. [PMID: 27458147 PMCID: PMC4960542 DOI: 10.1038/srep30336] [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: 02/15/2016] [Accepted: 06/30/2016] [Indexed: 12/20/2022] Open
Abstract
Shiga toxin (Stx), an AB5 toxin, binds specifically to the neutral glycosphingolipid Gb3 at the cell surface before being transported into cells. We here demonstrate that addition of conical lysophospholipids (LPLs) with large head groups inhibit Stx binding to cells whereas LPLs with small head groups do not. Lysophosphatidylinositol (LPI 18:0), the most efficient LPL with the largest head group, was selected for in-depth investigations to study how the binding of Stx is regulated. We show that the inhibition of Stx binding by LPI is reversible and possibly regulated by cholesterol since addition of methyl-β-cyclodextrin (mβCD) reversed the ability of LPI to inhibit binding. LPI-induced inhibition of Stx binding is independent of signalling and membrane turnover as it occurs in fixed cells as well as after depletion of cellular ATP. Furthermore, data obtained with fluorescent membrane dyes suggest that LPI treatment has a direct effect on plasma membrane lipid packing with shift towards a liquid disordered phase in the outer leaflet, while lysophosphoethanolamine (LPE), which has a small head group, does not. In conclusion, our data show that cellular treatment with conical LPLs with large head groups changes intrinsic properties of the plasma membrane and modulates Stx binding to Gb3.
Collapse
Affiliation(s)
- Ieva Ailte
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Anne Berit Dyve Lingelem
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Simona Kavaliauskiene
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jonas Bergan
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway.,Department of Research and Innovation, Østfold Hospital, Sarpsborg, Norway
| | - Audun Sverre Kvalvaag
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Anne-Grethe Myrann
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Tore Skotland
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
43
|
Abstract
Post-infectious hemolytic uremic syndrome (HUS) is caused by specific pathogens in patients with no identifiable HUS-associated genetic mutation or autoantibody. The majority of episodes is due to infections by Shiga toxin (Stx) producing Escherichia coli (STEC). This chapter reviews the epidemiology and pathogenesis of STEC-HUS, including bacterial-derived factors and host responses. STEC disease is characterized by hematological (microangiopathic hemolytic anemia), renal (acute kidney injury) and extrarenal organ involvement. Clinicians should always strive for an etiological diagnosis through the microbiological or molecular identification of Stx-producing bacteria and Stx or, if negative, serological assays. Treatment of STEC-HUS is supportive; more investigations are needed to evaluate the efficacy of putative preventive and therapeutic measures, such as non-phage-inducing antibiotics, volume expansion and anti-complement agents. The outcome of STEC-HUS is generally favorable, but chronic kidney disease, permanent extrarenal, mainly cerebral complication and death (in less than 5 %) occur and long-term follow-up is recommended. The remainder of this chapter highlights rarer forms of (post-infectious) HUS due to S. dysenteriae, S. pneumoniae, influenza A and HIV and discusses potential interactions between these pathogens and the complement system.
Collapse
Affiliation(s)
- Denis F. Geary
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Franz Schaefer
- Division of Pediatric Nephrology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
44
|
Shiga Toxins as Multi-Functional Proteins: Induction of Host Cellular Stress Responses, Role in Pathogenesis and Therapeutic Applications. Toxins (Basel) 2016; 8:toxins8030077. [PMID: 26999205 PMCID: PMC4810222 DOI: 10.3390/toxins8030077] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 12/17/2022] Open
Abstract
Shiga toxins (Stxs) produced by Shiga toxin-producing bacteria Shigella dysenteriae serotype 1 and select serotypes of Escherichia coli are primary virulence factors in the pathogenesis of hemorrhagic colitis progressing to potentially fatal systemic complications, such as hemolytic uremic syndrome and central nervous system abnormalities. Current therapeutic options to treat patients infected with toxin-producing bacteria are limited. The structures of Stxs, toxin-receptor binding, intracellular transport and the mode of action of the toxins have been well defined. However, in the last decade, numerous studies have demonstrated that in addition to being potent protein synthesis inhibitors, Stxs are also multifunctional proteins capable of activating multiple cell stress signaling pathways, which may result in apoptosis, autophagy or activation of the innate immune response. Here, we briefly present the current understanding of Stx-activated signaling pathways and provide a concise review of therapeutic applications to target tumors by engineering the toxins.
Collapse
|
45
|
Klokk TI, Kavaliauskiene S, Sandvig K. Cross-linking of glycosphingolipids at the plasma membrane: consequences for intracellular signaling and traffic. Cell Mol Life Sci 2016; 73:1301-16. [PMID: 26407609 PMCID: PMC11108300 DOI: 10.1007/s00018-015-2049-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/16/2015] [Accepted: 09/17/2015] [Indexed: 12/11/2022]
Abstract
Glycosphingolipids (GSLs) are predominantly found in the outer leaflet of the plasma membrane, where they play a role in important processes such as cell adhesion, migration and signaling. However, by which mechanisms GSLs regulate these processes remains elusive. In this study, we therefore took advantage of the fact that some GSLs also serve as receptors for certain protein toxins, which rely on receptor binding for internalization and intoxication. Here, we demonstrate that Shiga and cholera toxins, which both possess multivalent GSL-binding capacity, induce dissociation of the cytosolic cPLA2α-AnxA1 complex in HeLa and HMEC-1 cells. The dissociation is mediated through an increase in cytosolic calcium levels and activation of the tyrosine kinase Syk. Ricin, a protein toxin that does not cross-link surface molecules, has no effect on the same complex. Importantly, we find that antibody-mediated cross-linking of Gb3 and GM1, the GSL receptors for Shiga and cholera toxin, respectively, also induces dissociation. These data demonstrate that cross-linking of GSLs at the plasma membrane mediates the intracellular signaling events resulting in dissociation of the complex. After dissociation, cPLA2α and AnxA1 are translocated to intracellular membranes where they are known to function in regulating membrane transport processes. In conclusion, we have characterized a novel mechanism for cell surface-induced initiation of intracellular signaling and transport events.
Collapse
Affiliation(s)
- Tove Irene Klokk
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway.
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0316, Oslo, Norway.
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0316, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0316, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| |
Collapse
|
46
|
Helms JB, Kaloyanova DV, Strating JRP, van Hellemond JJ, van der Schaar HM, Tielens AGM, van Kuppeveld FJM, Brouwers JF. Targeting of the hydrophobic metabolome by pathogens. Traffic 2016; 16:439-60. [PMID: 25754025 PMCID: PMC7169838 DOI: 10.1111/tra.12280] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 12/12/2022]
Abstract
The hydrophobic molecules of the metabolome – also named the lipidome – constitute a major part of the entire metabolome. Novel technologies show the existence of a staggering number of individual lipid species, the biological functions of which are, with the exception of only a few lipid species, unknown. Much can be learned from pathogens that have evolved to take advantage of the complexity of the lipidome to escape the immune system of the host organism and to allow their survival and replication. Different types of pathogens target different lipids as shown in interaction maps, allowing visualization of differences between different types of pathogens. Bacterial and viral pathogens target predominantly structural and signaling lipids to alter the cellular phenotype of the host cell. Fungal and parasitic pathogens have complex lipidomes themselves and target predominantly the release of polyunsaturated fatty acids from the host cell lipidome, resulting in the generation of eicosanoids by either the host cell or the pathogen. Thus, whereas viruses and bacteria induce predominantly alterations in lipid metabolites at the host cell level, eukaryotic pathogens focus on interference with lipid metabolites affecting systemic inflammatory reactions that are part of the immune system. A better understanding of the interplay between host–pathogen interactions will not only help elucidate the fundamental role of lipid species in cellular physiology, but will also aid in the generation of novel therapeutic drugs.
Collapse
Affiliation(s)
- J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Róg T, Orłowski A, Llorente A, Skotland T, Sylvänne T, Kauhanen D, Ekroos K, Sandvig K, Vattulainen I. Interdigitation of long-chain sphingomyelin induces coupling of membrane leaflets in a cholesterol dependent manner. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:281-8. [DOI: 10.1016/j.bbamem.2015.12.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 11/16/2015] [Accepted: 12/01/2015] [Indexed: 12/15/2022]
|
48
|
Postila PA, Vattulainen I, Róg T. Selective effect of cell membrane on synaptic neurotransmission. Sci Rep 2016; 6:19345. [PMID: 26782980 PMCID: PMC4725992 DOI: 10.1038/srep19345] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022] Open
Abstract
Atomistic molecular dynamics simulations were performed with 13 non-peptidic neurotransmitters (NTs) in three different membrane environments. The results provide compelling evidence that NTs are divided into membrane-binding and membrane-nonbinding molecules. NTs adhere to the postsynaptic membrane surface whenever the ligand-binding sites of their synaptic receptors are buried in the lipid bilayer. In contrast, NTs that have extracellular ligand-binding sites do not have a similar tendency to adhere to the membrane surface. This finding is a seemingly simple yet important addition to the paradigm of neurotransmission, essentially dividing it into membrane-independent and membrane-dependent mechanisms. Moreover, the simulations also indicate that the lipid composition especially in terms of charged lipids can affect the membrane partitioning of NTs. The revised paradigm, highlighting the importance of cell membrane and specific lipids for neurotransmission, should to be of interest to neuroscientists, drug industry and the general public alike.
Collapse
Affiliation(s)
- Pekka A. Postila
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department of Chemistry and Biochemistry, University of California San Diego, 92093-0340 San Diego, CA, USA
| | - Ilpo Vattulainen
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- MEMPHYS– Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014, Helsinki, Finland
| | - Tomasz Róg
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| |
Collapse
|
49
|
Steil D, Schepers CL, Pohlentz G, Legros N, Runde J, Humpf HU, Karch H, Müthing J. Shiga toxin glycosphingolipid receptors of Vero-B4 kidney epithelial cells and their membrane microdomain lipid environment. J Lipid Res 2015; 56:2322-36. [PMID: 26464281 DOI: 10.1194/jlr.m063040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Indexed: 12/12/2022] Open
Abstract
Shiga toxins (Stxs) are produced by enterohemorrhagic Escherichia coli (EHEC), which cause human infections with an often fatal outcome. Vero cell lines, derived from African green monkey kidney, represent the gold standard for determining the cytotoxic effects of Stxs. Despite their global use, knowledge about the exact structures of the Stx receptor glycosphingolipids (GSLs) and their assembly in lipid rafts is poor. Here we present a comprehensive structural analysis of Stx receptor GSLs and their distribution to detergent-resistant membranes (DRMs), which were prepared from Vero-B4 cells and used as lipid raft equivalents. We identified globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) as the GSL receptors for Stx1a, Stx2a, and Stx2e subtypes using TLC overlay detection combined with MS. The uncommon Stx receptor, globopentaosylceramide (Gb5Cer, Galβ3GalNAcβ3Galα4Galβ4Glcβ1Cer), which was specifically recognized (in addition to Gb3Cer and Gb4Cer) by Stx2e, was fully structurally characterized. Lipoforms of Stx receptor GSLs were found to mainly harbor ceramide moieties composed of sphingosine (d18:1) and C24:0/C24:1 or C16:0 fatty acid. Moreover, co-occurrence with lipid raft markers, SM and cholesterol, in DRMs suggested GSL association with membrane microdomains. This study provides the basis for further exploring the functional impact of lipid raft-associated Stx receptors for toxin-mediated injury of Vero-B4 cells.
Collapse
Affiliation(s)
- Daniel Steil
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | | | | | - Nadine Legros
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | - Jana Runde
- Food Chemistry, University of Münster, D-48149 Münster, Germany
| | | | - Helge Karch
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
50
|
Kymre L, Simm R, Skotland T, Sandvig K. Different roles of the C-terminal end of Stx1A and Stx2A for AB5 complex integrity and retrograde transport of Stx in HeLa cells. Pathog Dis 2015; 73:ftv083. [PMID: 26443836 DOI: 10.1093/femspd/ftv083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2015] [Indexed: 11/14/2022] Open
Abstract
Shiga toxin 1 (Stx1) and Shiga toxin 2 (Stx2) differ regarding receptor affinity, cellular toxicity and clinical outcome. To this date, it is not clarified in detail why the subtypes display these differences. Even though the crystal structures of Stx1 and Stx2 share overall similarities, significant differences were found in the C-terminal end of the A-subunits. The aim of this study was to investigate the role of the C-terminal end of the A-subunit in complex stability and retrograde transport by generating truncated mutants where 2, 4, 6 and 8 amino acids were removed from the C-terminal end of Stx1A and Stx2A. The results obtained show that removal of 6 or 8 amino acids from the Stx1A C-terminus abolishes the AB5 complex integrity, while removing up to 8 amino acids from Stx2A does not affect the complex in vivo (in the bacteria). We also present results showing different levels of A1-subunit in HeLa cells after exposure to Stx1, Stx2 and their truncated mutants.
Collapse
Affiliation(s)
- Linn Kymre
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Roger Simm
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|